1
|
Zhou C, Zhang YF, Yang ZJ, Huang YQ, Da MX. Computed tomography-based deep learning radiomics model for preoperative prediction of tumor immune microenvironment in colorectal cancer. World J Gastrointest Oncol 2025; 17:106103. [DOI: 10.4251/wjgo.v17.i5.106103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2025] [Revised: 03/08/2025] [Accepted: 03/31/2025] [Indexed: 05/15/2025] Open
Abstract
BACKGROUND Colorectal cancer (CRC) is a leading cause of cancer-related death globally, with the tumor immune microenvironment (TIME) influencing prognosis and immunotherapy response. Current TIME evaluation relies on invasive biopsies, limiting its clinical application. This study hypothesized that computed tomography (CT)-based deep learning (DL) radiomics models can non-invasively predict key TIME biomarkers: Tumor-stroma ratio (TSR), tumor-infiltrating lymphocytes (TILs), and immune score (IS).
AIM To develop a non-invasive DL approach using preoperative CT radiomics to evaluate TIME components in CRC patients.
METHODS In this retrospective study, preoperative CT images of 315 pathologically confirmed CRC patients (220 in training cohort and 95 in validation cohort) were analyzed. Manually delineated regions of interest were used to extract DL features. Predictive models (DenseNet-121/169) for TSR, TILs, IS, and TIME classification were constructed. Performance was evaluated via receiver operating characteristic curves, calibration curves, and decision curve analysis (DCA).
RESULTS The DL-DenseNet-169 model achieved area under the curve (AUC) values of 0.892 [95% confidence interval (CI): 0.828-0.957] for TSR and 0.772 (95%CI: 0.674-0.870) for TIME score. The DenseNet-121 model yielded AUC values of 0.851 (95%CI: 0.768-0.933) for TILs and 0.852 (95%CI: 0.775-0.928) for IS. Calibration curves demonstrated strong prediction-observation agreement, and DCA confirmed clinical utility across threshold probabilities (P < 0.05 for all models).
CONCLUSION CT-based DL radiomics provides a reliable non-invasive method for preoperative TIME evaluation, enabling personalized immunotherapy strategies in CRC management.
Collapse
Affiliation(s)
- Chuan Zhou
- The First Clinical Medical College of Lanzhou University, Lanzhou University, Lanzhou 730000, Gansu Province, China
- NHC Key Laboratory of Diagnosis and Therapy of Gastrointestinal Tumor, Gansu Provincial Hospital, Lanzhou 730000, Gansu Province, China
- Key Laboratory of Molecular Diagnostics and Precision Medicine for Surgical Oncology in Gansu Province, Gansu Provincial Hospital, Lanzhou 730000, Gansu Province, China
| | - Yun-Feng Zhang
- The First Clinical Medical College of Lanzhou University, Lanzhou University, Lanzhou 730000, Gansu Province, China
| | - Zhi-Jun Yang
- The First Clinical Medical College of Lanzhou University, Lanzhou University, Lanzhou 730000, Gansu Province, China
| | - Yu-Qian Huang
- Center of Medical Cosmetology, Chengdu Second People’s Hospital, Chengdu 610017, Sichuan Province, China
| | - Ming-Xu Da
- The First Clinical Medical College of Lanzhou University, Lanzhou University, Lanzhou 730000, Gansu Province, China
- NHC Key Laboratory of Diagnosis and Therapy of Gastrointestinal Tumor, Gansu Provincial Hospital, Lanzhou 730000, Gansu Province, China
- Key Laboratory of Molecular Diagnostics and Precision Medicine for Surgical Oncology in Gansu Province, Gansu Provincial Hospital, Lanzhou 730000, Gansu Province, China
- Department of Surgical Oncology, Gansu Provincial Hospital, Lanzhou 730000, Gansu Province, China
| |
Collapse
|
2
|
Hong T, Zhang H, Zhao Q, Liu L, Sun J, Hu S, Mao Y. A Hybrid Machine Learning CT-Based Radiomics Nomogram for Predicting Cancer-Specific Survival in Curatively Resected Colorectal Cancer. Acad Radiol 2025; 32:2630-2641. [PMID: 39922745 DOI: 10.1016/j.acra.2024.12.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 12/10/2024] [Accepted: 12/10/2024] [Indexed: 02/10/2025]
Abstract
RATIONALE AND OBJECTIVES To develop and validate a computed tomography-based radiomics nomogram for cancer-specific survival (CSS) prediction in curatively resected colorectal cancer (CRC), and its performance was compared with the American Joint Committee on Cancer (AJCC) staging and clinical-pathological models. MATERIALS AND METHODS A total of 794 patients with curatively resected CRC from a prospective cancer registry program were included and randomly divided into the training (n = 556) and validation (n = 238) cohorts. A radiomics signature (RS) predicting CSS was constructed with a hybrid automatic machine learning strategy, and the prognostic value was assessed with Kaplan-Meier (KM) survival analysis. The performance of the established models was assessed by the discrimination, calibration, and clinical utility. RESULTS A 10-feature-based RS with independent prognostic value was developed. KM survival curves showed that high-risk patients defined by RS had a worse CSS than the low-risk patients (log-rank P<0.001). The radiomics nomogram integrating the RS and clinical-pathological factors had the optimal performance in predicting CSS in terms of Harrell's concordance index (0.803 [95% confidence interval: 0.761-0.845] for the primary cohort, 0.772 [95% confidence interval: 0.702-0.841] for the validation cohort), time-dependent receiver operating curves (time-ROC) (the area under the time-ROC curves [AUC] at three years were 84.06±2.86 and at five years were 86.35±2.19 in the primary cohort, the AUC at three years were 77.6±4.76, and at five years were 84±3.66 in the validation cohort), calibration curves and decision curve analysis, in comparison with the AJCC staging model, clinical-pathological model, and the RS alone. CONCLUSION The radiomics nomogram integrating the RS and clinical-pathological factors could be a valuable individualized predictor of the CSS for curatively resected CRC patients.
Collapse
Affiliation(s)
- Tingting Hong
- Department of Medical Oncology, the Affiliated Hospital of Jiangnan University, No.1000, Hefeng Road, Wuxi 214000, China (T.H., Y.M.).
| | - Heng Zhang
- Department of Radiology, the Affiliated Hospital of Jiangnan University, No.1000, Hefeng Road, Wuxi 214000, China (H.Z., S.H.).
| | - Qiming Zhao
- Department of Artificial Intelligence and Computer Science, Jiangnan University, No.1800, Lihu Big Road, Wuxi 214122, China (Q.Z., J.S.).
| | - Li Liu
- Big Data Center, the Affiliated Hospital of Jiangnan University, No.1000, Hefeng Road, Wuxi 214000, China (L.L.).
| | - Jun Sun
- Department of Artificial Intelligence and Computer Science, Jiangnan University, No.1800, Lihu Big Road, Wuxi 214122, China (Q.Z., J.S.).
| | - Shudong Hu
- Department of Radiology, the Affiliated Hospital of Jiangnan University, No.1000, Hefeng Road, Wuxi 214000, China (H.Z., S.H.).
| | - Yong Mao
- Department of Medical Oncology, the Affiliated Hospital of Jiangnan University, No.1000, Hefeng Road, Wuxi 214000, China (T.H., Y.M.).
| |
Collapse
|
3
|
Howell HJ, McGale JP, Choucair A, Shirini D, Aide N, Postow MA, Wang L, Tordjman M, Lopci E, Lecler A, Champiat S, Chen DL, Deandreis D, Dercle L. Artificial Intelligence for Drug Discovery: An Update and Future Prospects. Semin Nucl Med 2025; 55:406-422. [PMID: 39966029 DOI: 10.1053/j.semnuclmed.2025.01.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Revised: 01/23/2025] [Accepted: 01/24/2025] [Indexed: 02/20/2025]
Abstract
Artificial intelligence (AI) has become a pivotal tool for medical image analysis, significantly enhancing drug discovery through improved diagnostics, staging, prognostication, and response assessment. At a high level, AI-driven image analysis enables the quantification and synthesis of previously qualitative imaging characteristics, facilitating the identification of novel disease-specific biomarkers, patient risk stratification, prognostication, and adverse event prediction. In addition, AI can assist in response assessment by capturing changes in imaging "phenotype" over time, allowing for optimized treatment plans based on real-time analysis. Integrating this emerging technology into drug discovery pipelines has the potential to accelerate the identification and development of new pharmaceuticals by assisting in target identification and patient selection, as well as reducing the incidence, and therefore cost, of failed trials through high-throughput, reproducible, and data-driven insights. Continued progress in AI applications will shape the future of medical imaging, ultimately fostering more efficient, accurate, and tailored drug discovery processes. Herein, we offer a comprehensive overview of how AI enhances medical imaging to inform drug development and therapeutic strategies.
Collapse
Affiliation(s)
- Harrison J Howell
- Department of Radiology, New York-Presbyterian Hospital, Columbia University Vagelos College of Physicians and Surgeons, New York, NY
| | - Jeremy P McGale
- Department of Radiology, New York-Presbyterian Hospital, Columbia University Vagelos College of Physicians and Surgeons, New York, NY
| | | | - Dorsa Shirini
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Nicolas Aide
- Centre Havrais d'Imagerie Nucléaire, Octeville, France
| | - Michael A Postow
- Department of Medicine, Memorial Sloan Kettering and Department of Medicine, Weill Cornell Medical College, New York, NY
| | - Lucy Wang
- School of Medicine, New York Medical College, Valhalla, NY
| | - Mickael Tordjman
- Department of Radiology, Biomedical Engineering & Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Egesta Lopci
- Nuclear Medicine Unit, IRCCS-Humanitas Research Hospital, Rozzano, Italy
| | - Augustin Lecler
- Department of Neuroradiology, Foundation Adolphe de Rothschild Hospital, Université Paris Cité, Paris, France
| | - Stéphane Champiat
- Department of Investigational Cancer Therapeutics, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Delphine L Chen
- Department of Radiology, University of Washington, Seattle, WA
| | | | - Laurent Dercle
- Department of Radiology, New York-Presbyterian Hospital, Columbia University Vagelos College of Physicians and Surgeons, New York, NY.
| |
Collapse
|
4
|
Guo X, Song J, Zhu L, Liu S, Huang C, Zhou L, Chen W, Lin G, Zhao Z, Tu J, Chen M, Chen F, Zheng L, Ji J. Multiparametric MRI-based radiomics and clinical nomogram predicts the recurrence of hepatocellular carcinoma after postoperative adjuvant transarterial chemoembolization. BMC Cancer 2025; 25:683. [PMID: 40229712 PMCID: PMC11995621 DOI: 10.1186/s12885-025-14079-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: 03/02/2024] [Accepted: 04/03/2025] [Indexed: 04/16/2025] Open
Abstract
BACKGROUND This study was undertaken to develop and validate a radiomics model based on multiparametric magnetic resonance imaging (MRI) for predicting recurrence in patients with hepatocellular carcinoma (HCC) following postoperative adjuvant transarterial chemoembolization (PA-TACE). METHODS In this retrospective study, 149 HCC patients (81 for training, 36 for internal validation, 32 for external validation) treated with PA-TACE were included in two medical centers. Multiparametric radiomics features were extracted from three MRI sequences. Least absolute shrinkage and selection operator (LASSO)-COX regression was utilized to select radiomics features. Optimal clinical characteristics selected by multivariate Cox analysis were integrated with Rad-score to develop a recurrence-free survival (RFS) prediction model. The model performance was evaluated by time-dependent receiver operating characteristic (ROC) curves, Harrell's concordance index (C-index), and calibration curve. RESULTS Fifteen optimal radiomic features were selected and the median Rad-score value was 0.434. Multivariate Cox analysis indicated that neutrophil-to-lymphocyte ratio (NLR) (hazard ratio (HR) = 1.49, 95% confidence interval (CI): 1.1-2.1, P = 0.022) and tumor size (HR = 1.28, 95% CI: 1.1-1.5, P = 0.001) were the independent predictors of RFS after PA-TACE. A combined model was established by integrating Rad-score, NLR, and tumor size in the training cohort (C-index 0.822; 95% CI 0.805-0.861), internal validation cohort (0.823; 95% CI 0.771-0.876) and external validation cohort (0.846; 95% CI 0.768-0.924). The calibration curve exhibited a satisfactory correspondence. CONCLUSION A multiparametric MRI-based radiomics model can predict RFS of HCC patients receiving PA-TACE and a nomogram can be served as an individualized tool for prognosis.
Collapse
Affiliation(s)
- Xinyu Guo
- Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, Zhejiang Engineering Research Center of Interventional Medicine Engineering and Biotechnology, Zhejiang Key Laboratory of Imaging and Interventional Medicine, Lishui Hospital of Zhejiang University, Lishui, 323000, China
- Department of Radiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, Zhejiang, China
| | - Jingjing Song
- Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, Zhejiang Engineering Research Center of Interventional Medicine Engineering and Biotechnology, Zhejiang Key Laboratory of Imaging and Interventional Medicine, Lishui Hospital of Zhejiang University, Lishui, 323000, China
| | - Lingyi Zhu
- Second Clinical Medical School, Zhejiang Chinese Medicine University, Hangzhou, 310003, Zhejiang, China
| | - Shuang Liu
- Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, Zhejiang Engineering Research Center of Interventional Medicine Engineering and Biotechnology, Zhejiang Key Laboratory of Imaging and Interventional Medicine, Lishui Hospital of Zhejiang University, Lishui, 323000, China
- Department of Radiology, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, 323000, China
| | - Chaoming Huang
- Second Clinical Medical School, Zhejiang Chinese Medicine University, Hangzhou, 310003, Zhejiang, China
| | - Lingling Zhou
- Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, Zhejiang Engineering Research Center of Interventional Medicine Engineering and Biotechnology, Zhejiang Key Laboratory of Imaging and Interventional Medicine, Lishui Hospital of Zhejiang University, Lishui, 323000, China
- Department of Radiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, Zhejiang, China
| | - Weiyue Chen
- Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, Zhejiang Engineering Research Center of Interventional Medicine Engineering and Biotechnology, Zhejiang Key Laboratory of Imaging and Interventional Medicine, Lishui Hospital of Zhejiang University, Lishui, 323000, China
- Department of Radiology, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, 323000, China
| | - Guihan Lin
- Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, Zhejiang Engineering Research Center of Interventional Medicine Engineering and Biotechnology, Zhejiang Key Laboratory of Imaging and Interventional Medicine, Lishui Hospital of Zhejiang University, Lishui, 323000, China
- Department of Radiology, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, 323000, China
| | - Zhongwei Zhao
- Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, Zhejiang Engineering Research Center of Interventional Medicine Engineering and Biotechnology, Zhejiang Key Laboratory of Imaging and Interventional Medicine, Lishui Hospital of Zhejiang University, Lishui, 323000, China
- Department of Radiology, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, 323000, China
| | - Jianfei Tu
- Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, Zhejiang Engineering Research Center of Interventional Medicine Engineering and Biotechnology, Zhejiang Key Laboratory of Imaging and Interventional Medicine, Lishui Hospital of Zhejiang University, Lishui, 323000, China
- Department of Radiology, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, 323000, China
| | - Minjiang Chen
- Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, Zhejiang Engineering Research Center of Interventional Medicine Engineering and Biotechnology, Zhejiang Key Laboratory of Imaging and Interventional Medicine, Lishui Hospital of Zhejiang University, Lishui, 323000, China
- Department of Radiology, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, 323000, China
| | - Feng Chen
- Department of Radiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, Zhejiang, China
| | - Liyun Zheng
- Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, Zhejiang Engineering Research Center of Interventional Medicine Engineering and Biotechnology, Zhejiang Key Laboratory of Imaging and Interventional Medicine, Lishui Hospital of Zhejiang University, Lishui, 323000, China.
- Department of Radiology, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, 323000, China.
| | - Jiansong Ji
- Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, Zhejiang Engineering Research Center of Interventional Medicine Engineering and Biotechnology, Zhejiang Key Laboratory of Imaging and Interventional Medicine, Lishui Hospital of Zhejiang University, Lishui, 323000, China.
- Department of Radiology, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, 323000, China.
| |
Collapse
|
5
|
Jiang C, Fang W, Wei N, Ma W, Dai C, Liu R, Cai A, Feng Q. Node Reporting and Data System Combined With Computed Tomography Radiomics Can Improve the Prediction of Nonenlarged Lymph Node Metastasis in Gastric Cancer. J Comput Assist Tomogr 2025; 49:215-224. [PMID: 39438281 DOI: 10.1097/rct.0000000000001673] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2024]
Abstract
OBJECTIVES To investigate the diagnostic performance of Node Reporting and Data System (Node-RADS) combined with computed tomography (CT) radiomics for assessing nonenlargement regional lymph nodes in gastric cancer (GC). METHODS Preoperative CT images were retrospectively collected from 376 pathologically confirmed of gastric adenocarcinoma from January 2019 to December 2023, with 605 lymph nodes included for analysis. They were divided into training (n = 362) and validation (n = 243) sets. Radiomics features were extracted from venous-phase, and the radiomics score was obtained. Clinical information, CT parameters, and Node-RADS classification were collected. A combined model was built using machine-learning approach and tested in validation set using receiver operating characteristic curve analysis. Further validation was conducted in different subgroups of lymph node short-axis diameter (SD) range. RESULTS Node-RADS score, SD, maximum diameter of thickness of tumor, and radiomics were identified as the most predictive factors. The results demonstrated that the integrated model combining SD, maximum diameter of thickness of tumor, Node-RADS, and radiomics outperformed the model excluding radiomics, yielding an area under the receiver operating characteristic curve of 0.82 compared with 0.79, with a statistically significant difference ( P < 0.001). Subgroup analysis based on different SDs of lymph nodes also revealed enhanced diagnostic accuracy when incorporating the radiomics score for the 4- to 7.9-mm subgroups, all P < 0.05. However, for the 8- to 9.9-mm subgroup, the combination of the radiomics did not significantly improve the prediction, with an area under the receiver operating characteristic curve of 0.85 versus 0.85, P = 0.877. CONCLUSION The integration of radiomics scores with Node-RADS assessments significantly enhances the accuracy of lymph node metastasis evaluation for GC. This combined model is particularly effective for lymph nodes with smaller standard deviations, yielding a marked improvement in diagnostic precision. CLINICAL RELEVANCE STATEMENT The findings of this study indicate that a composite model, which incorporates Node-RADS, radiomics features, and conventional parameters, may serve as an effective method for the assessment of nonenlarged lymph nodes in GC.
Collapse
Affiliation(s)
| | - Wei Fang
- Radiology Department, Yidu Central Hospital of Shandong Second Medical University, Qingzhou, Shandong
| | - Na Wei
- Yidu Central Hospital of Shandong Second Medical University, Qingzhou
| | - Wenwen Ma
- Radiology Department, Affiliated Hospital of Shandong Second Medical University, Weifang
| | - Cong Dai
- Radiology Department, Yidu Central Hospital of Shandong Second Medical University, Qingzhou, Shandong
| | - Ruixue Liu
- Pathology Department, Yidu Central Hospital of Shandong Second Medical University, Qingzhou, Shandong Province, China
| | - Anzhen Cai
- Radiology Department, Yidu Central Hospital of Shandong Second Medical University, Qingzhou, Shandong
| | - Qiang Feng
- Radiology Department, Yidu Central Hospital of Shandong Second Medical University, Qingzhou, Shandong
| |
Collapse
|
6
|
Mian A, Kamnitsas K, Gordon-Weeks A. Radiomics for Treatment Planning in Liver Cancers. JAMA Surg 2025:2830620. [PMID: 40009391 DOI: 10.1001/jamasurg.2024.4346] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/27/2025]
Abstract
This Surgical Innovation delves into the transformative potential of radiomics in liver cancer treatment, highlighting its advancements in pretreatment prognosis, noninvasive tumor profiling, and treatment response prediction, thus paving the way for more personalized therapeutic strategies.
Collapse
Affiliation(s)
- Areeb Mian
- Nuffield Department of Surgical Sciences, John Radcliffe Hospital, University of Oxford, Oxford, United Kingdom
- Department of Hepato-Pancreato-Biliary (HPB) Surgery, Oxford University Hospitals NHS Foundation Trust, Oxford, United Kingdom
| | - Konstantinos Kamnitsas
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford, United Kingdom
| | - Alex Gordon-Weeks
- Nuffield Department of Surgical Sciences, John Radcliffe Hospital, University of Oxford, Oxford, United Kingdom
- Department of Hepato-Pancreato-Biliary (HPB) Surgery, Oxford University Hospitals NHS Foundation Trust, Oxford, United Kingdom
| |
Collapse
|
7
|
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
|
8
|
Naemi A, Tashk A, Sorayaie Azar A, Samimi T, Tavassoli G, Bagherzadeh Mohasefi A, Nasiri Khanshan E, Heshmat Najafabad M, Tarighi V, Wiil UK, Bagherzadeh Mohasefi J, Pirnejad H, Niazkhani Z. Applications of Artificial Intelligence for Metastatic Gastrointestinal Cancer: A Systematic Literature Review. Cancers (Basel) 2025; 17:558. [PMID: 39941923 PMCID: PMC11817159 DOI: 10.3390/cancers17030558] [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: 12/10/2024] [Revised: 01/18/2025] [Accepted: 02/05/2025] [Indexed: 02/16/2025] Open
Abstract
BACKGROUND/OBJECTIVES This systematic literature review examines the application of Artificial Intelligence (AI) in the diagnosis, treatment, and follow-up of metastatic gastrointestinal cancers. METHODS The databases PubMed, Scopus, Embase (Ovid), and Google Scholar were searched for published articles in English from January 2010 to January 2022, focusing on AI models in metastatic gastrointestinal cancers. RESULTS forty-six studies were included in the final set of reviewed papers. The critical appraisal and data extraction followed the checklist for systematic reviews of prediction modeling studies. The risk of bias in the included papers was assessed using the prediction risk of bias assessment tool. CONCLUSIONS AI techniques, including machine learning and deep learning models, have shown promise in improving diagnostic accuracy, predicting treatment outcomes, and identifying prognostic biomarkers. Despite these advancements, challenges persist, such as reliance on retrospective data, variability in imaging protocols, small sample sizes, and data preprocessing and model interpretability issues. These challenges limit the generalizability, clinical application, and integration of AI models.
