1
|
Luo Y, Li X, Sun J, Liu S, Zhong P, Liu H, Chen X, Fang J. Predicting higher-risk growth patterns in invasive lung adenocarcinoma with multiphase multidetector computed tomography and 18 F-fluorodeoxyglucose PET radiomics. Nucl Med Commun 2025; 46:171-179. [PMID: 39575614 DOI: 10.1097/mnm.0000000000001931] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2025]
Abstract
PURPOSE To develop a predictive model for identifying the higher-risk growth pattern of invasive lung adenocarcinoma using multiphase multidetector computed tomography (MDCT) and 18 F-fluorodeoxyglucose (FDG) PET radiomics. METHODS A total of 203 patients with confirmed invasive lung adenocarcinoma between January 2018 and December 2021 were enrolled and randomly divided into