Collapse
Affiliation(s)
- Amin Naemi
- Nordcee, Department of Biology, University of Southern Denmark, 5230 Odense, Denmark
| | - Ashkan Tashk
- Cognitive Systems, DTU Compute, The Technical University of Denmark (DTU), 2800 Copenhagen, Denmark;
| | - Amir Sorayaie Azar
- SDU Health Informatics and Technology, The Maersk Mc-Kinney Moller Institute, University of Southern Denmark, 5230 Odense, Denmark; (A.S.A.); (U.K.W.); (J.B.M.)
- Department of Computer Engineering, Urmia University, Urmia 165, Iran; (A.B.M.); (E.N.K.); (M.H.N.); (V.T.)
| | - Tahereh Samimi
- Student Research Committee, Urmia University of Medical Sciences, Urmia 1138, Iran;
- Department of Medical Informatics, Urmia University of Medical Sciences, Urmia 1138, Iran
| | - Ghanbar Tavassoli
- Department of Computer Engineering, Urmia Branch, Islamic Azad University, Urmia 969, Iran;
| | - Anita Bagherzadeh Mohasefi
- Department of Computer Engineering, Urmia University, Urmia 165, Iran; (A.B.M.); (E.N.K.); (M.H.N.); (V.T.)
| | - Elaheh Nasiri Khanshan
- Department of Computer Engineering, Urmia University, Urmia 165, Iran; (A.B.M.); (E.N.K.); (M.H.N.); (V.T.)
| | - Mehrdad Heshmat Najafabad
- Department of Computer Engineering, Urmia University, Urmia 165, Iran; (A.B.M.); (E.N.K.); (M.H.N.); (V.T.)
| | - Vafa Tarighi
- Department of Computer Engineering, Urmia University, Urmia 165, Iran; (A.B.M.); (E.N.K.); (M.H.N.); (V.T.)
| | - Uffe Kock Wiil
- SDU Health Informatics and Technology, The Maersk Mc-Kinney Moller Institute, University of Southern Denmark, 5230 Odense, Denmark; (A.S.A.); (U.K.W.); (J.B.M.)
| | - Jamshid Bagherzadeh Mohasefi
- SDU Health Informatics and Technology, The Maersk Mc-Kinney Moller Institute, University of Southern Denmark, 5230 Odense, Denmark; (A.S.A.); (U.K.W.); (J.B.M.)
- Department of Computer Engineering, Urmia University, Urmia 165, Iran; (A.B.M.); (E.N.K.); (M.H.N.); (V.T.)
| | - Habibollah Pirnejad
- Patient Safety Research Center, Clinical Research Institute, Urmia University of Medical Sciences, Urmia 1138, Iran;
- Department of Family Medicine, Amsterdam University Medical Center, 7057 Amsterdam, The Netherlands
| | - Zahra Niazkhani
- Nephrology and Kidney Transplant Research Center, Clinical Research Institute, Urmia University of Medical Sciences, Urmia 1138, Iran;
- Erasmus School of Health Policy and Management (ESHPM), Erasmus University Rotterdam, 3000 Rotterdam, The Netherlands
| |
Collapse
|
9
|
Kou J, Peng JY, Lv WB, Wu CF, Chen ZH, Zhou GQ, Wang YQ, Lin L, Lu LJ, Sun Y. A Serial MRI-based Deep Learning Model to Predict Survival in Patients with Locoregionally Advanced Nasopharyngeal Carcinoma. Radiol Artif Intell 2025; 7:e230544. [PMID: 39812582 DOI: 10.1148/ryai.230544] [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: 01/16/2025]
Abstract
Purpose To develop and evaluate a deep learning-based prognostic model for predicting survival in locoregionally advanced nasopharyngeal carcinoma (LA-NPC) using serial MRI before and after induction chemotherapy (IC). Materials and Methods This multicenter retrospective study included 1039 patients with LA-NPC (779 male and 260 female patients; mean age, 44 years ± 11 [SD]) diagnosed between December 2011 and January 2016. A radiomics-clinical prognostic model (model RC) was developed using pre- and post-IC MRI acquisitions and other clinical factors using graph convolutional neural networks. The concordance index (C-index) was used to evaluate model performance in predicting disease-free survival (DFS). The survival benefits of concurrent chemoradiation therapy (CCRT) were analyzed in model-defined risk groups. Results The C-indexes of model RC for predicting DFS were significantly higher than those of TNM staging in the internal (0.79 vs 0.53) and external (0.79 vs 0.62, both P < .001) testing cohorts. The 5-year DFS for the model RC-defined low-risk group was significantly better than that of the high-risk group (90.6% vs 58.9%, P < .001). In high-risk patients, those who underwent CCRT had a higher 5-year DFS rate than those who did not (58.7% vs 28.6%, P = .03). There was no evidence of a difference in 5-year DFS rate in low-risk patients who did or did not undergo CCRT (91.9% vs 81.3%, P = .19). Conclusion Serial MRI before and after IC can effectively help predict survival in LA-NPC. The radiomics-clinical prognostic model developed using a graph convolutional network-based deep learning method showed good risk discrimination capabilities and may facilitate risk-adapted therapy. Keywords: Nasopharyngeal Carcinoma, Deep Learning, Induction Chemotherapy, Serial MRI, MR Imaging, Radiomics, Prognosis, Radiation Therapy/Oncology, Head/Neck Supplemental material is available for this article. © RSNA, 2025.
Collapse
Affiliation(s)
- Jia Kou
- From the Department of Radiation Oncology, State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, 651 Dongfeng Road East, Guangzhou 510060, China (J.K., C.F.W., Z.H.C., G.Q.Z., Y.Q.W., L.L., Y.S.); Department of Radiotherapy Center, The Sixth Affiliated Hospital, School of Medicine, South China University of Technology, Foshan, China (J.Y.P.); School of Biomedical Engineering and Guangdong Provincial Key Laboratory of Medical Image Processing, Southern Medical University, Guangzhou, China (L.J.L.); and Department of Electronic Engineering, Information School, Yunnan University, Kunming, China (W.B.L.)
| | - Jun-Yi Peng
- From the Department of Radiation Oncology, State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, 651 Dongfeng Road East, Guangzhou 510060, China (J.K., C.F.W., Z.H.C., G.Q.Z., Y.Q.W., L.L., Y.S.); Department of Radiotherapy Center, The Sixth Affiliated Hospital, School of Medicine, South China University of Technology, Foshan, China (J.Y.P.); School of Biomedical Engineering and Guangdong Provincial Key Laboratory of Medical Image Processing, Southern Medical University, Guangzhou, China (L.J.L.); and Department of Electronic Engineering, Information School, Yunnan University, Kunming, China (W.B.L.)
| | - Wen-Bing Lv
- From the Department of Radiation Oncology, State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, 651 Dongfeng Road East, Guangzhou 510060, China (J.K., C.F.W., Z.H.C., G.Q.Z., Y.Q.W., L.L., Y.S.); Department of Radiotherapy Center, The Sixth Affiliated Hospital, School of Medicine, South China University of Technology, Foshan, China (J.Y.P.); School of Biomedical Engineering and Guangdong Provincial Key Laboratory of Medical Image Processing, Southern Medical University, Guangzhou, China (L.J.L.); and Department of Electronic Engineering, Information School, Yunnan University, Kunming, China (W.B.L.)
| | - Chen-Fei Wu
- From the Department of Radiation Oncology, State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, 651 Dongfeng Road East, Guangzhou 510060, China (J.K., C.F.W., Z.H.C., G.Q.Z., Y.Q.W., L.L., Y.S.); Department of Radiotherapy Center, The Sixth Affiliated Hospital, School of Medicine, South China University of Technology, Foshan, China (J.Y.P.); School of Biomedical Engineering and Guangdong Provincial Key Laboratory of Medical Image Processing, Southern Medical University, Guangzhou, China (L.J.L.); and Department of Electronic Engineering, Information School, Yunnan University, Kunming, China (W.B.L.)
| | - Zi-Hang Chen
- From the Department of Radiation Oncology, State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, 651 Dongfeng Road East, Guangzhou 510060, China (J.K., C.F.W., Z.H.C., G.Q.Z., Y.Q.W., L.L., Y.S.); Department of Radiotherapy Center, The Sixth Affiliated Hospital, School of Medicine, South China University of Technology, Foshan, China (J.Y.P.); School of Biomedical Engineering and Guangdong Provincial Key Laboratory of Medical Image Processing, Southern Medical University, Guangzhou, China (L.J.L.); and Department of Electronic Engineering, Information School, Yunnan University, Kunming, China (W.B.L.)
| | - Guan-Qun Zhou
- From the Department of Radiation Oncology, State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, 651 Dongfeng Road East, Guangzhou 510060, China (J.K., C.F.W., Z.H.C., G.Q.Z., Y.Q.W., L.L., Y.S.); Department of Radiotherapy Center, The Sixth Affiliated Hospital, School of Medicine, South China University of Technology, Foshan, China (J.Y.P.); School of Biomedical Engineering and Guangdong Provincial Key Laboratory of Medical Image Processing, Southern Medical University, Guangzhou, China (L.J.L.); and Department of Electronic Engineering, Information School, Yunnan University, Kunming, China (W.B.L.)
| | - Ya-Qin Wang
- From the Department of Radiation Oncology, State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, 651 Dongfeng Road East, Guangzhou 510060, China (J.K., C.F.W., Z.H.C., G.Q.Z., Y.Q.W., L.L., Y.S.); Department of Radiotherapy Center, The Sixth Affiliated Hospital, School of Medicine, South China University of Technology, Foshan, China (J.Y.P.); School of Biomedical Engineering and Guangdong Provincial Key Laboratory of Medical Image Processing, Southern Medical University, Guangzhou, China (L.J.L.); and Department of Electronic Engineering, Information School, Yunnan University, Kunming, China (W.B.L.)
| | - Li Lin
- From the Department of Radiation Oncology, State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, 651 Dongfeng Road East, Guangzhou 510060, China (J.K., C.F.W., Z.H.C., G.Q.Z., Y.Q.W., L.L., Y.S.); Department of Radiotherapy Center, The Sixth Affiliated Hospital, School of Medicine, South China University of Technology, Foshan, China (J.Y.P.); School of Biomedical Engineering and Guangdong Provincial Key Laboratory of Medical Image Processing, Southern Medical University, Guangzhou, China (L.J.L.); and Department of Electronic Engineering, Information School, Yunnan University, Kunming, China (W.B.L.)
| | - Li-Jun Lu
- From the Department of Radiation Oncology, State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, 651 Dongfeng Road East, Guangzhou 510060, China (J.K., C.F.W., Z.H.C., G.Q.Z., Y.Q.W., L.L., Y.S.); Department of Radiotherapy Center, The Sixth Affiliated Hospital, School of Medicine, South China University of Technology, Foshan, China (J.Y.P.); School of Biomedical Engineering and Guangdong Provincial Key Laboratory of Medical Image Processing, Southern Medical University, Guangzhou, China (L.J.L.); and Department of Electronic Engineering, Information School, Yunnan University, Kunming, China (W.B.L.)
| | - Ying Sun
- From the Department of Radiation Oncology, State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, 651 Dongfeng Road East, Guangzhou 510060, China (J.K., C.F.W., Z.H.C., G.Q.Z., Y.Q.W., L.L., Y.S.); Department of Radiotherapy Center, The Sixth Affiliated Hospital, School of Medicine, South China University of Technology, Foshan, China (J.Y.P.); School of Biomedical Engineering and Guangdong Provincial Key Laboratory of Medical Image Processing, Southern Medical University, Guangzhou, China (L.J.L.); and Department of Electronic Engineering, Information School, Yunnan University, Kunming, China (W.B.L.)
| |
Collapse
|
10
|
Miyamoto Y, Nakaura T, Ohuchi M, Ogawa K, Kato R, Maeda Y, Eto K, Iwatsuki M, Baba Y, Hirai T, Baba H. Radiomics-based Machine Learning Approach to Predict Chemotherapy Responses in Colorectal Liver Metastases. J Anus Rectum Colon 2025; 9:117-126. [PMID: 39882217 PMCID: PMC11772800 DOI: 10.23922/jarc.2024-077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Accepted: 10/15/2024] [Indexed: 01/31/2025] Open
Abstract
Objectives This study explored the clinical utility of CT radiomics-driven machine learning as a predictive marker for chemotherapy response in colorectal liver metastasis (CRLM) patients. Methods We included 150 CRLM patients who underwent first-line doublet chemotherapy, dividing them into a training cohort (n=112) and a test cohort (n=38). We manually delineated three-dimensional tumor volumes, selecting the largest liver metastasis for measurement, using pretreatment portal-phase CT images and extracted 107 radiomics features. Treatment response was classified as responder (complete or partial response) or non-responder (stable or progressive disease), based on the best overall response according to RECIST criteria, version 1.1. Employing Random Forest and Boruta algorithms, we identified significant features for responder-non-responder differentiation. Radiomics signatures were developed and validated in the training cohort using five-fold cross-validation, and performance was assessed using the area under the curve (AUC). Results Among the patients, 91 (61%) were responders and 59 (39%) were non-responders. Variable selection with Boruta revealed three key parameters ("DependenceVariance," "ClusterShade," and "RunVariance"). In the training cohort, individual CT texture parameter AUCs ranged from 0.4 to 0.65, while the machine learning analysis incorporating all valid parameters exhibited a significantly higher AUC of 0.94 (p<0.01). The validation cohort also demonstrated strong predictive accuracy, with an AUC of 0.87 for treatment response. Conclusions This study highlights the potential of CT radiomics-driven machine learning in predicting chemotherapy responses among CRLM patients.
Collapse
Affiliation(s)
- Yuji Miyamoto
- Department of Gastroenterological Surgery, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Takeshi Nakaura
- Department of Diagnostic Radiology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Mayuko Ohuchi
- Department of Gastroenterological Surgery, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Katsuhiro Ogawa
- Department of Gastroenterological Surgery, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Rikako Kato
- Department of Gastroenterological Surgery, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Yuto Maeda
- Department of Gastroenterological Surgery, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Kojiro Eto
- Department of Gastroenterological Surgery, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Masaaki Iwatsuki
- Division of Translational Research and Advanced Treatment Against Gastrointestinal Cancer, Kumamoto University, Kumamoto, Japan
| | - Yoshifumi Baba
- Department of Next-Generation Surgical Therapy Development, Kumamoto University, Kumamoto, Japan
| | - Toshinori Hirai
- Department of Diagnostic Radiology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Hideo Baba
- Department of Gastroenterological Surgery, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| |
Collapse
|
11
|
Parillo M, Quattrocchi CC. Node Reporting and Data System 1.0 (Node-RADS) for the Assessment of Oncological Patients' Lymph Nodes in Clinical Imaging. J Clin Med 2025; 14:263. [PMID: 39797344 PMCID: PMC11722337 DOI: 10.3390/jcm14010263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2024] [Revised: 01/01/2025] [Accepted: 01/03/2025] [Indexed: 01/13/2025] Open
Abstract
The assessment of lymph node (LN) involvement with clinical imaging is a key factor in cancer staging. Node Reporting and Data System 1.0 (Node-RADS) was introduced in 2021 as a new system specifically tailored for classifying and reporting LNs on computed tomography (CT) and magnetic resonance imaging scans. The aim of this review is to compile the scientific evidence that has emerged since the introduction of Node-RADS, with a specific focus on its diagnostic performance and reliability. Node-RADS's performance has been evaluated in various cancer types and anatomical sites, revealing a trend where higher Node-RADS scores correspond to a greater probability of metastatic LN with better diagnostic performances compared to using short axis diameter alone. Moreover, Node-RADS exhibits encouraging diagnostic value for both Node-RADS ≥ 3 and Node-RADS ≥ 4 cutoffs in predicting metastatic LN. In terms of Node-RADS scoring reliability, preliminary studies show promising but partially conflicting results, with agreement levels, mostly between two readers, ranging from fair to almost perfect. This review highlights a wide variation in methodologies across different studies. Thus, to fully realize the potential of Node-RADS in clinical practice, future studies should comprehensively evaluate its diagnostic accuracy, category-specific malignancy rates, and inter-observer agreement. Finally, although limited, promising evidence has suggested the following: a potential prognostic role for Node-RADS; the possible value of diffusion-weighted imaging for LNs classified as Node-RADS ≥ 3; a correlation between Node-RADS and certain texture features in CT; and improved diagnostic performance when Node-RADS is integrated into radiomics or clinical models.
Collapse
Affiliation(s)
- Marco Parillo
- Radiology, Multizonal Unit of Rovereto and Arco, APSS Provincia Autonoma Di Trento, 38123 Trento, Italy;
| | - Carlo Cosimo Quattrocchi
- Radiology, Multizonal Unit of Rovereto and Arco, APSS Provincia Autonoma Di Trento, 38123 Trento, Italy;
- Centre for Medical Sciences—CISMed, University of Trento, 38122 Trento, Italy
| |
Collapse
|
12
|
Viganò L, Zanuso V, Fiz F, Cerri L, Laino ME, Ammirabile A, Ragaini EM, Viganò S, Terracciano LM, Francone M, Ieva F, Di Tommaso L, Rimassa L. CT-based radiogenomics of intrahepatic cholangiocarcinoma. Dig Liver Dis 2025; 57:118-124. [PMID: 39003163 DOI: 10.1016/j.dld.2024.06.033] [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: 04/19/2024] [Revised: 06/21/2024] [Accepted: 06/28/2024] [Indexed: 07/15/2024]
Abstract
BACKGROUND Intrahepatic cholangiocarcinoma (ICC) is an aggressive disease with increasing incidence and its genetic alterations could be the target of systemic therapies. AIMS To elucidate if radiomics extracted from computed tomography (CT) may non-invasively predict ICC genetic alterations. METHODS All consecutive patients with a diagnosis of a mass-forming ICC (01/2016-06/2022) were considered. Inclusion criteria were availability of a high-quality contrast-enhanced CT and molecular profiling by NGS or FISH for FGFR2 fusion/rearrangement. The CT scan at diagnosis was considered. Genetic analyses were performed on surgical specimens (resectable patients) or biopsies (unresectable ones). The radiomic features were extracted using the LifeX software. Multivariate predictive models of the commonest genetic alterations were built. RESULTS In the 90 enrolled patients (58 NGS/32 FISH, median age 65 years), the most common genetic alterations were FGFR2 (20/90), IDH1 (10/58), and KRAS (9/58). At internal validation, the combined clinical-radiomic models achieved the best performance for the prediction of FGFR2 (AUC = 0.892) and IDH1 status (AUC = 0.819), outperforming the pure clinical and radiomic models. The radiomic model for predicting KRAS mutations achieved an AUC = 0.767 (vs. 0.660 of the clinical model) without further improvements with the addition of clinical features. CONCLUSIONS CT-based radiomics provides a reliable non-invasive prediction of ICC genetic status with a major impact on therapeutic strategies.
Collapse
Affiliation(s)
- Luca Viganò
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy; Hepatobiliary Unit, Department of Minimally Invasive General & Oncologic Surgery, Humanitas Gavazzeni University Hospital, Bergamo, Italy.
| | - Valentina Zanuso
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy; Medical Oncology and Hematology Unit, Humanitas Cancer Center, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Francesco Fiz
- Nuclear Medicine Unit, Department of Diagnostic Imaging, Ente Ospedaliero "Ospedali Galliera", Genoa, Italy; Department of Nuclear Medicine and Clinical Molecular Imaging, University Hospital, Tübingen, Germany
| | - Luca Cerri
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy
| | | | - Angela Ammirabile
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy; Department of Radiology, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Elisa Maria Ragaini
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy
| | - Samuele Viganò
- MOX laboratory, Department of Mathematics, Politecnico di Milano, Milan, Italy
| | - Luigi Maria Terracciano
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy; Pathology Unit, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Marco Francone
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy; Department of Radiology, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Francesca Ieva
- MOX laboratory, Department of Mathematics, Politecnico di Milano, Milan, Italy; CHDS - Center for Health Data Science, Human Technopole, Milan, Italy
| | - Luca Di Tommaso
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy; Pathology Unit, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Lorenza Rimassa
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy; Medical Oncology and Hematology Unit, Humanitas Cancer Center, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| |
Collapse
|
13
|
Liu L, Cui WC, Sun Y, Wang H, Liang ZN, Wu W, Yan K, Ji YL, Dong L, Yang W. Classification of Neoadjuvant Therapy Response in Patients With Colorectal Liver Metastases Using Contrast-Enhanced Ultrasound-With Histological Pathology as the Gold Standard. ULTRASOUND IN MEDICINE & BIOLOGY 2025; 51:102-111. [PMID: 39414406 DOI: 10.1016/j.ultrasmedbio.2024.09.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 08/29/2024] [Accepted: 09/16/2024] [Indexed: 10/18/2024]
Abstract
OBJECTIVE To evaluate the response to neoadjuvant therapy in patients with colorectal liver metastases (CRLMs) using ultrasound(US) and contrast-enhanced ultrasound(CEUS), with correction to the tumor regression grade (TRG) of pathological results. METHODS This study included patients with resectable CRLMs admitted from February to December 2022. After at least 4 cycles neoadjuvant therapy, all the patients received US and CEUS examinations within two weeks before hepatectomy. CEUS clips were postprocessed with color parameter imaging (CPI) and microflow imaging (MFI) analysis. Logistic regression analyses were used to develop an evaluation Nomogram. Ultrasound-based model was constructed to discriminate between the response (TRG1/2/3) and nonresponse (TRG4/5) groups at the lesion level. The model's predictive ability was evaluated using the C index and calibration curve, with decision curve analysis assessing the Nomogram's added value. RESULTS The study analyzed 105 CRLM lesions (the lesion with the highest diameter analyzed for each patient), with 43.8% showing a response to therapy. Univariate analysis identified calcification on US (p = 0.039), CEUS enhancement degree (p < 0.001), CEUS enhancement pattern (p<0.001), CEUS washout type (p < 0.001), CEUS necrosis (p < 0.001), CPI feeding artery (p = 0.003) and MFI pattern (p < 0.001) were significantly associated with TRG. The multivariate analysis showed CEUS enhancement pattern (p = 0.026), CEUS washout type (p = 0.018) and CEUS necrosis (p = 0.005) were independently associated with the neoadjuvant therapy response. A Nomogram with the three independent predictors was developed, with an AUC of 0.898. CONCLUSION The ultrasound-based model provided accurate evaluation of pathological tumor response to preoperative chemotherapy in patients with CRLM, and may help to decide the individualized treatment strategy.
Collapse
Affiliation(s)
- Li Liu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Ultrasound, Peking University Cancer Hospital & Institute, Beijing, China
| | - Wen-Chao Cui
- Department of Ultrasonography, Shengli Oil Field Center Hospital, Dongying, Shandong Province, China
| | - Yu Sun
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Pathology, Peking University Cancer Hospital & Institute, Beijing, China
| | - Hong Wang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Ultrasound, Peking University Cancer Hospital & Institute, Beijing, China
| | - Zi-Nan Liang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Ultrasound, Peking University Cancer Hospital & Institute, Beijing, China
| | - Wei Wu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Ultrasound, Peking University Cancer Hospital & Institute, Beijing, China
| | - Kun Yan
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Ultrasound, Peking University Cancer Hospital & Institute, Beijing, China
| | - Yong-Li Ji
- Department of Ultrasonography, Shengli Oil Field Center Hospital, Dongying, Shandong Province, China
| | - Liang Dong
- Department of Ultrasonography, Shengli Oil Field Center Hospital, Dongying, Shandong Province, China
| | - Wei Yang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Ultrasound, Peking University Cancer Hospital & Institute, Beijing, China.
| |
Collapse
|
14
|
Zhang Z, Zhang W, He C, Xie J, Liang F, Zhao Y, Tan L, Lai S, Jiang X, Wei X, Zhen X, Yang R. Identification of macrotrabecular-massive hepatocellular carcinoma through multiphasic CT-based representation learning method. Med Phys 2024; 51:9017-9030. [PMID: 39311438 DOI: 10.1002/mp.17401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 07/17/2024] [Accepted: 08/21/2024] [Indexed: 12/20/2024] Open
Abstract
BACKGROUND Macrotrabecular-massive hepatocellular carcinoma (MTM-HCC) represents an aggressive subtype of HCC and is associated with poor survival. PURPOSE To investigate the performance of a representation learning-based feature fusion strategy that employs a multiphase contrast-enhanced CT (mpCECT)-based latent feature fusion (MCLFF) model for MTM-HCC identification. METHODS A total of 206 patients (54 MTM HCC, 152 non-MTM HCC) who underwent preoperative mpCECT with surgically confirmed HCC between July 2017 and December 2022 were retrospectively included from two medical centers. Multiphasic radiomics features were extracted from manually delineated volume of interest (VOI) of all lesions on each mpCECT phase. Representation learning based MCLFF model was built to fuse multiphasic features for MTM HCC prediction, and compared with competing models using other fusion methods. Conventional imaging features and clinical factors were also evaluated and analyzed. Prediction performance was validated by ROC analysis and statistical comparisons on an internal validation and an external testing dataset. RESULTS Fusion of radiomics features from the arterial phase (AP) and portal venous phase (PAP) using MCLFF demonstrated superior performance in MTM HCC prediction, with a higher AUC of 0.857 compared with all competing models in the internal validation set. Integration of multiple radiological or clinical features further improved the overall performance, with the highest AUCs of 0.857 and 0.836 respectively achieved in the internal validation and external testing set. CONCLUSIONS Multiphasic radiomics features of AP and PVP fused by the MCLFF have demonstrated substantial potential in the accurate prediction of MTM HCC. Clinical factors and Radiological features in mpCECT contribute incremental values to the developed MCLFF strategy.
Collapse
Affiliation(s)
- Zhenyang Zhang
- Department of Radiology, the Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, China
- Department of Radiology, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China
- School of Biomedical Engineering, Southern Medical University, Guangzhou, Guangdong, China
| | - Wanli Zhang
- Department of Radiology, the Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, China
- Department of Radiology, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Chutong He
- Medical Imaging Center, Jinan University First Affiliated Hospital, Guangzhou, Guangdong, China
| | - Jincheng Xie
- School of Biomedical Engineering, Southern Medical University, Guangzhou, Guangdong, China
| | - Fangrong Liang
- Department of Radiology, the Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, China
- Department of Radiology, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Yandong Zhao
- Department of Radiology, the Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, China
- Department of Radiology, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Lilian Tan
- Department of Radiology, the Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Shengsheng Lai
- School of Medical Equipment, Guangdong Food and Drug Vocational College, Guangzhou, Guangdong, China
| | - Xinqing Jiang
- Department of Radiology, the Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, China
- Department of Radiology, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Xinhua Wei
- Department of Radiology, the Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, China
- Department of Radiology, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Xin Zhen
- School of Biomedical Engineering, Southern Medical University, Guangzhou, Guangdong, China
- Medical Big Data Center, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, China
| | - Ruimeng Yang
- Department of Radiology, the Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, China
- Department of Radiology, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China
| |
Collapse
|
15
|
Zhao B, Dercle L, Yang H, Riely GJ, Kris MG, Schwartz LH. Annotated test-retest dataset of lung cancer CT scan images reconstructed at multiple imaging parameters. Sci Data 2024; 11:1259. [PMID: 39567508 PMCID: PMC11579286 DOI: 10.1038/s41597-024-04085-3] [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: 03/25/2024] [Accepted: 11/05/2024] [Indexed: 11/22/2024] Open
Abstract
Quantitative imaging biomarkers (QIB) are increasingly used in clinical research to advance precision medicine approaches in oncology. Computed tomography (CT) is a modality of choice for cancer diagnosis, prognosis, and response assessment due to its reliability and global accessibility. Here, we contribute to the cancer imaging community through The Cancer Imaging Archive (TCIA) by providing investigator-initiated, same-day repeat CT scan images of 32 non-small cell lung cancer (NSCLC) patients, along with radiologist-annotated lesion contours as a reference standard. Each scan was reconstructed into 6 image settings using various combinations of three slice thicknesses (1.25 mm, 2.5 mm, 5 mm) and two reconstruction kernels (lung, standard; GE CT equipment), which spans a wide range of CT imaging reconstruction parameters commonly used in lung cancer clinical practice and clinical trials. This holds considerable value for advancing the development of robust Radiomics, Artificial Intelligence (AI) and machine learning (ML) methods.
Collapse
Affiliation(s)
- Binsheng Zhao
- Memorial Sloan-Kettering Cancer Center, New York, NY, 10021, USA.
| | - Laurent Dercle
- Memorial Sloan-Kettering Cancer Center, New York, NY, 10021, USA
- Department of Radiology, Columbia University New York, New York, NY, 10032, USA
| | - Hao Yang
- Memorial Sloan-Kettering Cancer Center, New York, NY, 10021, USA
| | - Gregory J Riely
- Memorial Sloan-Kettering Cancer Center, New York, NY, 10021, USA
| | - Mark G Kris
- Memorial Sloan-Kettering Cancer Center, New York, NY, 10021, USA
| | | |
Collapse
|
16
|
Chen X, Chen Q, Liu Y, Qiu Y, Lv L, Zhang Z, Yin X, Shu F. Radiomics models to predict bone marrow metastasis of neuroblastoma using CT. CANCER INNOVATION 2024; 3:e135. [PMID: 38948899 PMCID: PMC11212276 DOI: 10.1002/cai2.135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/25/2023] [Revised: 02/01/2024] [Accepted: 02/02/2024] [Indexed: 07/02/2024]
Abstract
BACKGROUND Bone marrow is the leading site for metastasis from neuroblastoma and affects the prognosis of patients with neuroblastoma. However, the accurate diagnosis of bone marrow metastasis is limited by the high spatial and temporal heterogeneity of neuroblastoma. Radiomics analysis has been applied in various cancers to build accurate diagnostic models but has not yet been applied to bone marrow metastasis of neuroblastoma. METHODS We retrospectively collected information from 187 patients pathologically diagnosed with neuroblastoma and divided them into training and validation sets in a ratio of 7:3. A total of 2632 radiomics features were retrieved from venous and arterial phases of contrast-enhanced computed tomography (CT), and nine machine learning approaches were used to build radiomics models, including multilayer perceptron (MLP), extreme gradient boosting, and random forest. We also constructed radiomics-clinical models that combined radiomics features with clinical predictors such as age, gender, ascites, and lymph gland metastasis. The performance of the models was evaluated with receiver operating characteristics (ROC) curves, calibration curves, and risk decile plots. RESULTS The MLP radiomics model yielded an area under the ROC curve (AUC) of 0.97 (95% confidence interval [CI]: 0.95-0.99) on the training set and 0.90 (95% CI: 0.82-0.95) on the validation set. The radiomics-clinical model using an MLP yielded an AUC of 0.93 (95% CI: 0.89-0.96) on the training set and 0.91 (95% CI: 0.85-0.97) on the validation set. CONCLUSIONS MLP-based radiomics and radiomics-clinical models can precisely predict bone marrow metastasis in patients with neuroblastoma.
Collapse
Affiliation(s)
- Xiong Chen
- Department of Paediatric Urology, Guangzhou Women and Children's Medical CenterGuangzhou Medical UniversityGuangzhouChina
- Department of Paediatric Surgery, Guangzhou Institute of PaediatricsGuangzhou Medical UniversityGuangzhouChina
| | - Qinchang Chen
- Department of Pediatric Cardiology, Guangdong Provincial People's HospitalGuangdong Academy of Medical Sciences, Guangdong Cardiovascular Institute, Guangdong Provincial Key Laboratory of Structural Heart DiseaseGuangzhouChina
| | - Yuanfang Liu
- Department of Radiology, Sun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhouChina
| | - Ya Qiu
- Department of Radiologythe First People's Hospital of Kashi PrefectureKashiChina
| | - Lin Lv
- Medical SchoolSun Yat‐sen UniversityGuangzhouChina
| | - Zhengtao Zhang
- Department of Paediatric Urology, Guangzhou Women and Children's Medical CenterGuangzhou Medical UniversityGuangzhouChina
- Department of Paediatric Surgery, Guangzhou Institute of PaediatricsGuangzhou Medical UniversityGuangzhouChina
| | - Xuntao Yin
- Department of RadiologyGuangzhou Women and Children's Medical CenterGuangzhouChina
| | - Fangpeng Shu
- Department of Paediatric Urology, Guangzhou Women and Children's Medical CenterGuangzhou Medical UniversityGuangzhouChina
- Department of Paediatric Surgery, Guangzhou Institute of PaediatricsGuangzhou Medical UniversityGuangzhouChina
| |
Collapse
|
17
|
Dimopoulos P, Mulita A, Antzoulas A, Bodard S, Leivaditis V, Akrida I, Benetatos N, Katsanos K, Anagnostopoulos CN, Mulita F. The role of artificial intelligence and image processing in the diagnosis, treatment, and prognosis of liver cancer: a narrative-review. PRZEGLAD GASTROENTEROLOGICZNY 2024; 19:221-230. [PMID: 39802971 PMCID: PMC11718495 DOI: 10.5114/pg.2024.143147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Accepted: 03/29/2024] [Indexed: 12/09/2024]
Abstract
Artificial intelligence (AI) and image processing are revolutionising the diagnosis and management of liver cancer. Recent advancements showcase AI's ability to analyse medical imaging data, like computed tomography scans and magnetic resonance imaging, accurately detecting and classifying liver cancer lesions for early intervention. Predictive models aid prognosis estimation and recurrence pattern identification, facilitating personalised treatment planning. Image processing techniques enhance data analysis by precise segmentation of liver structures, fusion of information from multiple modalities, and feature extraction for informed decision-making. Despite progress, challenges persist, including the need for standardised datasets and regulatory considerations.
Collapse
Affiliation(s)
- Platon Dimopoulos
- Department of Interventional Radiology, General University Hospital of Patras, Patras, Greece
| | - Admir Mulita
- Medical Physics Department, Democritus University of Thrace, University Hospital of Alexandroupolis, Alexandroupolis, Greece
- Intelligent Systems Lab, Department of Cultural Technology and Communication, University of the Aegean, Mytilene, Greece
| | - Andreas Antzoulas
- Department of Surgery, General University Hospital of Patras, Patras, Greece
| | - Sylvain Bodard
- Department of Radiology, University of Paris Cite, Necker Hospital, Paris, France
| | - Vasileios Leivaditis
- Department of Cardiothoracic and Vascular Surgery, Westpfalz Klinikum, Kaiserslautern, Germany
| | - Ioanna Akrida
- Department of Surgery, General University Hospital of Patras, Patras, Greece
| | - Nikolaos Benetatos
- Department of Surgery, General University Hospital of Patras, Patras, Greece
| | - Konstantinos Katsanos
- Department of Interventional Radiology, General University Hospital of Patras, Patras, Greece
| | | | - Francesk Mulita
- Department of Surgery, General University Hospital of Patras, Patras, Greece
| |
Collapse
|
18
|
Wang N, Dai M, Jing F, Liu Y, Zhao Y, Zhang Z, Wang J, Zhang J, Wang Y, Zhao X. Value of 18F-FDG PET/CT-based radiomics features for differentiating primary lung cancer and solitary lung metastasis in patients with colorectal adenocarcinoma. Int J Radiat Biol 2024:1-9. [PMID: 39288285 DOI: 10.1080/09553002.2024.2404465] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2024] [Revised: 08/20/2024] [Accepted: 09/03/2024] [Indexed: 09/19/2024]
Abstract
OBJECTIVE To investigate the value and applicability of 18F-fluorodeoxyglucose positron emission tomography/computed tomography (18F-FDG PET/CT) radiomics in differentiating primary lung cancer (PLC) from solitary lung metastasis (SLM) in patients with colorectal cancer (CRC). MATERIALS AND METHODS This retrospective study included 103 patients with CRC and solitary pulmonary nodules (SPNs). The least absolute shrinkage and selection operator (LASSO) was used to screen for optimal radiomics features and establish a PET/CT radiomics model. PET/CT Visual and complex models (combining radiomics with PET/CT visual features) were developed. The area under the receiver operating characteristic (ROC) curve (AUC) was used to determine the predictive value and diagnostic efficiency of the models. RESULTS The AUC of the PET/CT radiomics model for differentiating PLC from SLM was 0.872 (95% CI: 0.806-0.939), which was not different from that of the visual (0.829 [95% CI: 0.749-0.908; p = .352]). However, the AUC of the complex model (0.936 [95% CI:0.892-0.981]) was significantly higher than that of the PET/CT radiomics (p = .005) and visual model (p = .001). The sensitivity (SEN), specificity (SPE), accuracy (ACC), positive predictive value (PPV), and negative predictive value (NPV) of PET/CT radiomics for differentiating PLC from SLM were 0.720, 0.887, 0.806, 0.857, and 0.770, respectively. CONCLUSION PET/CT radiomics can effectively distinguish PLC and SLM in patients with CRC and SPNs and guide the implementation of personalized treatment.
Collapse
Affiliation(s)
- Na Wang
- Department of Nuclear Medicine, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
- Hebei Provincial Key Laboratory of Tumor Microenvironment and Drug Resistance, Shijiazhuang, Hebei, China
| | - Meng Dai
- Department of Nuclear Medicine, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
- Hebei Provincial Key Laboratory of Tumor Microenvironment and Drug Resistance, Shijiazhuang, Hebei, China
| | - Fenglian Jing
- Department of Nuclear Medicine, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Yunuan Liu
- Department of Nuclear Medicine, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Yan Zhao
- Department of Oncology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Zhaoqi Zhang
- Department of Nuclear Medicine, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
- Hebei Provincial Key Laboratory of Tumor Microenvironment and Drug Resistance, Shijiazhuang, Hebei, China
| | - Jianfang Wang
- Department of Nuclear Medicine, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
- Hebei Provincial Key Laboratory of Tumor Microenvironment and Drug Resistance, Shijiazhuang, Hebei, China
| | - Jingmian Zhang
- Department of Nuclear Medicine, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
- Hebei Provincial Key Laboratory of Tumor Microenvironment and Drug Resistance, Shijiazhuang, Hebei, China
| | - Yingchen Wang
- Department of Nuclear Medicine, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Xinming Zhao
- Department of Nuclear Medicine, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
- Hebei Provincial Key Laboratory of Tumor Microenvironment and Drug Resistance, Shijiazhuang, Hebei, China
| |
Collapse
|
19
|
Montagnon E, Cerny M, Hamilton V, Derennes T, Ilinca A, Elforaici MEA, Jabbour G, Rafie E, Wu A, Perdigon Romero F, Cadrin-Chênevert A, Kadoury S, Turcotte S, Tang A. Radiomics analysis of baseline computed tomography to predict oncological outcomes in patients treated for resectable colorectal cancer liver metastasis. PLoS One 2024; 19:e0307815. [PMID: 39259736 PMCID: PMC11389941 DOI: 10.1371/journal.pone.0307815] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Accepted: 07/11/2024] [Indexed: 09/13/2024] Open
Abstract
OBJECTIVE The purpose of this study was to determine and compare the performance of pre-treatment clinical risk score (CRS), radiomics models based on computed (CT), and their combination for predicting time to recurrence (TTR) and disease-specific survival (DSS) in patients with colorectal cancer liver metastases. METHODS We retrospectively analyzed a prospectively maintained registry of 241 patients treated with systemic chemotherapy and surgery for colorectal cancer liver metastases. Radiomics features were extracted from baseline, pre-treatment, contrast-enhanced CT images. Multiple aggregation strategies were investigated for cases with multiple metastases. Radiomics signatures were derived using feature selection methods. Random survival forests (RSF) and neural network survival models (DeepSurv) based on radiomics features, alone or combined with CRS, were developed to predict TTR and DSS. Leveraging survival models predictions, classification models were trained to predict TTR within 18 months and DSS within 3 years. Classification performance was assessed with area under the receiver operating characteristic curve (AUC) on the test set. RESULTS For TTR prediction, the concordance index (95% confidence interval) was 0.57 (0.57-0.57) for CRS, 0.61 (0.60-0.61) for RSF in combination with CRS, and 0.70 (0.68-0.73) for DeepSurv in combination with CRS. For DSS prediction, the concordance index was 0.59 (0.59-0.59) for CRS, 0.57 (0.56-0.57) for RSF in combination with CRS, and 0.60 (0.58-0.61) for DeepSurv in combination with CRS. For TTR classification, the AUC was 0.33 (0.33-0.33) for CRS, 0.77 (0.75-0.78) for radiomics signature alone, and 0.58 (0.57-0.59) for DeepSurv score alone. For DSS classification, the AUC was 0.61 (0.61-0.61) for CRS, 0.57 (0.56-0.57) for radiomics signature, and 0.75 (0.74-0.76) for DeepSurv score alone. CONCLUSION Radiomics-based survival models outperformed CRS for TTR prediction. More accurate, noninvasive, and early prediction of patient outcome may help reduce exposure to ineffective yet toxic chemotherapy or high-risk major hepatectomies.
Collapse
Affiliation(s)
- Emmanuel Montagnon
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Montréal, QC, Canada
| | - Milena Cerny
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Montréal, QC, Canada
- Department of Radiology, CISSS des Laurentides, Hôpital de Saint-Eustache, Saint-Eustache, QC, Canada
| | - Vincent Hamilton
- Department of Radiology, Radiation Oncology and Nuclear Medicine, Université de Montréal, Montréal, QC, Canada
| | - Thomas Derennes
- Department of Radiology, Radiation Oncology and Nuclear Medicine, Université de Montréal, Montréal, QC, Canada
| | - André Ilinca
- Department of Radiology, Radiation Oncology and Nuclear Medicine, Université de Montréal, Montréal, QC, Canada
| | - Mohamed El Amine Elforaici
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Montréal, QC, Canada
- MedICAL Laboratory, Polytechnique Montréal, Montréal, QC, Canada
| | - Gilbert Jabbour
- Department of Radiology, Radiation Oncology and Nuclear Medicine, Université de Montréal, Montréal, QC, Canada
- Division of Internal Medicine, Department of Medicine, Hôpital du Sacré-Cœur-de-Montréal, Montréal, QC, Canada
| | - Edmond Rafie
- Department of Radiology, Radiation Oncology and Nuclear Medicine, Université de Montréal, Montréal, QC, Canada
| | - Anni Wu
- Department of Radiology, Radiation Oncology and Nuclear Medicine, Université de Montréal, Montréal, QC, Canada
| | | | | | - Samuel Kadoury
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Montréal, QC, Canada
- MedICAL Laboratory, Polytechnique Montréal, Montréal, QC, Canada
| | - Simon Turcotte
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Montréal, QC, Canada
- Hepatopancreatobiliary and Liver Transplantation Division, Department of Surgery, Centre Hospitalier de l'Université de Montréal (CHUM), Montréal, QC, Canada
| | - An Tang
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Montréal, QC, Canada
- Department of Radiology, Radiation Oncology and Nuclear Medicine, Université de Montréal, Montréal, QC, Canada
- Department of Radiology, Centre Hospitalier de l'Université de Montréal (CHUM), Montréal, QC, Canada
| |
Collapse
|
20
|
Nardone V, Marmorino F, Germani MM, Cichowska-Cwalińska N, Menditti VS, Gallo P, Studiale V, Taravella A, Landi M, Reginelli A, Cappabianca S, Girnyi S, Cwalinski T, Boccardi V, Goyal A, Skokowski J, Oviedo RJ, Abou-Mrad A, Marano L. The Role of Artificial Intelligence on Tumor Boards: Perspectives from Surgeons, Medical Oncologists and Radiation Oncologists. Curr Oncol 2024; 31:4984-5007. [PMID: 39329997 PMCID: PMC11431448 DOI: 10.3390/curroncol31090369] [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: 07/29/2024] [Revised: 08/24/2024] [Accepted: 08/26/2024] [Indexed: 09/28/2024] Open
Abstract
The integration of multidisciplinary tumor boards (MTBs) is fundamental in delivering state-of-the-art cancer treatment, facilitating collaborative diagnosis and management by a diverse team of specialists. Despite the clear benefits in personalized patient care and improved outcomes, the increasing burden on MTBs due to rising cancer incidence and financial constraints necessitates innovative solutions. The advent of artificial intelligence (AI) in the medical field offers a promising avenue to support clinical decision-making. This review explores the perspectives of clinicians dedicated to the care of cancer patients-surgeons, medical oncologists, and radiation oncologists-on the application of AI within MTBs. Additionally, it examines the role of AI across various clinical specialties involved in cancer diagnosis and treatment. By analyzing both the potential and the challenges, this study underscores how AI can enhance multidisciplinary discussions and optimize treatment plans. The findings highlight the transformative role that AI may play in refining oncology care and sustaining the efficacy of MTBs amidst growing clinical demands.
Collapse
Affiliation(s)
- Valerio Nardone
- Department of Precision Medicine, University of Campania “L. Vanvitelli”, 80131 Naples, Italy; (V.N.); (V.S.M.); (P.G.); (A.R.); (S.C.)
| | - Federica Marmorino
- Unit of Medical Oncology 2, Azienda Ospedaliera Universitaria Pisana, 56126 Pisa, Italy; (F.M.); (M.M.G.); (V.S.); (A.T.); (M.L.)
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, 56126 Pisa, Italy
| | - Marco Maria Germani
- Unit of Medical Oncology 2, Azienda Ospedaliera Universitaria Pisana, 56126 Pisa, Italy; (F.M.); (M.M.G.); (V.S.); (A.T.); (M.L.)
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, 56126 Pisa, Italy
| | | | - Vittorio Salvatore Menditti
- Department of Precision Medicine, University of Campania “L. Vanvitelli”, 80131 Naples, Italy; (V.N.); (V.S.M.); (P.G.); (A.R.); (S.C.)
| | - Paolo Gallo
- Department of Precision Medicine, University of Campania “L. Vanvitelli”, 80131 Naples, Italy; (V.N.); (V.S.M.); (P.G.); (A.R.); (S.C.)
| | - Vittorio Studiale
- Unit of Medical Oncology 2, Azienda Ospedaliera Universitaria Pisana, 56126 Pisa, Italy; (F.M.); (M.M.G.); (V.S.); (A.T.); (M.L.)
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, 56126 Pisa, Italy
| | - Ada Taravella
- Unit of Medical Oncology 2, Azienda Ospedaliera Universitaria Pisana, 56126 Pisa, Italy; (F.M.); (M.M.G.); (V.S.); (A.T.); (M.L.)
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, 56126 Pisa, Italy
| | - Matteo Landi
- Unit of Medical Oncology 2, Azienda Ospedaliera Universitaria Pisana, 56126 Pisa, Italy; (F.M.); (M.M.G.); (V.S.); (A.T.); (M.L.)
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, 56126 Pisa, Italy
| | - Alfonso Reginelli
- Department of Precision Medicine, University of Campania “L. Vanvitelli”, 80131 Naples, Italy; (V.N.); (V.S.M.); (P.G.); (A.R.); (S.C.)
| | - Salvatore Cappabianca
- Department of Precision Medicine, University of Campania “L. Vanvitelli”, 80131 Naples, Italy; (V.N.); (V.S.M.); (P.G.); (A.R.); (S.C.)
| | - Sergii Girnyi
- Department of General Surgery and Surgical Oncology, “Saint Wojciech” Hospital, “Nicolaus Copernicus” Health Center, 80-462 Gdańsk, Poland; (S.G.); (T.C.); (J.S.); (L.M.)
| | - Tomasz Cwalinski
- Department of General Surgery and Surgical Oncology, “Saint Wojciech” Hospital, “Nicolaus Copernicus” Health Center, 80-462 Gdańsk, Poland; (S.G.); (T.C.); (J.S.); (L.M.)
| | - Virginia Boccardi
- Division of Gerontology and Geriatrics, Department of Medicine and Surgery, University of Perugia, 06123 Perugia, Italy;
| | - Aman Goyal
- Adesh Institute of Medical Sciences and Research, Bathinda 151109, Punjab, India;
| | - Jaroslaw Skokowski
- Department of General Surgery and Surgical Oncology, “Saint Wojciech” Hospital, “Nicolaus Copernicus” Health Center, 80-462 Gdańsk, Poland; (S.G.); (T.C.); (J.S.); (L.M.)
- Department of Medicine, Academy of Applied Medical and Social Sciences-AMiSNS: Akademia Medycznych I Spolecznych Nauk Stosowanych, 82-300 Elbląg, Poland
| | - Rodolfo J. Oviedo
- Nacogdoches Medical Center, Nacogdoches, TX 75965, USA
- Tilman J. Fertitta Family College of Medicine, University of Houston, Houston, TX 77021, USA
- College of Osteopathic Medicine, Sam Houston State University, Conroe, TX 77304, USA
| | - Adel Abou-Mrad
- Centre Hospitalier Universitaire d’Orléans, 45100 Orléans, France;
| | - Luigi Marano
- Department of General Surgery and Surgical Oncology, “Saint Wojciech” Hospital, “Nicolaus Copernicus” Health Center, 80-462 Gdańsk, Poland; (S.G.); (T.C.); (J.S.); (L.M.)
- Department of Medicine, Academy of Applied Medical and Social Sciences-AMiSNS: Akademia Medycznych I Spolecznych Nauk Stosowanych, 82-300 Elbląg, Poland
| |
Collapse
|
21
|
Hua Y, Sun Z, Xiao Y, Li H, Ma X, Luo X, Tan W, Xie Z, Zhang Z, Tang C, Zhuang H, Xu W, Zhu H, Chen Y, Shang C. Pretreatment CT-based machine learning radiomics model predicts response in unresectable hepatocellular carcinoma treated with lenvatinib plus PD-1 inhibitors and interventional therapy. J Immunother Cancer 2024; 12:e008953. [PMID: 39029924 PMCID: PMC11261678 DOI: 10.1136/jitc-2024-008953] [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] [Accepted: 06/25/2024] [Indexed: 07/21/2024] Open
Abstract
BACKGROUND Lenvatinib plus PD-1 inhibitors and interventional (LPI) therapy have demonstrated promising treatment effects in unresectable hepatocellular carcinoma (HCC). However, biomarkers for predicting the response to LPI therapy remain to be further explored. We aimed to develop a radiomics model to noninvasively predict the efficacy of LPI therapy. METHODS Clinical data of patients with HCC receiving LPI therapy were collected in our institution. The clinical model was built with clinical information. Nine machine learning classifiers were tested and the multilayer perceptron classifier with optimal performance was used as the radiomics model. The clinical-radiomics model was constructed by integrating clinical and radiomics scores through logistic regression analysis. RESULTS 151 patients were enrolled in this study (2:1 randomization, 101 and 50 in the training and validation cohorts), of which three achieved complete response, 69 showed partial response, 46 showed stable disease, and 33 showed progressive disease. The objective response rate, disease control rate, and conversion resection rates were 47.7, 78.1 and 23.2%. 14 features were selected from the initially extracted 1223 for radiomics model construction. The area under the curves of the radiomics model (0.900 for training and 0.893 for validation) were comparable to that of the clinical-radiomics model (0.912 for training and 0.892 for validation), and both were superior to the clinical model (0.669 for training and 0.585 for validation). Meanwhile, the radiomics model can categorize participants into high-risk and low-risk groups for progression-free survival (PFS) and overall survival (OS) in the training (HR 1.913, 95% CI 1.121 to 3.265, p=0.016 for PFS; HR 4.252, 95% CI 2.051 to 8.816, p=0.001 for OS) and validation sets (HR 2.347, 95% CI 1.095 to 5.031, p=0.012 for PFS; HR 2.592, 95% CI 1.050 to 6.394, p=0.019 for OS). CONCLUSION The promising machine learning radiomics model was developed and validated to predict the efficacy of LPI therapy for patients with HCC and perform risk stratification, with comparable performance to clinical-radiomics model.
Collapse
Affiliation(s)
- Yonglin Hua
- Department of Hepatobiliary Surgery, Sun Yat-Sen Memorial Hospital, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Zhixian Sun
- Breast Disease Center, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
- Laboratory of General Surgery, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
- Breast Tumor Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yuxin Xiao
- Department of Hepatobiliary Surgery, Sun Yat-Sen Memorial Hospital, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Huilong Li
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Xiaowu Ma
- Department of Hepatobiliary Surgery, Sun Yat-Sen Memorial Hospital, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Xuan Luo
- Department of Hepatobiliary Surgery, Sun Yat-Sen Memorial Hospital, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Wenliang Tan
- Department of Hepatobiliary and Pancreatic Surgery, Zhuzhou Hospital Affiliated to Central South University Xiangya School of Medicine, Zhuzhou, Hunan, China
| | - Zhiqin Xie
- Department of Hepatobiliary and Pancreatic Surgery, Zhuzhou Hospital Affiliated to Central South University Xiangya School of Medicine, Zhuzhou, Hunan, China
| | - Ziyu Zhang
- Department of Hepatobiliary Surgery, Sun Yat-Sen Memorial Hospital, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Chenwei Tang
- Department of Hepatobiliary Surgery, Sun Yat-Sen Memorial Hospital, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Hongkai Zhuang
- Department of Hepatobiliary Surgery, Sun Yat-Sen Memorial Hospital, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Weikai Xu
- Department of Hepatobiliary Surgery, Sun Yat-Sen Memorial Hospital, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Haihong Zhu
- Department of General Surgery, Qinghai Provincial People’s Hospital, Xining, Qinghai, China
| | - Yajin Chen
- Department of Hepatobiliary Surgery, Sun Yat-Sen Memorial Hospital, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Changzhen Shang
- Department of Hepatobiliary Surgery, Sun Yat-Sen Memorial Hospital, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
| |
Collapse
|
22
|
Napolitano S, Martini G, Ciardiello D, Del Tufo S, Martinelli E, Troiani T, Ciardiello F. Targeting the EGFR signalling pathway in metastatic colorectal cancer. Lancet Gastroenterol Hepatol 2024; 9:664-676. [PMID: 38697174 DOI: 10.1016/s2468-1253(23)00479-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Revised: 12/21/2023] [Accepted: 12/28/2023] [Indexed: 05/04/2024]
Abstract
Epidermal growth factor receptor (EGFR) and its activated downstream signalling pathways play a crucial role in colorectal cancer development and progression. After four decades of preclinical, translational, and clinical research, it has been shown that blocking the EGFR signalling pathway at different molecular levels represents a fundamental therapeutic strategy for patients with metastatic colorectal cancer. Nevertheless, the efficacy of molecularly targeted therapies is inescapably limited by the insurgence of mechanisms of acquired cancer cell resistance. Thus, in the era of precision medicine, a deeper understanding of the complex molecular landscape of metastatic colorectal cancer is required to deliver the best treatment choices to all patients. Major efforts are currently ongoing to improve patient selection, improve the efficacy of available treatments targeting the EGFR pathway, and develop novel combination strategies to overcome therapy resistance within the continuum of care of metastatic colorectal cancer.
Collapse
Affiliation(s)
- Stefania Napolitano
- Department of Precision Medicine, Università degli studi della Campania Luigi Vanvitelli, Napoli, Italy
| | - Giulia Martini
- Department of Precision Medicine, Università degli studi della Campania Luigi Vanvitelli, Napoli, Italy
| | - Davide Ciardiello
- Department of Precision Medicine, Università degli studi della Campania Luigi Vanvitelli, Napoli, Italy; Division of Gastrointestinal Medical Oncology and Neuroendocrine Tumors, European Institute of Oncology, IEO, IRCCS, Milan, Italy
| | - Sara Del Tufo
- Department of Precision Medicine, Università degli studi della Campania Luigi Vanvitelli, Napoli, Italy
| | - Erika Martinelli
- Department of Precision Medicine, Università degli studi della Campania Luigi Vanvitelli, Napoli, Italy
| | - Teresa Troiani
- Department of Precision Medicine, Università degli studi della Campania Luigi Vanvitelli, Napoli, Italy
| | - Fortunato Ciardiello
- Department of Precision Medicine, Università degli studi della Campania Luigi Vanvitelli, Napoli, Italy.
| |
Collapse
|
23
|
Granata V, Fusco R, Setola SV, Brunese MC, Di Mauro A, Avallone A, Ottaiano A, Normanno N, Petrillo A, Izzo F. Machine learning and radiomics analysis by computed tomography in colorectal liver metastases patients for RAS mutational status prediction. LA RADIOLOGIA MEDICA 2024; 129:957-966. [PMID: 38761342 DOI: 10.1007/s11547-024-01828-5] [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: 01/17/2024] [Accepted: 05/13/2024] [Indexed: 05/20/2024]
Abstract
PURPOSE To assess the efficacy of machine learning and radiomics analysis by computed tomography (CT) in presurgical setting, to predict RAS mutational status in colorectal liver metastases. METHODS Patient selection in a retrospective study was carried out from January 2018 to May 2021 considering the following inclusion criteria: patients subjected to surgical resection for liver metastases; proven pathological liver metastases; patients subjected to enhanced CT examination in the presurgical setting with a good quality of images; and RAS assessment as standard reference. A total of 851 radiomics features were extracted using the PyRadiomics Python package from the Slicer 3D image computing platform after slice-by-slice segmentation on CT portal phase by two expert radiologists of each individual liver metastasis performed first independently by the individual reader and then in consensus. Balancing technique was performed, and inter- and intraclass correlation coefficients were calculated to assess the between-observer and within-observer reproducibility of features. Receiver operating characteristics (ROC) analysis with the calculation of area under the ROC curve (AUC), sensitivity (SENS), specificity (SPEC), positive predictive value (PPV), negative predictive value (NPV) and accuracy (ACC) were assessed for each parameter. Linear and non-logistic regression model (LRM and NLRM) and different machine learning-based classifiers were considered. Moreover, features selection was performed before and after a normalized procedure using two different methods (3-sigma and z-score). RESULTS Seventy-seven liver metastases in 28 patients with a mean age of 60 years (range 40-80 years) were analyzed. The best predictors, at univariate analysis for both normalized procedures, were original_shape_Maximum2DDiameter and wavelet_HLL_glcm_InverseVariance that reached an accuracy of 80%, an AUC ≥ 0.75, a sensitivity ≥ 80% and a specificity ≥ 70% (p value < < 0.01). However, a multivariate analysis significantly increased the accuracy in RAS prediction when a linear regression model (LRM) was used. The best performance was obtained using a LRM combining linearly 12 robust features after a z-score normalization procedure: AUC of 0.953, accuracy 98%, sensitivity 96%, specificity of 100%, PPV 100% and NPV 96% (p value < < 0.01). No statistically significant increase was obtained considering the tested machine learning both without normalization and with normalization methods. CONCLUSIONS Normalized approach in CT radiomics analysis allows to predict RAS mutational status in colorectal liver metastases patients.
Collapse
Affiliation(s)
- Vincenza Granata
- Division of Radiology, Istituto Nazionale Tumori IRCCS Fondazione Pascale-IRCCS di Napoli, Naples, Italy.
| | | | - Sergio Venanzio Setola
- Division of Radiology, Istituto Nazionale Tumori IRCCS Fondazione Pascale-IRCCS di Napoli, Naples, Italy
| | - Maria Chiara Brunese
- Department of Medicine and Health Sciences V. Tiberio, University of Molise, 86100, Campobasso, Italy
| | - Annabella Di Mauro
- Pathological Anatomy and Cytopathology Unit, Istituto Nazionale Tumori, IRCCS Fondazione G. Pascale, 80131, Naples, Italy
| | - Antonio Avallone
- Clinical Sperimental Abdominal Oncology Unit, Istituto Nazionale Tumori, IRCCS Fondazione G. Pascale, 80131, Naples, Italy
| | - Alessandro Ottaiano
- Clinical Sperimental Abdominal Oncology Unit, Istituto Nazionale Tumori, IRCCS Fondazione G. Pascale, 80131, Naples, Italy
| | - Nicola Normanno
- IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", 47014, Mendola, Italy
| | - Antonella Petrillo
- Division of Radiology, Istituto Nazionale Tumori IRCCS Fondazione Pascale-IRCCS di Napoli, Naples, Italy
| | - Francesco Izzo
- Division of Epatobiliary Surgical Oncology, Istituto Nazionale Tumori IRCCS Fondazione Pascale-IRCCS di Napoli, 80131, Naples, Italy
| |
Collapse
|
24
|
Liu X, Li H, Wang S, Yang S, Zhang G, Xu Y, Yang H, Shan F. CT radiomics to differentiate neuroendocrine neoplasm from adenocarcinoma in patients with a peripheral solid pulmonary nodule: a multicenter study. Front Oncol 2024; 14:1420213. [PMID: 38952551 PMCID: PMC11215045 DOI: 10.3389/fonc.2024.1420213] [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: 04/19/2024] [Accepted: 06/03/2024] [Indexed: 07/03/2024] Open
Abstract
Purpose To construct and validate a computed tomography (CT) radiomics model for differentiating lung neuroendocrine neoplasm (LNEN) from lung adenocarcinoma (LADC) manifesting as a peripheral solid nodule (PSN) to aid in early clinical decision-making. Methods A total of 445 patients with pathologically confirmed LNEN and LADC from June 2016 to July 2023 were retrospectively included from five medical centers. Those patients were split into the training set (n = 316; 158 LNEN) and external test set (n = 129; 43 LNEN), the former including the cross-validation (CV) training set and CV test set using ten-fold CV. The support vector machine (SVM) classifier was used to develop the semantic, radiomics and merged models. The diagnostic performances were evaluated by the area under the receiver operating characteristic curve (AUC) and compared by Delong test. Preoperative neuron-specific enolase (NSE) levels were collected as a clinical predictor. Results In the training set, the AUCs of the radiomics model (0.878 [95% CI: 0.836, 0.915]) and merged model (0.884 [95% CI: 0.844, 0.919]) significantly outperformed the semantic model (0.718 [95% CI: 0.663, 0.769], p both<.001). In the external test set, the AUCs of the radiomics model (0.787 [95% CI: 0.696, 0.871]), merged model (0.807 [95%CI: 0.720, 0.889]) and semantic model (0.729 [95% CI: 0.631, 0.811]) did not exhibit statistical differences. The radiomics model outperformed NSE in sensitivity in the training set (85.3% vs 20.0%; p <.001) and external test set (88.9% vs 40.7%; p = .002). Conclusion The CT radiomics model could non-invasively, effectively and sensitively predict LNEN and LADC presenting as a PSN to assist in treatment strategy selection.
Collapse
Affiliation(s)
- Xiaoyu Liu
- Department of Radiology, Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Hongjian Li
- Department of Radiology, Affiliated Hospital of North Sichuan Medical College, North Sichuan Medical College, Nanchong, China
| | - Shengping Wang
- Department of Radiology, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, China
| | - Shan Yang
- Department of Radiology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Guobin Zhang
- Department of Radiology, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yonghua Xu
- Department of Imaging and Interventional Radiology, Zhongshan-Xuhui Hospital of Fudan University, Fudan University, Shanghai, China
| | - Hanfeng Yang
- Department of Radiology, Affiliated Hospital of North Sichuan Medical College, North Sichuan Medical College, Nanchong, China
| | - Fei Shan
- Department of Radiology, Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| |
Collapse
|
25
|
Wang Y, Wang X, Chen J, Huang S, Huang Y. Comparative analysis of preoperative chemoradiotherapy and upfront surgery in the treatment of upper-half rectal cancer: oncological benefits, surgical outcomes, and cost implications. Updates Surg 2024; 76:949-962. [PMID: 38240957 DOI: 10.1007/s13304-023-01744-9] [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: 11/11/2023] [Accepted: 12/24/2023] [Indexed: 05/28/2024]
Abstract
The value of neoadjuvant chemoradiotherapy (CRT) is not absolutely clear for upper-half (> 7-15 cm) rectal cancer. This study aimed to compare the efficacy and safety of radical surgery with preoperative CRT vs. upfront surgery (US) in Chinese patients with stage II and III upper-half rectal cancer. A total of 809 patients with locally advanced upper-half rectal cancer between 2017 and 2021 were enrolled retrospectively (280 treated with CRT and 529 treated with US). Through 1:1 propensity score matching, the CRT (172 patients) and US (172 patients) groups were compared for short-term postoperative results and long-term oncological and functional outcomes. In the entire cohort, patients in the CRT group had a younger age, lower distance from the anal verge (DAV), and higher rates of cT4 stage, cN2 stage, mrCRM positivity, EMVI positivity, CEA elevation, and CA-199 elevation than those in the US group. The 5-year disease-free survival (DFS) was lower in the CRT group than in the US group (76% vs. 84%, p = 0.022), while the 5-year overall survival (OS) was comparable between the two groups (85% and 88%, p = 0.084). The distant metastasis rate was higher in the CRT group than in the US group (12.5% vs. 7.8%, p = 0.028), though the local recurrence rate was similar between the two groups (1.1% and 1.3%, p = 1.000). After performing PSM, the 5-year OS (86% vs. 88% p = 0.312), the 5-year DFS (79% vs. 80%, p = 0.435), the local recurrence rate (1.2% vs. 1.7%, p = 1.000), and the distant metastasis rate (11.0% vs. 9.3%, p = 0.593) were comparable between the two groups. Notable pathological downstaging was observed in the CRT group, with a pathological complete response (PCR) rate of 14.5%. In addition, patients in the CRT group had a lower proportion of pT3 (61.6% vs. 77.9%, p < 0.001), pN + (pN1, 15.1% vs. 30.2%, pN2, 9.3% vs. 20.3%, p < 0.001), stage III (24.4% vs. 50.6%, p < 0.001), perineural invasion (19.8% vs. 32.0%, p = 0.014), and lymphovascular invasion (9.3% vs. 25.6%, p < 0.001) than those in the US group. Postoperative complications and long-term functional results were similar, yet there was a trend toward a higher conversion to laparotomy rate (5 (2.9%) vs. 0 (0.0%), p = 0.061) and higher rates of robotic surgery (11.6% vs. 4.7%, p < 0.001), open surgery (7.0% vs. 0.6%, p < 0.001), diverting stoma (47.1% vs. 25.6%, p < 0.001), and surgery costs (1473.6 ± 106.5 vs. 1140.3 ± 54.3$, p = 0.006) in the CRT group. In addition, EMVI (OR = 2.516, p = 0.001) was the only independent risk factor associated with poor response to CRT, and in subgroup analysis of EMVI + , CRT group patients presented a lower 5-year DFS (72.9% vs. 80.5%, p = 0.025) compared to US group patients. CRT prior to surgery has no additional oncological benefits over US in the treatment of upper-half rectal cancer. In contrast, CRT is associated with increased rates of conversion to laparotomy, stoma creation and higher surgery costs. Surgeons tend to favor robotic surgery in the treatment of complex cases such as radiated upper-half rectal cancers. Notably, EMVI + patients with upper-half rectal cancer should be encouraged to undergo upfront surgery, as preoperative CRT may not provide benefits and may lead to delayed treatment effects.
Collapse
Affiliation(s)
- Yangyang Wang
- Department of Colorectal Surgery, Union Hospital, Fujian Medical University, 29 Xin-Quan Road, Fuzhou, Fujian, 350001, People's Republic of China
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Shandong First Medical University, Tai'an, People's Republic of China
| | - Xiaojie Wang
- Department of Colorectal Surgery, Union Hospital, Fujian Medical University, 29 Xin-Quan Road, Fuzhou, Fujian, 350001, People's Republic of China
| | - Jinhua Chen
- Follow-Up Center, Union Hospital, Fujian Medical University, Fuzhou, People's Republic of China
| | - Shenghui Huang
- Department of Colorectal Surgery, Union Hospital, Fujian Medical University, 29 Xin-Quan Road, Fuzhou, Fujian, 350001, People's Republic of China
| | - Ying Huang
- Department of Colorectal Surgery, Union Hospital, Fujian Medical University, 29 Xin-Quan Road, Fuzhou, Fujian, 350001, People's Republic of China.
| |
Collapse
|
26
|
Xie X, Li D, Pei Y, Zhu W, Du X, Jiang X, Zhang L, Wang HQ. Personalized anti-tumor drug efficacy prediction based on clinical data. Heliyon 2024; 10:e27300. [PMID: 38500995 PMCID: PMC10945121 DOI: 10.1016/j.heliyon.2024.e27300] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Revised: 02/27/2024] [Accepted: 02/27/2024] [Indexed: 03/20/2024] Open
Abstract
Anti-tumor drug efficacy prediction poses an unprecedented challenge to realizing personalized medicine. This paper proposes to predict personalized anti-tumor drug efficacy based on clinical data. Specifically, we encode the clinical text as numeric vectors featured with hidden topics for patients using Latent Dirichlet Allocation model. Then, to classify patients into two classes, responsive or non-responsive to a drug, drug efficacy predictors are established by machine learning based on the Latent Dirichlet Allocation topic representation. To evaluate the proposed method, we collected and collated clinical records of lung and bowel cancer patients treated with platinum. Experimental results on the data sets show the efficacy and effectiveness of the proposed method, suggesting the potential value of clinical data in cancer precision medicine. We hope that it will promote the research of drug efficacy prediction based on clinical data.
Collapse
Affiliation(s)
- Xinping Xie
- School of Mathematics and Physics, Anhui Jianzhu University, Hefei, China
| | - Dandan Li
- School of Mathematics and Physics, Anhui Jianzhu University, Hefei, China
| | - Yangyang Pei
- School of Mathematics and Physics, Anhui Jianzhu University, Hefei, China
| | - Weiwei Zhu
- Institute of Intelligent Machines/Zhongqi AI Lab., Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, China
| | - Xiaodong Du
- Experimental Teaching Center, Hefei University, Hefei, China
| | - Xiaodong Jiang
- Medical Oncology Department, The First Affiliated Hospital of University of Science and Technology of China, Hefei, Anhui, 230001, China
| | - Lei Zhang
- Pharmacy Department, The First Affiliated Hospital of University of Science and Technology of China, Hefei, Anhui, 230001, China
| | - Hong-Qiang Wang
- Institute of Intelligent Machines/Zhongqi AI Lab., Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, China
| |
Collapse
|
27
|
Granata V, Fusco R, Brunese MC, Di Mauro A, Avallone A, Ottaiano A, Izzo F, Normanno N, Petrillo A. Machine learning-based radiomics analysis in predicting RAS mutational status using magnetic resonance imaging. LA RADIOLOGIA MEDICA 2024; 129:420-428. [PMID: 38308061 DOI: 10.1007/s11547-024-01779-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/25/2023] [Accepted: 01/04/2024] [Indexed: 02/04/2024]
Abstract
PURPOSE To assess the efficacy of radiomics features, obtained by magnetic resonance imaging (MRI) with hepatospecific contrast agent, in pre-surgical setting, to predict RAS mutational status in liver metastases. METHODS Patients with MRI in pre-surgical setting were enrolled in a retrospective study. Manual segmentation was made by means 3D Slicer image computing, and 851 radiomics features were extracted as median values using the PyRadiomics Python package. The features were extracted considering the agreement with the Imaging Biomarker Standardization Initiative (IBSI). Balancing was performed through synthesis of samples for the underrepresented classes using the self-adaptive synthetic oversampling (SASYNO) approach. Inter- and intraclass correlation coefficients (ICC) were calculated to assess the between-observer and within-observer reproducibility of all radiomics characteristics. For continuous variables, nonparametric Wilcoxon-Mann-Whitney test was utilized. Benjamini and Hochberg's false discovery rate (FDR) adjustment for multiple testing was used. Receiver operating characteristics (ROC) analysis with the calculation of area under the ROC curve (AUC), sensitivity (SENS), specificity (SPEC), positive predictive value (PPV), negative predictive value (NPV) and accuracy (ACC) were assessed for each parameter. Linear and non-logistic regression model (LRM and NLRM) and different machine learning-based classifiers including decision tree (DT), k-nearest neighbor (KNN) and support vector machine (SVM) were considered. Moreover, features selection were performed before and after a normalized procedure using two different methods (3-sigma and z-score). McNemar test was used to assess differences statistically significant between dichotomic tables. All statistical procedures were done using MATLAB R2021b Statistics and Machine Toolbox (MathWorks, Natick, MA, USA). RESULTS Seven normalized radiomics features, extracted from arterial phase, 11 normalized radiomics features, from portal phase, 12 normalized radiomics features from hepatobiliary phase and 12 normalized features from T2-W SPACE sequence were robust predictors of RAS mutational status. The multivariate analysis increased significantly the accuracy in RAS prediction when a LRM was used, combining 12 robust normalized features extracted by VIBE hepatobiliary phase reaching an accuracy of 99%, a sensitivity 97%, a specificity of 100%, a PPV of 100% and a NPV of 98%. No statistically significant increase was obtained, considering the tested classifiers DT, KNN and SVM, both without normalization and with normalization methods. CONCLUSIONS Normalized approach in MRI radiomics analysis allows to predict RAS mutational status.
Collapse
Affiliation(s)
- Vincenza Granata
- Radiology Unit, Istituto Nazionale Tumori IRCCS Fondazione Pascale-IRCCS di Napoli, Naples, Italy.
| | | | - Maria Chiara Brunese
- Department of Medicine and Health Sciences V. Tiberio, University of Molise, 86100, Campobasso, Italy
| | - Annabella Di Mauro
- Pathological Anatomy and Cytopathology Unit, Istituto Nazionale Tumori, IRCCS Fondazione G. Pascale, 80131, Naples, Italy
| | - Antonio Avallone
- Clinical Sperimental Abdominal Oncology Unit, Istituto Nazionale Tumori, IRCCS Fondazione G. Pascale, 80131, Naples, Italy
| | - Alessandro Ottaiano
- Clinical Sperimental Abdominal Oncology Unit, Istituto Nazionale Tumori, IRCCS Fondazione G. Pascale, 80131, Naples, Italy
| | - Francesco Izzo
- Epatobiliary Surgical Oncology Unit, Istituto Nazionale Tumori IRCCS Fondazione Pascale-IRCCS di Napoli, 80131, Naples, Italy
| | - Nicola Normanno
- Cell Biology and Biotherapy Unit, Istituto Nazionale Tumori IRCCS Fondazione Pascale-IRCCS Di Napoli, 80131, Naples, Italy
| | - Antonella Petrillo
- Radiology Unit, Istituto Nazionale Tumori IRCCS Fondazione Pascale-IRCCS di Napoli, Naples, Italy
| |
Collapse
|
28
|
Maino C, Vernuccio F, Cannella R, Franco PN, Giannini V, Dezio M, Pisani AR, Blandino AA, Faletti R, De Bernardi E, Ippolito D, Gatti M, Inchingolo R. Radiomics and liver: Where we are and where we are headed? Eur J Radiol 2024; 171:111297. [PMID: 38237517 DOI: 10.1016/j.ejrad.2024.111297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 01/03/2024] [Accepted: 01/07/2024] [Indexed: 02/10/2024]
Abstract
Hepatic diffuse conditions and focal liver lesions represent two of the most common scenarios to face in everyday radiological clinical practice. Thanks to the advances in technology, radiology has gained a central role in the management of patients with liver disease, especially due to its high sensitivity and specificity. Since the introduction of computed tomography (CT) and magnetic resonance imaging (MRI), radiology has been considered the non-invasive reference modality to assess and characterize liver pathologies. In recent years, clinical practice has moved forward to a quantitative approach to better evaluate and manage each patient with a more fitted approach. In this setting, radiomics has gained an important role in helping radiologists and clinicians characterize hepatic pathological entities, in managing patients, and in determining prognosis. Radiomics can extract a large amount of data from radiological images, which can be associated with different liver scenarios. Thanks to its wide applications in ultrasonography (US), CT, and MRI, different studies were focused on specific aspects related to liver diseases. Even if broadly applied, radiomics has some advantages and different pitfalls. This review aims to summarize the most important and robust studies published in the field of liver radiomics, underlying their main limitations and issues, and what they can add to the current and future clinical practice and literature.
Collapse
Affiliation(s)
- Cesare Maino
- Department of Radiology, Fondazione IRCCS San Gerardo dei Tintori, Monza 20900, Italy.
| | - Federica Vernuccio
- Institute of Radiology, University Hospital of Padova, Padova 35128, Italy
| | - Roberto Cannella
- Department of Biomedicine, Neuroscience and Advanced Diagnostics (BiND), University of Palermo, Palermo 90127, Italy
| | - Paolo Niccolò Franco
- Department of Radiology, Fondazione IRCCS San Gerardo dei Tintori, Monza 20900, Italy
| | - Valentina Giannini
- Department of Surgical Sciences, University of Turin, Turin 10126, Italy
| | - Michele Dezio
- Department of Radiology, Miulli Hospital, Acquaviva delle Fonti 70021, Bari, Italy
| | - Antonio Rosario Pisani
- Nuclear Medicine Unit, Interdisciplinary Department of Medicine, University of Bari, Bari 70121, Italy
| | - Antonino Andrea Blandino
- Department of Biomedicine, Neuroscience and Advanced Diagnostics (BiND), University of Palermo, Palermo 90127, Italy
| | - Riccardo Faletti
- Department of Surgical Sciences, University of Turin, Turin 10126, Italy
| | - Elisabetta De Bernardi
- Bicocca Bioinformatics Biostatistics and Bioimaging Centre - B4, University of Milano Bicocca, Milano 20100, Italy; School of Medicine, University of Milano Bicocca, Milano 20100, Italy
| | - Davide Ippolito
- Department of Radiology, Fondazione IRCCS San Gerardo dei Tintori, Monza 20900, Italy; School of Medicine, University of Milano Bicocca, Milano 20100, Italy
| | - Marco Gatti
- Department of Surgical Sciences, University of Turin, Turin 10126, Italy
| | - Riccardo Inchingolo
- Unit of Interventional Radiology, F. Miulli Hospital, Acquaviva delle Fonti 70021, Italy
| |
Collapse
|
29
|
Sheng L, Yang C, Chen Y, Song B. Machine Learning Combined with Radiomics Facilitating the Personal Treatment of Malignant Liver Tumors. Biomedicines 2023; 12:58. [PMID: 38255165 PMCID: PMC10813632 DOI: 10.3390/biomedicines12010058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Revised: 12/13/2023] [Accepted: 12/18/2023] [Indexed: 01/24/2024] Open
Abstract
In the realm of managing malignant liver tumors, the convergence of radiomics and machine learning has redefined the landscape of medical practice. The field of radiomics employs advanced algorithms to extract thousands of quantitative features (including intensity, texture, and structure) from medical images. Machine learning, including its subset deep learning, aids in the comprehensive analysis and integration of these features from diverse image sources. This potent synergy enables the prediction of responses of malignant liver tumors to various treatments and outcomes. In this comprehensive review, we examine the evolution of the field of radiomics and its procedural framework. Furthermore, the applications of radiomics combined with machine learning in the context of personalized treatment for malignant liver tumors are outlined in aspects of surgical therapy and non-surgical treatments such as ablation, transarterial chemoembolization, radiotherapy, and systemic therapies. Finally, we discuss the current challenges in the amalgamation of radiomics and machine learning in the study of malignant liver tumors and explore future opportunities.
Collapse
Affiliation(s)
- Liuji Sheng
- Department of Radiology, West China Hospital, Sichuan University, Chengdu 610041, China; (L.S.); (C.Y.)
- Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Chongtu Yang
- Department of Radiology, West China Hospital, Sichuan University, Chengdu 610041, China; (L.S.); (C.Y.)
- Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yidi Chen
- Department of Radiology, West China Hospital, Sichuan University, Chengdu 610041, China; (L.S.); (C.Y.)
- Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Bin Song
- Department of Radiology, West China Hospital, Sichuan University, Chengdu 610041, China; (L.S.); (C.Y.)
- Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu 610041, China
- Department of Radiology, Sanya People’s Hospital, Sanya 572000, China
| |
Collapse
|
30
|
Dercle L, Yang M, Gönen M, Flynn J, Moskowitz CS, Connors DE, Yang H, Lu L, Reidy-Lagunes D, Fojo T, Karovic S, Zhao B, Schwartz LH, Henick BS. Ethnic diversity in treatment response for colorectal cancer: proof of concept for radiomics-driven enrichment trials. Eur Radiol 2023; 33:9254-9261. [PMID: 37368111 DOI: 10.1007/s00330-023-09862-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 05/11/2023] [Accepted: 05/15/2023] [Indexed: 06/28/2023]
Abstract
BACKGROUND Several barriers hamper recruitment of diverse patient populations in multicenter clinical trials which determine efficacy of new systemic cancer therapies. PURPOSE We assessed if quantitative analysis of computed tomography (CT) scans of metastatic colorectal cancer (mCRC) patients using imaging features that predict overall survival (OS) can unravel the association between ethnicity and efficacy. METHODS We retrospectively analyzed CT images from 1584 mCRC patients in two phase III trials evaluating FOLFOX ± panitumumab (n = 331, 350) and FOLFIRI ± aflibercept (n = 437, 466) collected from August 2006 to March 2013. Primary and secondary endpoints compared RECIST1.1 response at month-2 and delta tumor volume at month-2, respectively. An ancillary study compared imaging phenotype using a peer-reviewed radiomics-signature combining 3 imaging features to predict OS landmarked from month-2. Analysis was stratified by ethnicity. RESULTS In total, 1584 patients were included (mean age, 60.25 ± 10.57 years; 969 men). Ethnicity was as follows: African (n = 50, 3.2%), Asian (n = 66, 4.2%), Caucasian (n = 1413, 89.2%), Latino (n = 27, 1.7%), Other (n = 28, 1.8%). Overall baseline tumor volume demonstrated Africans and Caucasians had more advanced disease (p < 0.001). Ethnicity was associated with treatment response. Response per RECIST1.1 at month-2 was distinct between ethnicities (p = 0.048) with higher response rate (55.6%) in Latinos. Overall delta tumor volume at month-2 demonstrated that Latino patients more likely experienced response to treatment (p = 0.021). Radiomics phenotype was also distinct in terms of tumor radiomics heterogeneity (p = 0.023). CONCLUSION This study highlights how clinical trials that inadequately represent minority groups may impact associated translational work. In appropriately powered studies, radiomics features may allow us to unravel associations between ethnicity and treatment efficacy, better elucidate mechanisms of resistance, and promote diversity in trials through predictive enrichment. CLINICAL RELEVANCE STATEMENT Radiomics could promote clinical trial diversity through predictive enrichment, hence benefit to historically underrepresented racial/ethnic groups that may respond variably to treatment due to socioeconomic factors and built environment, collectively referred to as social determinants of health. KEY POINTS •Findings indicate ethnicity was associated with treatment response across all 3 endpoints. First, response per RECIST1.1 at month-2 was distinct between ethnicities (p = 0.048) with higher response rate (55.6%) in Latinos. •Second, the overall delta tumor volume at month-2 demonstrated that Latino patients were more likely to experience response to treatment (p = 0.021). Radiomics phenotype was also distinct in terms of tumor radiomics heterogeneity (p = 0.023).
Collapse
Affiliation(s)
- Laurent Dercle
- Department of Radiology, Columbia University Irving Medical Center and New York Presbyterian Hospital, 710 West 168th St, New York, NY, 10032, USA.
| | - Melissa Yang
- Department of Radiology, Columbia University Irving Medical Center and New York Presbyterian Hospital, 710 West 168th St, New York, NY, 10032, USA
| | - Mithat Gönen
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA
| | - Jessica Flynn
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA
| | - Chaya S Moskowitz
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA
| | - Dana E Connors
- Foundation for the National Institutes of Health (FNIH), 11400 Rockville Pike, Suite 600, North Bethesda, MD, 20852, USA
| | - Hao Yang
- Department of Radiology, Columbia University Irving Medical Center and New York Presbyterian Hospital, 710 West 168th St, New York, NY, 10032, USA
| | - Lin Lu
- Department of Radiology, Columbia University Irving Medical Center and New York Presbyterian Hospital, 710 West 168th St, New York, NY, 10032, USA
| | - Diane Reidy-Lagunes
- Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA
| | - Tito Fojo
- Columbia University Herbert Irving Comprehensive Cancer Center, 161 Fort Washington Ave, New York, NY, 10032, USA
| | - Sanja Karovic
- Inova Center for Personalized Health and Schar Cancer Institute, 8100 Innovation Park Dr, Fairfax, VA, 22031, USA
- University of Virginia Cancer Center, 1240 Lee St, Charlottesville, VA, 22903, USA
| | - Binsheng Zhao
- Department of Radiology, Columbia University Irving Medical Center and New York Presbyterian Hospital, 710 West 168th St, New York, NY, 10032, USA
| | - Lawrence H Schwartz
- Department of Radiology, Columbia University Irving Medical Center and New York Presbyterian Hospital, 710 West 168th St, New York, NY, 10032, USA
| | - Brian S Henick
- Columbia University Herbert Irving Comprehensive Cancer Center, 161 Fort Washington Ave, New York, NY, 10032, USA
| |
Collapse
|
31
|
Ricci Lara MA, Esposito MI, Aineseder M, López Grove R, Cerini MA, Verzura MA, Luna DR, Benítez SE, Spina JC. Radiomics and Machine Learning for prediction of two-year disease-specific mortality and KRAS mutation status in metastatic colorectal cancer. Surg Oncol 2023; 51:101986. [PMID: 37729816 DOI: 10.1016/j.suronc.2023.101986] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 08/23/2023] [Accepted: 09/07/2023] [Indexed: 09/22/2023]
Abstract
PURPOSE Colorectal cancer is usually accompanied by liver metastases. The prediction of patient evolution is essential for the choice of the appropriate therapy. The aim of this study is to develop and evaluate machine learning models to predict KRAS gene mutations and 2-year disease-specific mortality from medical images. METHODS Clinical and follow-up information was collected from patients with metastatic colorectal cancer who had undergone computed tomography prior to liver resection. The dominant liver lesion was segmented in each scan and radiomic features were extracted from the volumes of interest. The 65% of the cases were employed to perform feature selection and to train machine learning algorithms through cross-validation. The best performing models were assembled and evaluated in the remaining cases of the cohort. RESULTS For the mortality model development, 101 cases were used as training set (64 alive, 37 deceased) and 35 as test set (22 alive, 13 deceased); while for KRAS mutation models, 55 cases were used for training (31 wild-type, 24 mutated) and 30 for testing (17 wild-type, 13 mutated). The ensemble of top performing models resulted in an area under the receiver operating characteristic curve of 0.878 for mortality and 0.905 for KRAS prediction. CONCLUSIONS Predicting the prognosis of patients with metastatic colorectal cancer is useful for making timely decisions about the best treatment options. This study presents a noninvasive method based on quantitative analysis of baseline images to identify factors influencing patient outcomes, with the aim of incorporating these tools as support systems.
Collapse
Affiliation(s)
- María Agustina Ricci Lara
- Health Informatics Department, Hospital Italiano de Buenos Aires, Tte. Gral. Juan Domingo Perón 4190, 1199, Ciudad Autónoma de Buenos Aires, Argentina; Universidad Tecnológica Nacional, Av. Medrano 951, 1179, Ciudad Autónoma de Buenos Aires, Argentina.
| | - Marco Iván Esposito
- Health Informatics Department, Hospital Italiano de Buenos Aires, Tte. Gral. Juan Domingo Perón 4190, 1199, Ciudad Autónoma de Buenos Aires, Argentina; Instituto Tecnológico de Buenos Aires, Iguazú 341, 1437, Ciudad Autónoma de Buenos Aires, Argentina.
| | - Martina Aineseder
- Radiology Department, Hospital Italiano de Buenos Aires, Tte. Gral. Juan Domingo Perón 4190, 1199, Ciudad Autónoma de Buenos Aires, Argentina.
| | - Roy López Grove
- Radiology Department, Hospital Italiano de Buenos Aires, Tte. Gral. Juan Domingo Perón 4190, 1199, Ciudad Autónoma de Buenos Aires, Argentina.
| | - Matías Alejandro Cerini
- Oncology Department, Hospital Italiano de Buenos Aires, Tte. Gral. Juan Domingo Perón 4190, 1199, Ciudad Autónoma de Buenos Aires, Argentina.
| | - María Alicia Verzura
- Oncology Department, Hospital Italiano de Buenos Aires, Tte. Gral. Juan Domingo Perón 4190, 1199, Ciudad Autónoma de Buenos Aires, Argentina.
| | - Daniel Roberto Luna
- Health Informatics Department, Hospital Italiano de Buenos Aires, Tte. Gral. Juan Domingo Perón 4190, 1199, Ciudad Autónoma de Buenos Aires, Argentina; Instituto de Medicina Traslacional e Ingeniería Biomédica (IMTIB), UE de triple dependencia CONICET- Instituto Universitario del Hospital Italiano (IUHI) - Hospital ITaliano (HIBA), Tte. Gral. Juan Domingo Perón 4190, 1199, Ciudad Autónoma de Buenos Aires, Argentina.
| | - Sonia Elizabeth Benítez
- Health Informatics Department, Hospital Italiano de Buenos Aires, Tte. Gral. Juan Domingo Perón 4190, 1199, Ciudad Autónoma de Buenos Aires, Argentina; Instituto Universitario del Hospital Italiano, Potosí 4265, 1199, Ciudad Autónoma de Buenos Aires, Argentina.
| | - Juan Carlos Spina
- Radiology Department, Hospital Italiano de Buenos Aires, Tte. Gral. Juan Domingo Perón 4190, 1199, Ciudad Autónoma de Buenos Aires, Argentina.
| |
Collapse
|
32
|
Granata V, Fusco R, De Muzio F, Brunese MC, Setola SV, Ottaiano A, Cardone C, Avallone A, Patrone R, Pradella S, Miele V, Tatangelo F, Cutolo C, Maggialetti N, Caruso D, Izzo F, Petrillo A. Radiomics and machine learning analysis by computed tomography and magnetic resonance imaging in colorectal liver metastases prognostic assessment. LA RADIOLOGIA MEDICA 2023; 128:1310-1332. [PMID: 37697033 DOI: 10.1007/s11547-023-01710-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/18/2023] [Accepted: 08/22/2023] [Indexed: 09/13/2023]
Abstract
OBJECTIVE The aim of this study was the evaluation radiomics analysis efficacy performed using computed tomography (CT) and magnetic resonance imaging in the prediction of colorectal liver metastases patterns linked to patient prognosis: tumor growth front; grade; tumor budding; mucinous type. Moreover, the prediction of liver recurrence was also evaluated. METHODS The retrospective study included an internal and validation dataset; the first was composed by 119 liver metastases from 49 patients while the second consisted to 28 patients with single lesion. Radiomic features were extracted using PyRadiomics. Univariate and multivariate approaches including machine learning algorithms were employed. RESULTS The best predictor to identify tumor growth was the Wavelet_HLH_glcm_MaximumProbability with an accuracy of 84% and to detect recurrence the best predictor was wavelet_HLH_ngtdm_Complexity with an accuracy of 90%, both extracted by T1-weigthed arterial phase sequence. The best predictor to detect tumor budding was the wavelet_LLH_glcm_Imc1 with an accuracy of 88% and to identify mucinous type was wavelet_LLH_glcm_JointEntropy with an accuracy of 92%, both calculated on T2-weigthed sequence. An increase statistically significant of accuracy (90%) was obtained using a linear weighted combination of 15 predictors extracted by T2-weigthed images to detect tumor front growth. An increase statistically significant of accuracy at 93% was obtained using a linear weighted combination of 11 predictors by the T1-weigthed arterial phase sequence to classify tumor budding. An increase statistically significant of accuracy at 97% was obtained using a linear weighted combination of 16 predictors extracted on CT to detect recurrence. An increase statistically significant of accuracy was obtained in the tumor budding identification considering a K-nearest neighbors and the 11 significant features extracted T1-weigthed arterial phase sequence. CONCLUSIONS The results confirmed the Radiomics capacity to recognize clinical and histopathological prognostic features that should influence the choice of treatments in colorectal liver metastases patients to obtain a more personalized therapy.
Collapse
Affiliation(s)
- Vincenza Granata
- Division of Radiology, Istituto Nazionale Tumori IRCCS Fondazione Pascale - IRCCS di Napoli, Naples, Italy.
| | | | - Federica De Muzio
- Department of Medicine and Health Sciences V. Tiberio, University of Molise, 86100, Campobasso, Italy
| | - Maria Chiara Brunese
- Department of Medicine and Health Sciences V. Tiberio, University of Molise, 86100, Campobasso, Italy
| | - Sergio Venanzio Setola
- Division of Radiology, Istituto Nazionale Tumori IRCCS Fondazione Pascale - IRCCS di Napoli, Naples, Italy
| | - Alessandro Ottaiano
- Clinical Experimental Abdominal Oncology Unit, Istituto Nazionale Tumori, IRCCS Fondazione G. Pascale, 80131, Naples, Italy
| | - Claudia Cardone
- Clinical Experimental Abdominal Oncology Unit, Istituto Nazionale Tumori, IRCCS Fondazione G. Pascale, 80131, Naples, Italy
| | - Antonio Avallone
- Clinical Experimental Abdominal Oncology Unit, Istituto Nazionale Tumori, IRCCS Fondazione G. Pascale, 80131, Naples, Italy
| | - Renato Patrone
- Division of Hepatobiliary Surgical Oncology, Istituto Nazionale Tumori IRCCS Fondazione Pascale-IRCCS di Napoli, 80131, Naples, Italy
| | - Silvia Pradella
- Department of Radiology, Careggi University Hospital, Largo Brambilla 3, 50134, Florence, Italy
- SIRM Foundation, Italian Society of Medical and Interventional Radiology (SIRM), 20122, Milan, Italy
| | - Vittorio Miele
- Department of Radiology, Careggi University Hospital, Largo Brambilla 3, 50134, Florence, Italy
- SIRM Foundation, Italian Society of Medical and Interventional Radiology (SIRM), 20122, Milan, Italy
| | - Fabiana Tatangelo
- Division of Pathological Anatomy and Cytopathology, Istituto Nazionale Tumori IRCCS Fondazione Pascale-IRCCS di Napoli, 80131, Naples, Italy
| | - Carmen Cutolo
- Department of Medicine, Surgery and Dentistry, University of Salerno, 84084, Salerno, Italy
| | - Nicola Maggialetti
- Department of Medical Science, Neuroscience and Sensory Organs (DSMBNOS), University of Bari "Aldo Moro", 70124, Bari, Italy
| | - Damiano Caruso
- Department of Medical Surgical Sciences and Translational Medicine, Radiology Unit-Sant'Andrea University Hospital, Sapienza-University of Rome, 00189, Rome, Italy
| | - Francesco Izzo
- Division of Hepatobiliary 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, Naples, Italy
| |
Collapse
|
33
|
Li M, Xu G, Cui Y, Wang M, Wang H, Xu X, Duan S, Shi J, Feng F. CT-based radiomics nomogram for the preoperative prediction of microsatellite instability and clinical outcomes in colorectal cancer: a multicentre study. Clin Radiol 2023; 78:e741-e751. [PMID: 37487841 DOI: 10.1016/j.crad.2023.06.012] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 06/15/2023] [Accepted: 06/29/2023] [Indexed: 07/26/2023]
Abstract
AIM To develop and validate a computed tomography (CT)-based radiomics nomogram for preoperative prediction of microsatellite instability (MSI) status and clinical outcomes in colorectal cancer (CRC) patients. MATERIALS AND METHODS This retrospective study enrolled 497 CRC patients from three centres. Least absolute shrinkage and selection operator regression was utilised for feature selection and constructing the radiomics signature. Univariate and multivariate logistic regression analyses were employed to identify significant clinical variables. The radiomics nomogram was constructed by integrating the radiomics signature and the identified clinical variables. The performance of the nomogram was evaluated through receiver operating characteristic curves, calibration curves, and decision curve analysis. Kaplan-Meier analysis was performed to investigate the prognostic value of the nomogram. RESULTS The radiomics signature comprised 10 radiomics features associated with MSI status. The nomogram, integrating the radiomics signature and independent predictors (age, location, and thickness), demonstrated favourable calibration and discrimination, achieving areas under the receiver operating characteristic (ROC) curves (AUCs) of 0.89 (95% confidence interval [CI]: 0.83-0.95), 0.87 (95% CI: 0.79-0.95), 0.88 (95% CI: 0.81-0.96), and 0.86 (95% CI: 0.78-0.93) in the training cohort, internal validation cohort, and two external validation cohorts, respectively. The nomogram exhibited superior performance compared to the clinical model (p<0.05). Additionally, survival analysis demonstrated that the nomogram successfully stratified stage II CRC patients based on prognosis (hazard ratio [HR]: 0.357, p=0.022). CONCLUSION The radiomics nomogram demonstrated promising performance in predicting MSI status and stratifying the prognosis of patients with CRC.
Collapse
Affiliation(s)
- M Li
- Department of Radiology, Affiliated Tumour Hospital of Nantong University, Nantong 226001, Jiangsu Province, China; Department of Radiology, Yancheng No. 1 People's Hospital, Yancheng 224006, Jiangsu Province, China
| | - G Xu
- Department of Radiology, Yancheng No. 1 People's Hospital, Yancheng 224006, Jiangsu Province, China; Department of Radiology, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, China
| | - Y Cui
- Department of Radiology, Shanxi Cancer Hospital, Shanxi 030013, Shanxi Province, China
| | - M Wang
- Department of Radiology, Yancheng No. 1 People's Hospital, Yancheng 224006, Jiangsu Province, China
| | - H Wang
- Department of Radiology, Affiliated Tumour Hospital of Nantong University, Nantong 226001, Jiangsu Province, China
| | - X Xu
- Department of Radiotherapy, Affiliated Tumour Hospital of Nantong University, Nantong 226001, Jiangsu Province, China
| | - S Duan
- GE Healthcare China, Shanghai 210000, China
| | - J Shi
- Department of Radiology, Affiliated Tumour Hospital of Nantong University, Nantong 226001, Jiangsu Province, China.
| | - F Feng
- Department of Radiology, Affiliated Tumour Hospital of Nantong University, Nantong 226001, Jiangsu Province, China.
| |
Collapse
|
34
|
Shamsabadipour A, Pourmadadi M, Davodabadi F, Rahdar A, Romanholo Ferreira LF. Applying thermodynamics as an applicable approach to cancer diagnosis, evaluation, and therapy: A review. J Drug Deliv Sci Technol 2023; 86:104681. [DOI: 10.1016/j.jddst.2023.104681] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
35
|
Xu B, Dong SY, Bai XL, Song TQ, Zhang BH, Zhou LD, Chen YJ, Zeng ZM, Wang K, Zhao HT, Lu N, Zhang W, Li XB, Zheng SS, Long G, Yang YC, Huang HS, Huang LQ, Wang YC, Liang F, Zhu XD, Huang C, Shen YH, Zhou J, Zeng MS, Fan J, Rao SX, Sun HC. Tumor Radiomic Features on Pretreatment MRI to Predict Response to Lenvatinib plus an Anti-PD-1 Antibody in Advanced Hepatocellular Carcinoma: A Multicenter Study. Liver Cancer 2023; 12:262-276. [PMID: 37601982 PMCID: PMC10433098 DOI: 10.1159/000528034] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Accepted: 10/02/2022] [Indexed: 08/22/2023] Open
Abstract
Introduction Lenvatinib plus an anti-PD-1 antibody has shown promising antitumor effects in patients with advanced hepatocellular carcinoma (HCC), but with clinical benefit limited to a subset of patients. We developed and validated a radiomic-based model to predict objective response to this combination therapy in advanced HCC patients. Methods Patients (N = 170) who received first-line combination therapy with lenvatinib plus an anti-PD-1 antibody were retrospectively enrolled from 9 Chinese centers; 124 and 46 into the training and validation cohorts, respectively. Radiomic features were extracted from pretreatment contrast-enhanced MRI. After feature selection, clinicopathologic, radiomic, and clinicopathologic-radiomic models were built using a neural network. The performance of models, incremental predictive value of radiomic features compared with clinicopathologic features and relationship between radiomic features and survivals were assessed. Results The clinicopathologic model modestly predicted objective response with an AUC of 0.748 (95% CI: 0.656-0.840) and 0.702 (95% CI: 0.547-0.884) in the training and validation cohorts, respectively. The radiomic model predicted response with an AUC of 0.886 (95% CI: 0.815-0.957) and 0.820 (95% CI: 0.648-0.984), respectively, with good calibration and clinical utility. The incremental predictive value of radiomic features to clinicopathologic features was confirmed with a net reclassification index of 47.9% (p < 0.001) and 41.5% (p = 0.025) in the training and validation cohorts, respectively. Furthermore, radiomic features were associated with overall survival and progression-free survival both in the training and validation cohorts, but modified albumin-bilirubin grade and neutrophil-to-lymphocyte ratio were not. Conclusion Radiomic features extracted from pretreatment MRI can predict individualized objective response to combination therapy with lenvatinib plus an anti-PD-1 antibody in patients with unresectable or advanced HCC, provide incremental predictive value over clinicopathologic features, and are associated with overall survival and progression-free survival after initiation of this combination regimen.
Collapse
Affiliation(s)
- Bin Xu
- Department of Liver Surgery and Transplantation, Liver Cancer Institute and Zhongshan Hospital, Fudan University, Shanghai, China
| | - San-Yuan Dong
- Department of Radiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Medical Imaging, Shanghai, China
| | - Xue-Li Bai
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Tian-Qiang Song
- Department of Hepatobiliary, National Clinical Research Center of Cancer, Oncology Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Bo-Heng Zhang
- Department of Hepatic Oncology, Xiamen Branch, Zhongshan Hospital, Fudan University, Xiamen, China
| | - Le-Du Zhou
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha, China
| | - Yong-Jun Chen
- Department of Hepatobiliary Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhi-Ming Zeng
- Department of Medical Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Kui Wang
- Department of Hepatic Surgery II, Eastern Hepatobiliary Surgery Hospital, Navy Medical University, Shanghai, China
| | - Hai-Tao Zhao
- Department of Liver Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Na Lu
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Wei Zhang
- Department of Hepatobiliary, National Clinical Research Center of Cancer, Oncology Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Xu-Bin Li
- Department of Radiology, National Clinical Research Center of Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Su-Su Zheng
- Department of Hepatic Oncology, Xiamen Branch, Zhongshan Hospital, Fudan University, Xiamen, China
| | - Guo Long
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha, China
| | - Yu-Chen Yang
- Department of Hepatobiliary Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hua-Sheng Huang
- Department of Medical Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Lan-Qing Huang
- Department of Hepatic Surgery II, Eastern Hepatobiliary Surgery Hospital, Navy Medical University, Shanghai, China
| | - Yun-Chao Wang
- Department of Liver Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Fei Liang
- Department of Biostatistics, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Xiao-Dong Zhu
- Department of Liver Surgery and Transplantation, Liver Cancer Institute and Zhongshan Hospital, Fudan University, Shanghai, China
| | - Cheng Huang
- Department of Liver Surgery and Transplantation, Liver Cancer Institute and Zhongshan Hospital, Fudan University, Shanghai, China
| | - Ying-Hao Shen
- Department of Liver Surgery and Transplantation, Liver Cancer Institute and Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jian Zhou
- Department of Liver Surgery and Transplantation, Liver Cancer Institute and Zhongshan Hospital, Fudan University, Shanghai, China
| | - Meng-Su Zeng
- Department of Radiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Medical Imaging, Shanghai, China
| | - Jia Fan
- Department of Liver Surgery and Transplantation, Liver Cancer Institute and Zhongshan Hospital, Fudan University, Shanghai, China
| | - Sheng-Xiang Rao
- Department of Radiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Medical Imaging, Shanghai, China
| | - Hui-Chuan Sun
- Department of Liver Surgery and Transplantation, Liver Cancer Institute and Zhongshan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
36
|
Costa G, Cavinato L, Fiz F, Sollini M, Chiti A, Torzilli G, Ieva F, Viganò L. Mapping Tumor Heterogeneity via Local Entropy Assessment: Making Biomarkers Visible. J Digit Imaging 2023; 36:1038-1048. [PMID: 36849835 PMCID: PMC10287605 DOI: 10.1007/s10278-023-00799-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 02/20/2023] [Accepted: 02/21/2023] [Indexed: 03/01/2023] Open
Abstract
Advanced imaging and analysis improve prediction of pathology data and outcomes in several tumors, with entropy-based measures being among the most promising biomarkers. However, entropy is often perceived as statistical data lacking clinical significance. We aimed to generate a voxel-by-voxel visual map of local tumor entropy, thus allowing to (1) make entropy explainable and accessible to clinicians; (2) disclose and quantitively characterize any intra-tumoral entropy heterogeneity; (3) evaluate associations between entropy and pathology data. We analyzed the portal phase of preoperative CT of 20 patients undergoing liver surgery for colorectal metastases. A three-dimensional core kernel (5 × 5 × 5 voxels) was created and used to compute the local entropy value for each voxel of the tumor. The map was encoded with a color palette. We performed two analyses: (a) qualitative assessment of tumors' detectability and pattern of entropy distribution; (b) quantitative analysis of the entropy values distribution. The latter data were compared with standard Hounsfield data as predictors of post-chemotherapy tumor regression grade (TRG). Entropy maps were successfully built for all tumors. Metastases were qualitatively hyper-entropic compared to surrounding parenchyma. In four cases hyper-entropic areas exceeded the tumor margin visible at CT. We identified four "entropic" patterns: homogeneous, inhomogeneous, peripheral rim, and mixed. At quantitative analysis, entropy-derived data (percentiles/mean/median/root mean square) predicted TRG (p < 0.05) better than Hounsfield-derived ones (p = n.s.). We present a standardized imaging technique to visualize tumor heterogeneity built on a voxel-by-voxel entropy assessment. The association of local entropy with pathology data supports its role as a biomarker.
Collapse
Affiliation(s)
- Guido Costa
- Division of Hepatobiliary and General Surgery, Department of Surgery, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy
| | - Lara Cavinato
- MOX Laboratory, Department of Mathematics, Politecnico di Milano, Piazza Leonardo da Vinci 32, 20133, Milan, Italy
| | - Francesco Fiz
- Department of Nuclear Medicine, IRCCS Humanitas Research Hospital, Milan, Italy
| | - Martina Sollini
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy
- Department of Nuclear Medicine, IRCCS Humanitas Research Hospital, Milan, Italy
| | - Arturo Chiti
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy
- Department of Nuclear Medicine, IRCCS Humanitas Research Hospital, Milan, Italy
| | - Guido Torzilli
- Division of Hepatobiliary and General Surgery, Department of Surgery, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy
| | - Francesca Ieva
- MOX Laboratory, Department of Mathematics, Politecnico di Milano, Piazza Leonardo da Vinci 32, 20133, Milan, Italy.
- CHDS - Center for Health Data Science, Human Technopole, Milan, Italy.
| | - Luca Viganò
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy.
- Hepatobiliary Unit, Department of Minimally Invasive General & Oncologic Surgery, Humanitas Gavazzeni University Hospital, Via M. Gavazzeni 21, 24125, Bergamo, Italy.
| |
Collapse
|
37
|
Inchingolo R, Maino C, Cannella R, Vernuccio F, Cortese F, Dezio M, Pisani AR, Giandola T, Gatti M, Giannini V, Ippolito D, Faletti R. Radiomics in colorectal cancer patients. World J Gastroenterol 2023; 29:2888-2904. [PMID: 37274803 PMCID: PMC10237092 DOI: 10.3748/wjg.v29.i19.2888] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Revised: 04/07/2023] [Accepted: 04/25/2023] [Indexed: 05/16/2023] Open
Abstract
The main therapeutic options for colorectal cancer are surgical resection and adjuvant chemotherapy in non-metastatic disease. However, the evaluation of the overall adjuvant chemotherapy benefit in patients with a high risk of recurrence is challenging. Radiological images can represent a source of data that can be analyzed by using automated computer-based techniques, working on numerical information coded within Digital Imaging and Communications in Medicine files: This image numerical analysis has been named "radiomics". Radiomics allows the extraction of quantitative features from radiological images, mainly invisible to the naked eye, that can be further analyzed by artificial intelligence algorithms. Radiomics is expanding in oncology to either understand tumor biology or for the development of imaging biomarkers for diagnosis, staging, and prognosis, prediction of treatment response and diseases monitoring and surveillance. Several efforts have been made to develop radiomics signatures for colorectal cancer patient using computed tomography (CT) images with different aims: The preoperative prediction of lymph node metastasis, detecting BRAF and RAS gene mutations. Moreover, the use of delta-radiomics allows the analysis of variations of the radiomics parameters extracted from CT scans performed at different timepoints. Most published studies concerning radiomics and magnetic resonance imaging (MRI) mainly focused on the response of advanced tumors that underwent neoadjuvant therapy. Nodes status is the main determinant of adjuvant chemotherapy. Therefore, several radiomics model based on MRI, especially on T2-weighted images and ADC maps, for the preoperative prediction of nodes metastasis in rectal cancer has been developed. Current studies mostly focused on the applications of radiomics in positron emission tomography/CT for the prediction of survival after curative surgical resection and assessment of response following neoadjuvant chemoradiotherapy. Since colorectal liver metastases develop in about 25% of patients with colorectal carcinoma, the main diagnostic tasks of radiomics should be the detection of synchronous and metachronous lesions. Radiomics could be an additional tool in clinical setting, especially in identifying patients with high-risk disease. Nevertheless, radiomics has numerous shortcomings that make daily use extremely difficult. Further studies are needed to assess performance of radiomics in stratifying patients with high-risk disease.
Collapse
Affiliation(s)
- Riccardo Inchingolo
- Unit of Interventional Radiology, F. Miulli Hospital, Acquaviva delle Fonti 70021, Italy
| | - Cesare Maino
- Department of Radiology, Fondazione IRCCS San Gerardo dei Tintori, Monza 20900, Italy
| | - Roberto Cannella
- Department of Biomedicine, Neuroscience and Advanced Diagnostics (BiND), University of Palermo, Palermo 90127, Italy
| | - Federica Vernuccio
- Institute of Radiology, University Hospital of Padova, Padova 35128, Italy
| | - Francesco Cortese
- Unit of Interventional Radiology, F. Miulli Hospital, Acquaviva delle Fonti 70021, Italy
| | - Michele Dezio
- Unit of Interventional Radiology, F. Miulli Hospital, Acquaviva delle Fonti 70021, Italy
| | - Antonio Rosario Pisani
- Interdisciplinary Department of Medicine, Section of Nuclear Medicine, University of Bari “Aldo Moro”, Bari 70121, Italy
| | - Teresa Giandola
- Department of Radiology, Fondazione IRCCS San Gerardo dei Tintori, Monza 20900, Italy
| | - Marco Gatti
- Department of Surgical Sciences, University of Turin, Turin 10126, Italy
| | - Valentina Giannini
- Department of Surgical Sciences, University of Turin, Turin 10126, Italy
| | - Davide Ippolito
- Department of Radiology, Fondazione IRCCS San Gerardo dei Tintori, Monza 20900, Italy
| | - Riccardo Faletti
- Department of Surgical Sciences, University of Turin, Turin 10126, Italy
| |
Collapse
|
38
|
Granata V, Fusco R, Setola SV, Galdiero R, Maggialetti N, Patrone R, Ottaiano A, Nasti G, Silvestro L, Cassata A, Grassi F, Avallone A, Izzo F, Petrillo A. Colorectal liver metastases patients prognostic assessment: prospects and limits of radiomics and radiogenomics. Infect Agent Cancer 2023; 18:18. [PMID: 36927442 PMCID: PMC10018963 DOI: 10.1186/s13027-023-00495-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Accepted: 03/07/2023] [Indexed: 03/18/2023] Open
Abstract
In this narrative review, we reported un up-to-date on the role of radiomics to assess prognostic features, which can impact on the liver metastases patient treatment choice. In the liver metastases patients, the possibility to assess mutational status (RAS or MSI), the tumor growth pattern and the histological subtype (NOS or mucinous) allows a better treatment selection to avoid unnecessary therapies. However, today, the detection of these features require an invasive approach. Recently, radiomics analysis application has improved rapidly, with a consequent growing interest in the oncological field. Radiomics analysis allows the textural characteristics assessment, which are correlated to biological data. This approach is captivating since it should allow to extract biological data from the radiological images, without invasive approach, so that to reduce costs and time, avoiding any risk for the patients. Several studies showed the ability of Radiomics to identify mutational status, tumor growth pattern and histological type in colorectal liver metastases. Although, radiomics analysis in a non-invasive and repeatable way, however features as the poor standardization and generalization of clinical studies results limit the translation of this analysis into clinical practice. Clear limits are data-quality control, reproducibility, repeatability, generalizability of results, and issues related to model overfitting.
Collapse
Affiliation(s)
- Vincenza Granata
- Division of Radiology, "Istituto Nazionale Tumori IRCCS Fondazione Pascale - IRCCS di Napoli", Naples, Italy.
| | - Roberta Fusco
- Medical Oncology Division, Igea SpA, Napoli, Italy.,Italian Society of Medical and Interventional Radiology (SIRM), SIRM Foundation, Via della Signora 2, Milan, 20122, Italy
| | - Sergio Venanzio Setola
- Division of Radiology, "Istituto Nazionale Tumori IRCCS Fondazione Pascale - IRCCS di Napoli", Naples, Italy
| | - Roberta Galdiero
- Division of Radiology, "Istituto Nazionale Tumori IRCCS Fondazione Pascale - IRCCS di Napoli", Naples, Italy
| | - Nicola Maggialetti
- Department of Medical Science, Neuroscience and Sensory Organs (DSMBNOS), University of Bari "Aldo Moro", Bari, 70124, Italy
| | - Renato Patrone
- Division of Epatobiliary Surgical Oncology, Istituto Nazionale Tumori IRCCS Fondazione Pascale-IRCCS di Napoli, Naples, 80131, Italy
| | - Alessandro Ottaiano
- Clinical Sperimental Abdominal Oncology Unit, Istituto Nazionale Tumori, IRCCS Fondazione G. Pascale, Napoli, 80131, Italy
| | - Guglielmo Nasti
- Clinical Sperimental Abdominal Oncology Unit, Istituto Nazionale Tumori, IRCCS Fondazione G. Pascale, Napoli, 80131, Italy
| | - Lucrezia Silvestro
- Clinical Sperimental Abdominal Oncology Unit, Istituto Nazionale Tumori, IRCCS Fondazione G. Pascale, Napoli, 80131, Italy
| | - Antonio Cassata
- Clinical Sperimental Abdominal Oncology Unit, Istituto Nazionale Tumori, IRCCS Fondazione G. Pascale, Napoli, 80131, Italy
| | - Francesca Grassi
- Division of Radiology, "Università degli Studi della Campania Luigi Vanvitelli", Naples, 80138, Italy
| | - Antonio Avallone
- Clinical Sperimental Abdominal Oncology Unit, Istituto Nazionale Tumori, IRCCS Fondazione G. Pascale, Napoli, 80131, Italy
| | - Francesco Izzo
- Division of Epatobiliary Surgical Oncology, Istituto Nazionale Tumori IRCCS Fondazione Pascale-IRCCS di Napoli, Naples, 80131, Italy
| | - Antonella Petrillo
- Division of Radiology, "Istituto Nazionale Tumori IRCCS Fondazione Pascale - IRCCS di Napoli", Naples, Italy
| |
Collapse
|
39
|
Predicting survival after radiosurgery in patients with lung cancer brain metastases using deep learning of radiomics and EGFR status. Phys Eng Sci Med 2023; 46:585-596. [PMID: 36857023 DOI: 10.1007/s13246-023-01234-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Accepted: 02/13/2023] [Indexed: 03/02/2023]
Abstract
The early prediction of overall survival (OS) in patients with lung cancer brain metastases (BMs) after Gamma Knife radiosurgery (GKRS) can facilitate patient management and outcome improvement. However, the disease progression is influenced by multiple factors, such as patient characteristics and treatment strategies, and hence satisfactory performance of OS prediction remains challenging. Accordingly, we proposed a deep learning approach based on comprehensive predictors, including clinical, imaging, and genetic information, to accomplish reliable and personalized OS prediction in patients with BMs after receiving GKRS. Overall 1793 radiomic features extracted from pre-GKRS magnetic resonance images (MRI), clinical information, and epidermal growth factor receptor (EGFR) mutation status were retrospectively collected from 237 BM patients who underwent GKRS. DeepSurv, a multi-layer perceptron model, with 4 different aggregation methods of radiomics was applied to predict personalized survival curves and survival status at 3, 6, 12, and 24 months. The model combining clinical features, EGFR status, and radiomics from the largest BM showed the best prediction performance with concordance index of 0.75 and achieved areas under the curve of 0.82, 0.80, 0.84, and 0.92 for predicting survival status at 3, 6, 12, and 24 months, respectively. The DeepSurv model showed a significant improvement (p < 0.001) in concordance index compared to the validated lung cancer BM prognostic molecular markers. Furthermore, the model provided a novel estimate of the risk-of-death period for patients. The personalized survival curves generated by the DeepSurv model effectively predicted the risk-of-death period which could facilitate personalized management of patients with lung cancer BMs.
Collapse
|
40
|
Huang M, Xu Q, Zhou M, Li X, Lv W, Zhou C, Wu R, Zhou Z, Chen X, Huang C, Lu G. Distinguishing multiple primary lung cancers from intrapulmonary metastasis using CT-based radiomics. Eur J Radiol 2023; 160:110671. [PMID: 36739831 DOI: 10.1016/j.ejrad.2022.110671] [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: 07/14/2022] [Revised: 11/29/2022] [Accepted: 12/22/2022] [Indexed: 12/27/2022]
Abstract
PURPOSE To develop CT-based radiomics models that can efficiently distinguish between multiple primary lung cancers (MPLCs) and intrapulmonary metastasis (IPMs). METHOD This retrospective study included 127 patients with 254 lung tumors pathologically proved as MPLCs or IPMs between May 2009 and January 2020. Radiomics features of lung tumors were extracted from baseline CT scans. Particularly, we incorporated tumor-focused, refined radiomics by calculating relative radiomics differences from paired tumors of individual patients. We applied the L1-norm regularization and analysis of variance to select informative radiomics features for constructing radiomics model (RM) and refined radiomics model (RRM). The performance was assessed by the area under the receiver operating characteristic curve (AUC-ROC). The two radiomics models were compared with the clinical-CT model (CCM, including clinical and CT semantic features). We incorporated both radiomics features to construct fusion model1 (FM1). We also, build fusion model2 (FM2) by combing both radiomics, clinical and CT semantic features. The performance of the FM1 and FM2 were further compared with that of the RRM. RESULTS On the validation set, the RM achieved an AUC of 0.857. The RRM demonstrated improved performance (validation set AUC, 0.870) than the RM, and showed significant differences compared with the CCM (validation set AUC, 0.782). Fusion models further led prediction performance (validation set AUC, FM1:0.885; FM2:0.889). There were no significant differences among the performance of the FM1, the FM2 and the RRM. CONCLUSIONS The CT-based radiomics models presented good performance on the discrimination between MPLCs and IPMs, demonstrating the potential for early diagnosis and treatment guidance for MPLCs and IPMs.
Collapse
Affiliation(s)
- Mei Huang
- Department of Medical Imaging, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Qinmei Xu
- Department of Medical Imaging, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing, China; Department of Radiology, Stanford University, School of Medicine, Stanford, United States
| | - Mu Zhou
- Department of Computer Science, Rutgers University, 110 Frelinghuysen Road, Piscataway, NJ 08854, United States
| | - Xinyu Li
- Department of Medical Imaging, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing, China; Department of Medical Imaging, Jinling Hospital, School of Medical Imaging, Nanjing Medical University, Nanjing, China
| | - Wenhui Lv
- Department of Medical Imaging, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Changsheng Zhou
- Department of Medical Imaging, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Ren Wu
- Department of Medical Imaging, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing, China; Department of Medical Imaging, Jinling Hospital, School of Medical Imaging, Nanjing Medical University, Nanjing, China
| | - Zhen Zhou
- Deepwise AI Lab, Deepwise Inc., Beijing, China
| | | | | | - Guangming Lu
- Department of Medical Imaging, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing, China.
| |
Collapse
|
41
|
Li Y, Gao X, Tang X, Lin S, Pang H. Research on automatic classification technology of kidney tumor and normal kidney tissue based on computed tomography radiomics. Front Oncol 2023; 13:1013085. [PMID: 36910615 PMCID: PMC9998940 DOI: 10.3389/fonc.2023.1013085] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2022] [Accepted: 02/13/2023] [Indexed: 03/14/2023] Open
Abstract
Purpose By using a radiomics-based approach, multiple radiomics features can be extracted from regions of interest in computed tomography (CT) images, which may be applied to automatically classify kidney tumors and normal kidney tissues. The study proposes a method based on CT radiomics and aims to use extracted radiomics features to automatically classify of kidney tumors and normal kidney tissues and to establish an automatic classification model. Methods CT data were retrieved from the 2019 Kidney and Kidney Tumor Segmentation Challenge (KiTS19) in The Cancer Imaging Archive (TCIA) open access database. Arterial phase-enhanced CT images from 210 cases were used to establish an automatic classification model. These CT images of patients were randomly divided into training (168 cases) and test (42 cases) sets. Furthermore, the radiomics features of gross tumor volume (GTV) and normal kidney tissues in the training set were extracted and screened, and a binary logistic regression model was established. For the test set, the radiomic features and cutoff value of P were consistent with the training set. Results Three radiomics features were selected to establish the binary logistic regression model. The accuracy (ACC), sensitivity (SENS), specificity (SPEC), area under the curve (AUC), and Youden index of the training and test sets based on the CT radiomics classification model were all higher than 0.85. Conclusion The automatic classification model of kidney tumors and normal kidney tissues based on CT radiomics exhibited good classification ability. Kidney tumors could be distinguished from normal kidney tissues. This study may complement automated tumor delineation techniques and warrants further research.
Collapse
Affiliation(s)
- Yunfei Li
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Xinrui Gao
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Xuemei Tang
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Sheng Lin
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Haowen Pang
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| |
Collapse
|
42
|
Feng Z, Li H, Liu Q, Duan J, Zhou W, Yu X, Chen Q, Liu Z, Wang W, Rong P. CT Radiomics to Predict Macrotrabecular-Massive Subtype and Immune Status in Hepatocellular Carcinoma. Radiology 2022; 307:e221291. [PMID: 36511807 DOI: 10.1148/radiol.221291] [Citation(s) in RCA: 54] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Background Macrotrabecular-massive (MTM) subtype of hepatocellular carcinoma (HCC) is an aggressive variant associated with angiogenesis and immunosuppressive tumor microenvironment, which is expected to be noninvasively identified using radiomics approaches. Purpose To construct a CT radiomics model to predict the MTM subtype and to investigate the underlying immune infiltration patterns. Materials and Methods This study included five retrospective data sets and one prospective data set from three academic medical centers between January 2015 and December 2021. The preoperative liver contrast-enhanced CT studies of 365 adult patients with resected HCC were evaluated. The Third Xiangya Hospital of Central South University provided the training set and internal test set, while Yueyang Central Hospital and Hunan Cancer Hospital provided the external test sets. Radiomic features were extracted and used to develop a radiomics model with machine learning in the training set, and the performance was verified in the two test sets. The outcomes cohort, including 58 adult patients with advanced HCC undergoing transarterial chemoembolization and antiangiogenic therapy, was used to evaluate the predictive value of the radiomics model for progression-free survival (PFS). Bulk RNA sequencing of tumors from 41 patients in The Cancer Genome Atlas (TCGA) and single-cell RNA sequencing from seven prospectively enrolled participants were used to investigate the radiomics-related immune infiltration patterns. Area under the receiver operating characteristics curve of the radiomics model was calculated, and Cox proportional regression was performed to identify predictors of PFS. Results Among 365 patients (mean age, 55 years ± 10 [SD]; 319 men) used for radiomics modeling, 122 (33%) were confirmed to have the MTM subtype. The radiomics model included 11 radiomic features and showed good performance for predicting the MTM subtype, with AUCs of 0.84, 0.80, and 0.74 in the training set, internal test set, and external test set, respectively. A low radiomics model score relative to the median value in the outcomes cohort was independently associated with PFS (hazard ratio, 0.4; 95% CI: 0.2, 0.8; P = .01). The radiomics model was associated with dysregulated humoral immunity involving B-cell infiltration and immunoglobulin synthesis. Conclusion Accurate prediction of the macrotrabecular-massive subtype in patients with hepatocellular carcinoma was achieved using a CT radiomics model, which was also associated with defective humoral immunity. Published under a CC BY 4.0 license. Supplemental material is available for this article. See also the editorial by Yoon and Kim in this issue.
Collapse
Affiliation(s)
- Zhichao Feng
- From the Departments of Radiology (Z.F., H.L., J.D., W.W., P.R.), Pathology (Q.C.), and Infectious Disease (Z.L.), The Third Xiangya Hospital of Central South University, No. 138 Tongzipo Rd, Changsha 410013, China; Department of Medical Imaging, Yueyang Central Hospital, Yueyang, China (Q.L., W.Z.); and Department of Diagnostic Radiology, Hunan Cancer Hospital, Changsha, China (X.Y.)
| | - Huiling Li
- From the Departments of Radiology (Z.F., H.L., J.D., W.W., P.R.), Pathology (Q.C.), and Infectious Disease (Z.L.), The Third Xiangya Hospital of Central South University, No. 138 Tongzipo Rd, Changsha 410013, China; Department of Medical Imaging, Yueyang Central Hospital, Yueyang, China (Q.L., W.Z.); and Department of Diagnostic Radiology, Hunan Cancer Hospital, Changsha, China (X.Y.)
| | - Qianyun Liu
- From the Departments of Radiology (Z.F., H.L., J.D., W.W., P.R.), Pathology (Q.C.), and Infectious Disease (Z.L.), The Third Xiangya Hospital of Central South University, No. 138 Tongzipo Rd, Changsha 410013, China; Department of Medical Imaging, Yueyang Central Hospital, Yueyang, China (Q.L., W.Z.); and Department of Diagnostic Radiology, Hunan Cancer Hospital, Changsha, China (X.Y.)
| | - Junhong Duan
- From the Departments of Radiology (Z.F., H.L., J.D., W.W., P.R.), Pathology (Q.C.), and Infectious Disease (Z.L.), The Third Xiangya Hospital of Central South University, No. 138 Tongzipo Rd, Changsha 410013, China; Department of Medical Imaging, Yueyang Central Hospital, Yueyang, China (Q.L., W.Z.); and Department of Diagnostic Radiology, Hunan Cancer Hospital, Changsha, China (X.Y.)
| | - Wenming Zhou
- From the Departments of Radiology (Z.F., H.L., J.D., W.W., P.R.), Pathology (Q.C.), and Infectious Disease (Z.L.), The Third Xiangya Hospital of Central South University, No. 138 Tongzipo Rd, Changsha 410013, China; Department of Medical Imaging, Yueyang Central Hospital, Yueyang, China (Q.L., W.Z.); and Department of Diagnostic Radiology, Hunan Cancer Hospital, Changsha, China (X.Y.)
| | - Xiaoping Yu
- From the Departments of Radiology (Z.F., H.L., J.D., W.W., P.R.), Pathology (Q.C.), and Infectious Disease (Z.L.), The Third Xiangya Hospital of Central South University, No. 138 Tongzipo Rd, Changsha 410013, China; Department of Medical Imaging, Yueyang Central Hospital, Yueyang, China (Q.L., W.Z.); and Department of Diagnostic Radiology, Hunan Cancer Hospital, Changsha, China (X.Y.)
| | - Qian Chen
- From the Departments of Radiology (Z.F., H.L., J.D., W.W., P.R.), Pathology (Q.C.), and Infectious Disease (Z.L.), The Third Xiangya Hospital of Central South University, No. 138 Tongzipo Rd, Changsha 410013, China; Department of Medical Imaging, Yueyang Central Hospital, Yueyang, China (Q.L., W.Z.); and Department of Diagnostic Radiology, Hunan Cancer Hospital, Changsha, China (X.Y.)
| | - Zhenguo Liu
- From the Departments of Radiology (Z.F., H.L., J.D., W.W., P.R.), Pathology (Q.C.), and Infectious Disease (Z.L.), The Third Xiangya Hospital of Central South University, No. 138 Tongzipo Rd, Changsha 410013, China; Department of Medical Imaging, Yueyang Central Hospital, Yueyang, China (Q.L., W.Z.); and Department of Diagnostic Radiology, Hunan Cancer Hospital, Changsha, China (X.Y.)
| | - Wei Wang
- From the Departments of Radiology (Z.F., H.L., J.D., W.W., P.R.), Pathology (Q.C.), and Infectious Disease (Z.L.), The Third Xiangya Hospital of Central South University, No. 138 Tongzipo Rd, Changsha 410013, China; Department of Medical Imaging, Yueyang Central Hospital, Yueyang, China (Q.L., W.Z.); and Department of Diagnostic Radiology, Hunan Cancer Hospital, Changsha, China (X.Y.)
| | - Pengfei Rong
- From the Departments of Radiology (Z.F., H.L., J.D., W.W., P.R.), Pathology (Q.C.), and Infectious Disease (Z.L.), The Third Xiangya Hospital of Central South University, No. 138 Tongzipo Rd, Changsha 410013, China; Department of Medical Imaging, Yueyang Central Hospital, Yueyang, China (Q.L., W.Z.); and Department of Diagnostic Radiology, Hunan Cancer Hospital, Changsha, China (X.Y.)
| |
Collapse
|
43
|
Dercle L, Ammari S, Roblin E, Bigorgne A, Champiat S, Taihi L, Plaian A, Hans S, Lakiss S, Tselikas L, Rouanne M, Deutsch E, Schwartz LH, Gönen M, Flynn J, Massard C, Soria JC, Robert C, Marabelle A. High serum LDH and liver metastases are the dominant predictors of primary cancer resistance to anti-PD(L)1 immunotherapy. Eur J Cancer 2022; 177:80-93. [PMID: 36332438 DOI: 10.1016/j.ejca.2022.08.034] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Accepted: 08/27/2022] [Indexed: 01/06/2023]
Abstract
AIM Anti-PD-(L)1 immunotherapies improve survival in multiple cancers but remain ineffective for most patients. We applied machine-learning algorithms and multivariate analyses on baseline medical data to estimate their relative impact on overall survival (OS) upon anti-PD-(L)1 monotherapies. METHOD This prognostic/predictive study retrospectively analysed 33 baseline routine medical variables derived from computed tomography (CT) images, clinical and biological meta-data. 695 patients with a diagnosis of advanced cancer were treated in prospective clinical trials in a single tertiary cancer centre in 3 cohorts including systemic anti-PD-(L)1 (251, 235 patients) versus other systemic therapies (209 patients). A random forest model combined variables to identify the combination (signature) which best estimated OS in patients treated with immunotherapy. The performance for estimating OS [95%CI] was measured using Kaplan-Meier Analysis and Log-Rank test. RESULTS Elevated serum lactate dehydrogenase (LDHhi) and presence of liver metastases (LM+) were dominant and independent predictors of short OS in independent cohorts of melanoma and non-melanoma solid tumours. Overall, LDHhiLM+ patients treated with anti-PD-(L)1 monotherapy had a poorer outcome (median OS: 3.1[2.4-7.8] months]) compared to LDHlowLM-patients (median OS: 15.3[8.9-NA] months; P < 0.0001). The OS of LDHlowLM-patients treated with immunotherapy was 28.8[17.9-NA] months (vs 13.1[10.8-18.5], P = 0.02) in the overall population and 30.3[19.93-NA] months (vs 14.1[8.69-NA], P = 0.0013) in patients with melanoma. CONCLUSION LDHhiLM+ status identifies patients who shall not benefit from anti-PD-(L)1 monotherapy. It could be used in clinical trials to stratify patients and eventually address this specific medical need.
Collapse
Affiliation(s)
- Laurent Dercle
- INSERM U1015 & CIC1428, Gustave Roussy, 94805 Villejuif, France.
| | - Samy Ammari
- Département de Radiologie, Gustave Roussy, 94805 Villejuif, France; BIOMAPS. UMR1281 INSERM. CEA. CNRS.Université Paris-Saclay, Villejuif, France
| | - Elvire Roblin
- Service de Biostatistique et D'Épidémiologie, Gustave Roussy, Université Paris-Saclay, Villejuif, France; Oncostat U1018, INSERM, Université Paris-Saclay, Équipe Labellisée Ligue Contre le Cancer, Villejuif, France
| | - Amelie Bigorgne
- INSERM U1015 & CIC1428, Gustave Roussy, 94805 Villejuif, France; INSERM U1163, Institut Imagine, Paris, France
| | | | - Lokmane Taihi
- Département de Radiologie, Gustave Roussy, 94805 Villejuif, France
| | - Athèna Plaian
- Département de Radiologie, Gustave Roussy, 94805 Villejuif, France
| | - Sophie Hans
- Département de Radiologie, Gustave Roussy, 94805 Villejuif, France
| | - Sara Lakiss
- Département de Radiologie, Gustave Roussy, 94805 Villejuif, France
| | | | - Mathieu Rouanne
- Hôpital Foch, UVSQ-Université Paris-Saclay, Suresnes, France; Departement D'Innovation Thérapeutique et des Essais Précoces (DITEP), Gustave Roussy, Universite Paris Saclay, Villejuif, France
| | - Eric Deutsch
- Departement de Radiothérapie, Gustave Roussy, Université Paris Saclay, 94805 Villejuif, France
| | - Lawrence H Schwartz
- Department of Radiology, NewYork-Presbyterian, Columbia University Irving Medical Center, NY, USA
| | - Mithat Gönen
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Jessica Flynn
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Christophe Massard
- Departement D'Innovation Thérapeutique et des Essais Précoces (DITEP), Gustave Roussy, Universite Paris Saclay, Villejuif, France
| | - Jean-Charles Soria
- Departement D'Innovation Thérapeutique et des Essais Précoces (DITEP), Gustave Roussy, Universite Paris Saclay, Villejuif, France; INSERM U981, Gustave Roussy, Villejuif, France
| | - Caroline Robert
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Departement de Médecine Oncologique, Gustave Roussy, Université Paris Saclay, Villejuif, France
| | - Aurélien Marabelle
- INSERM U1015 & CIC1428, Gustave Roussy, 94805 Villejuif, France; Departement D'Innovation Thérapeutique et des Essais Précoces (DITEP), Gustave Roussy, Universite Paris Saclay, Villejuif, France.
| |
Collapse
|
44
|
Imaging standardisation in metastatic colorectal cancer: A joint EORTC-ESOI-ESGAR expert consensus recommendation. Eur J Cancer 2022; 176:193-206. [PMID: 36274570 DOI: 10.1016/j.ejca.2022.09.012] [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: 07/19/2022] [Revised: 09/13/2022] [Accepted: 09/14/2022] [Indexed: 12/15/2022]
Abstract
BACKGROUND Treatment monitoring in metastatic colorectal cancer (mCRC) relies on imaging to evaluate the tumour burden. Response Evaluation Criteria in Solid Tumors provide a framework on reporting and interpretation of imaging findings yet offer no guidance on a standardised imaging protocol tailored to patients with mCRC. Imaging protocol heterogeneity remains a challenge for the reproducibility of conventional imaging end-points and is an obstacle for research on novel imaging end-points. PATIENTS AND METHODS Acknowledging the recently highlighted potential of radiomics and artificial intelligence tools as decision support for patient care in mCRC, a multidisciplinary, international and expert panel of imaging specialists was formed to find consensus on mCRC imaging protocols using the Delphi method. RESULTS Under the guidance of the European Organisation for Research and Treatment of Cancer (EORTC) Imaging and Gastrointestinal Tract Cancer Groups, the European Society of Oncologic Imaging (ESOI) and the European Society of Gastrointestinal and Abdominal Radiology (ESGAR), the EORTC-ESOI-ESGAR core imaging protocol was identified. CONCLUSION This consensus protocol attempts to promote standardisation and to diminish variations in patient preparation, scan acquisition and scan reconstruction. We anticipate that this standardisation will increase reproducibility of radiomics and artificial intelligence studies and serve as a catalyst for future research on imaging end-points. For ongoing and future mCRC trials, we encourage principal investigators to support the dissemination of these imaging standards across recruiting centres.
Collapse
|
45
|
Alshohoumi F, Al-Hamdani A, Hedjam R, AlAbdulsalam A, Al Zaabi A. A Review of Radiomics in Predicting Therapeutic Response in Colorectal Liver Metastases: From Traditional to Artificial Intelligence Techniques. Healthcare (Basel) 2022; 10:2075. [PMID: 36292522 PMCID: PMC9602631 DOI: 10.3390/healthcare10102075] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 10/12/2022] [Accepted: 10/13/2022] [Indexed: 12/12/2024] Open
Abstract
An early evaluation of colorectal cancer liver metastasis (CRCLM) is crucial in determining treatment options that ultimately affect patient survival rates and outcomes. Radiomics (quantitative imaging features) have recently gained popularity in diagnostic and therapeutic strategies. Despite this, radiomics faces many challenges and limitations. This study sheds light on these limitations by reviewing the studies that used radiomics to predict therapeutic response in CRCLM. Despite radiomics' potential to enhance clinical decision-making, it lacks standardization. According to the results of this study, the instability of radiomics quantification is caused by changes in CT scan parameters used to obtain CT scans, lesion segmentation methods used for contouring liver metastases, feature extraction methods, and dataset size used for experimentation and validation. Accordingly, the study recommends combining radiomics with deep learning to improve prediction accuracy.
Collapse
Affiliation(s)
- Fatma Alshohoumi
- Department of Computer Science, College of Science, Sultan Qaboos University, P.O. Box 36, Muscat 123, Oman
| | - Abdullah Al-Hamdani
- Department of Computer Science, College of Science, Sultan Qaboos University, P.O. Box 36, Muscat 123, Oman
| | - Rachid Hedjam
- Department of Computer Science, College of Science, Sultan Qaboos University, P.O. Box 36, Muscat 123, Oman
| | - AbdulRahman AlAbdulsalam
- Department of Computer Science, College of Science, Sultan Qaboos University, P.O. Box 36, Muscat 123, Oman
| | - Adhari Al Zaabi
- Department of Human and Clinical Anatomy, College of Medicine & Health Sciences, Sultan Qaboos University, P.O. Box 36, Muscat 123, Oman
| |
Collapse
|
46
|
Budai BK, Stollmayer R, Rónaszéki AD, Körmendy B, Zsombor Z, Palotás L, Fejér B, Szendrõi A, Székely E, Maurovich-Horvat P, Kaposi PN. Radiomics analysis of contrast-enhanced CT scans can distinguish between clear cell and non-clear cell renal cell carcinoma in different imaging protocols. Front Med (Lausanne) 2022; 9:974485. [PMID: 36314024 PMCID: PMC9606401 DOI: 10.3389/fmed.2022.974485] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Accepted: 09/28/2022] [Indexed: 11/13/2022] Open
Abstract
Introduction This study aimed to construct a radiomics-based machine learning (ML) model for differentiation between non-clear cell and clear cell renal cell carcinomas (ccRCC) that is robust against institutional imaging protocols and scanners. Materials and methods Preoperative unenhanced (UN), corticomedullary (CM), and excretory (EX) phase CT scans from 209 patients diagnosed with RCCs were retrospectively collected. After the three-dimensional segmentation, 107 radiomics features (RFs) were extracted from the tumor volumes in each contrast phase. For the ML analysis, the cases were randomly split into training and test sets with a 3:1 ratio. Highly correlated RFs were filtered out based on Pearson’s correlation coefficient (r > 0.95). Intraclass correlation coefficient analysis was used to select RFs with excellent reproducibility (ICC ≥ 0.90). The most predictive RFs were selected by the least absolute shrinkage and selection operator (LASSO). A support vector machine algorithm-based binary classifier (SVC) was constructed to predict tumor types and its performance was evaluated based-on receiver operating characteristic curve (ROC) analysis. The “Kidney Tumor Segmentation 2019” (KiTS19) publicly available dataset was used during external validation of the model. The performance of the SVC was also compared with an expert radiologist’s. Results The training set consisted of 121 ccRCCs and 38 non-ccRCCs, while the independent internal test set contained 40 ccRCCs and 13 non-ccRCCs. For external validation, 50 ccRCCs and 23 non-ccRCCs were identified from the KiTS19 dataset with the available UN, CM, and EX phase CTs. After filtering out the highly correlated and poorly reproducible features, the LASSO algorithm selected 10 CM phase RFs that were then used for model construction. During external validation, the SVC achieved an area under the ROC curve (AUC) value, accuracy, sensitivity, and specificity of 0.83, 0.78, 0.80, and 0.74, respectively. UN and/or EX phase RFs did not further increase the model’s performance. Meanwhile, in the same comparison, the expert radiologist achieved similar performance with an AUC of 0.77, an accuracy of 0.79, a sensitivity of 0.84, and a specificity of 0.69. Conclusion Radiomics analysis of CM phase CT scans combined with ML can achieve comparable performance with an expert radiologist in differentiating ccRCCs from non-ccRCCs.
Collapse
Affiliation(s)
- Bettina Katalin Budai
- Department of Radiology, Faculty of Medicine, Medical Imaging Centre, Semmelweis University, Budapest, Hungary,*Correspondence: Bettina Katalin Budai,
| | - Róbert Stollmayer
- Department of Radiology, Faculty of Medicine, Medical Imaging Centre, Semmelweis University, Budapest, Hungary
| | - Aladár Dávid Rónaszéki
- Department of Radiology, Faculty of Medicine, Medical Imaging Centre, Semmelweis University, Budapest, Hungary
| | - Borbála Körmendy
- Department of Radiology, Faculty of Medicine, Medical Imaging Centre, Semmelweis University, Budapest, Hungary
| | - Zita Zsombor
- Department of Radiology, Faculty of Medicine, Medical Imaging Centre, Semmelweis University, Budapest, Hungary
| | - Lõrinc Palotás
- Department of Radiology, Faculty of Medicine, Medical Imaging Centre, Semmelweis University, Budapest, Hungary
| | - Bence Fejér
- Department of Radiology, Faculty of Medicine, Medical Imaging Centre, Semmelweis University, Budapest, Hungary
| | - Attila Szendrõi
- Department of Urology, Faculty of Medicine, Semmelweis University, Budapest, Hungary
| | - Eszter Székely
- Department of Pathology, Forensic and Insurance Medicine, Faculty of Medicine, Semmelweis University, Budapest, Hungary
| | - Pál Maurovich-Horvat
- Department of Radiology, Faculty of Medicine, Medical Imaging Centre, Semmelweis University, Budapest, Hungary
| | - Pál Novák Kaposi
- Department of Radiology, Faculty of Medicine, Medical Imaging Centre, Semmelweis University, Budapest, Hungary
| |
Collapse
|
47
|
Ibrahim A, Lu L, Yang H, Akin O, Schwartz LH, Zhao B. The Impact of Image Acquisition Parameters and ComBat Harmonization on the Predictive Performance of Radiomics: A Renal Cell Carcinoma Model. APPLIED SCIENCES (BASEL, SWITZERLAND) 2022; 12:9824. [PMID: 37091743 PMCID: PMC10121203 DOI: 10.3390/app12199824] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 04/25/2023]
Abstract
Radiomics, one of the potential methods for developing clinical biomarker, is one of the exponentially growing research fields. In addition to its potential, several limitations have been identified in this field, and most importantly the effects of variations in imaging parameters on radiomic features (RFs). In this study, we investigate the potential of RFs to predict overall survival in patients with clear cell renal cell carcinoma, as well as the impact of ComBat harmonization on the performance of RF models. We assessed the robustness of the results by performing the analyses a thousand times. Publicly available CT scans of 179 patients were retrospectively collected and analyzed. The scans were acquired using different imaging vendors and parameters in different medical centers. The performance was calculated by averaging the metrics over all runs. On average, the clinical model significantly outperformed the radiomic models. The use of ComBat harmonization, on average, did not significantly improve the performance of radiomic models. Hence, the variability in image acquisition and reconstruction parameters significantly affect the performance of radiomic models. The development of radiomic specific harmonization techniques remain a necessity for the advancement of the field.
Collapse
Affiliation(s)
- Abdalla Ibrahim
- Department of Radiology, Columbia University Irving Medical Center, New York, NY 10032, USA
- Correspondence:
| | - Lin Lu
- Department of Radiology, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Hao Yang
- Department of Radiology, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Oguz Akin
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Lawrence H. Schwartz
- Department of Radiology, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Binsheng Zhao
- Department of Radiology, Columbia University Irving Medical Center, New York, NY 10032, USA
| |
Collapse
|
48
|
Hewitt DB, Brown ZJ, Pawlik TM. The Role of Biomarkers in the Management of Colorectal Liver Metastases. Cancers (Basel) 2022; 14:cancers14194602. [PMID: 36230522 PMCID: PMC9559307 DOI: 10.3390/cancers14194602] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Accepted: 09/17/2022] [Indexed: 11/16/2022] Open
Abstract
Simple Summary Colorectal cancer remains one of the most significant sources of cancer-related morbidity and mortality worldwide. The liver is the most common site of metastatic spread. Multiple modalities exist to manage and potentially cure patients with metastatic colorectal cancer. However, reliable biomarkers to assist with clinical decision-making are limited. Recent advances in genomic sequencing technology have greatly expanded our knowledge of colorectal cancer carcinogenesis and significantly reduced the cost and timing of the investigation. In this article, we discuss the current utility of biomarkers in the management of colorectal cancer liver metastases. Abstract Surgical management combined with improved systemic therapies have extended 5-year overall survival beyond 50% among patients with colorectal liver metastases (CRLM). Furthermore, a multitude of liver-directed therapies has improved local disease control for patients with unresectable CRLM. Unfortunately, a significant portion of patients treated with curative-intent hepatectomy develops disease recurrence. Traditional markers fail to risk-stratify and prognosticate patients with CRLM appropriately. Over the last few decades, advances in molecular sequencing technology have greatly expanded our knowledge of the pathophysiology and tumor microenvironment characteristics of CRLM. These investigations have revealed biomarkers with the potential to better inform management decisions in patients with CRLM. Actionable biomarkers such as RAS and BRAF mutations, microsatellite instability/mismatch repair status, and tumor mutational burden have been incorporated into national and societal guidelines. Other biomarkers, including circulating tumor DNA and radiomic features, are under active investigation to evaluate their clinical utility. Given the plethora of therapeutic modalities and lack of evidence on timing and sequence, reliable biomarkers are needed to assist clinicians with the development of patient-tailored management plans. In this review, we discuss the current evidence regarding biomarkers for patients with CRLM.
Collapse
|
49
|
Chen Q, Zhang L, Liu S, You J, Chen L, Jin Z, Zhang S, Zhang B. Radiomics in precision medicine for gastric cancer: opportunities and challenges. Eur Radiol 2022; 32:5852-5868. [PMID: 35316364 DOI: 10.1007/s00330-022-08704-8] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2021] [Revised: 02/20/2022] [Accepted: 02/28/2022] [Indexed: 01/19/2023]
Abstract
OBJECTIVES Radiomic features derived from routine medical images show great potential for personalized medicine in gastric cancer (GC). We aimed to evaluate the current status and quality of radiomic research as well as its potential for identifying biomarkers to predict therapy response and prognosis in patients with GC. METHODS We performed a systematic search of the PubMed and Embase databases for articles published from inception through July 10, 2021. The phase classification criteria for image mining studies and the radiomics quality scoring (RQS) tool were applied to evaluate scientific and reporting quality. RESULTS Twenty-five studies consisting of 10,432 patients were included. 96% of studies extracted radiomic features from CT images. Association between radiomic signature and therapy response was evaluated in seven (28%) studies; association with survival was evaluated in 17 (68%) studies; one (4%) study analyzed both. All results of the included studies showed significant associations. Based on the phase classification criteria for image mining studies, 18 (72%) studies were classified as phase II, with two, four, and one studies as discovery science, phase 0 and phase I, respectively. The median RQS score for the radiomic studies was 44.4% (range, 0 to 55.6%). There was extensive heterogeneity in the study population, tumor stage, treatment protocol, and radiomic workflow amongst the studies. CONCLUSIONS Although radiomic research in GC is highly heterogeneous and of relatively low quality, it holds promise for predicting therapy response and prognosis. Efforts towards standardization and collaboration are needed to utilize radiomics for clinical application. KEY POINTS • Radiomics application of gastric cancer is increasingly being reported, particularly in predicting therapy response and survival. • Although radiomics research in gastric cancer is highly heterogeneous and relatively low quality, it holds promise for predicting clinical outcomes. • Standardized imaging protocols and radiomic workflow are needed to facilitate radiomics into clinical use.
Collapse
Affiliation(s)
- Qiuying Chen
- Department of Radiology, the First Affiliated Hospital, Jinan University, No.613, Huangpu West Road, Tianhe District, Guangzhou, 510627, Guangdong, China.,Graduate College, Jinan University, Guangzhou, Guangdong, China
| | - Lu Zhang
- Department of Radiology, the First Affiliated Hospital, Jinan University, No.613, Huangpu West Road, Tianhe District, Guangzhou, 510627, Guangdong, China.,Graduate College, Jinan University, Guangzhou, Guangdong, China
| | - Shuyi Liu
- Department of Radiology, the First Affiliated Hospital, Jinan University, No.613, Huangpu West Road, Tianhe District, Guangzhou, 510627, Guangdong, China.,Graduate College, Jinan University, Guangzhou, Guangdong, China
| | - Jingjing You
- Department of Radiology, the First Affiliated Hospital, Jinan University, No.613, Huangpu West Road, Tianhe District, Guangzhou, 510627, Guangdong, China.,Graduate College, Jinan University, Guangzhou, Guangdong, China
| | - Luyan Chen
- Department of Radiology, the First Affiliated Hospital, Jinan University, No.613, Huangpu West Road, Tianhe District, Guangzhou, 510627, Guangdong, China.,Graduate College, Jinan University, Guangzhou, Guangdong, China
| | - Zhe Jin
- Department of Radiology, the First Affiliated Hospital, Jinan University, No.613, Huangpu West Road, Tianhe District, Guangzhou, 510627, Guangdong, China.,Graduate College, Jinan University, Guangzhou, Guangdong, China
| | - Shuixing Zhang
- Department of Radiology, the First Affiliated Hospital, Jinan University, No.613, Huangpu West Road, Tianhe District, Guangzhou, 510627, Guangdong, China. .,Graduate College, Jinan University, Guangzhou, Guangdong, China.
| | - Bin Zhang
- Department of Radiology, the First Affiliated Hospital, Jinan University, No.613, Huangpu West Road, Tianhe District, Guangzhou, 510627, Guangdong, China. .,Graduate College, Jinan University, Guangzhou, Guangdong, China.
| |
Collapse
|
50
|
Russo V, Lallo E, Munnia A, Spedicato M, Messerini L, D’Aurizio R, Ceroni EG, Brunelli G, Galvano A, Russo A, Landini I, Nobili S, Ceppi M, Bruzzone M, Cianchi F, Staderini F, Roselli M, Riondino S, Ferroni P, Guadagni F, Mini E, Peluso M. Artificial Intelligence Predictive Models of Response to Cytotoxic Chemotherapy Alone or Combined to Targeted Therapy for Metastatic Colorectal Cancer Patients: A Systematic Review and Meta-Analysis. Cancers (Basel) 2022; 14:4012. [PMID: 36011003 PMCID: PMC9406544 DOI: 10.3390/cancers14164012] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 07/26/2022] [Accepted: 08/12/2022] [Indexed: 12/24/2022] Open
Abstract
Tailored treatments for metastatic colorectal cancer (mCRC) have not yet completely evolved due to the variety in response to drugs. Therefore, artificial intelligence has been recently used to develop prognostic and predictive models of treatment response (either activity/efficacy or toxicity) to aid in clinical decision making. In this systematic review, we have examined the ability of learning methods to predict response to chemotherapy alone or combined with targeted therapy in mCRC patients by targeting specific narrative publications in Medline up to April 2022 to identify appropriate original scientific articles. After the literature search, 26 original articles met inclusion and exclusion criteria and were included in the study. Our results show that all investigations conducted on this field have provided generally promising results in predicting the response to therapy or toxic side-effects. By a meta-analytic approach we found that the overall weighted means of the area under the receiver operating characteristic (ROC) curve (AUC) were 0.90, 95% C.I. 0.80-0.95 and 0.83, 95% C.I. 0.74-0.89 in training and validation sets, respectively, indicating a good classification performance in discriminating response vs. non-response. The calculation of overall HR indicates that learning models have strong ability to predict improved survival. Lastly, the delta-radiomics and the 74 gene signatures were able to discriminate response vs. non-response by correctly identifying up to 99% of mCRC patients who were responders and up to 100% of patients who were non-responders. Specifically, when we evaluated the predictive models with tests reaching 80% sensitivity (SE) and 90% specificity (SP), the delta radiomics showed an SE of 99% and an SP of 94% in the training set and an SE of 85% and SP of 92 in the test set, whereas for the 74 gene signatures the SE was 97.6% and the SP 100% in the training set.
Collapse
Affiliation(s)
- Valentina Russo
- Research and Development Branch, Regional Cancer Prevention Laboratory, ISPRO-Study, Prevention and Oncology Network Institute, 50139 Florence, Italy
| | - Eleonora Lallo
- Research and Development Branch, Regional Cancer Prevention Laboratory, ISPRO-Study, Prevention and Oncology Network Institute, 50139 Florence, Italy
| | - Armelle Munnia
- Research and Development Branch, Regional Cancer Prevention Laboratory, ISPRO-Study, Prevention and Oncology Network Institute, 50139 Florence, Italy
| | - Miriana Spedicato
- Research and Development Branch, Regional Cancer Prevention Laboratory, ISPRO-Study, Prevention and Oncology Network Institute, 50139 Florence, Italy
| | - Luca Messerini
- Department of Experimental and Clinical Medicine, University of Florence, 50134 Florence, Italy
| | - Romina D’Aurizio
- Institute of Informatics and Telematics, National Research Council, 56124 Pisa, Italy
| | - Elia Giuseppe Ceroni
- Institute of Informatics and Telematics, National Research Council, 56124 Pisa, Italy
| | - Giulia Brunelli
- Institute of Informatics and Telematics, National Research Council, 56124 Pisa, Italy
| | - Antonio Galvano
- Department of Surgical, Oncological and Oral Sciences, University of Palermo, 90127 Palermo, Italy
| | - Antonio Russo
- Department of Surgical, Oncological and Oral Sciences, University of Palermo, 90127 Palermo, Italy
| | - Ida Landini
- Department of Health Sciences, University of Florence, 50139 Florence, Italy
| | - Stefania Nobili
- Department of Neurosciences, Imaging and Clinical Sciences, “G. D’Annunzio” Chieti-Pescara, 66100 Chieti, Italy
| | - Marcello Ceppi
- Clinical Epidemiology Unit, IRCCS-Ospedale Policlinico San Martino, 16131 Genova, Italy
| | - Marco Bruzzone
- Clinical Epidemiology Unit, IRCCS-Ospedale Policlinico San Martino, 16131 Genova, Italy
| | - Fabio Cianchi
- Department of Experimental and Clinical Medicine, University of Florence, 50134 Florence, Italy
| | - Fabio Staderini
- Department of Experimental and Clinical Medicine, University of Florence, 50134 Florence, Italy
| | - Mario Roselli
- Medical Oncology Unit, Department of Systems Medicine, Tor Vergata University, 00133 Rome, Italy
| | - Silvia Riondino
- Medical Oncology Unit, Department of Systems Medicine, Tor Vergata University, 00133 Rome, Italy
| | - Patrizia Ferroni
- BioBIM (InterInstitutional Multidisciplinary Biobank), IRCCS San Raffaele Roma, 00166 Rome, Italy
- Department of Human Sciences & Quality of Life Promotion, San Raffaele Roma Open University, 00166 Rome, Italy
| | - Fiorella Guadagni
- BioBIM (InterInstitutional Multidisciplinary Biobank), IRCCS San Raffaele Roma, 00166 Rome, Italy
- Department of Human Sciences & Quality of Life Promotion, San Raffaele Roma Open University, 00166 Rome, Italy
| | - Enrico Mini
- Department of Health Sciences, University of Florence, 50139 Florence, Italy
| | - Marco Peluso
- Research and Development Branch, Regional Cancer Prevention Laboratory, ISPRO-Study, Prevention and Oncology Network Institute, 50139 Florence, Italy
| |
Collapse
|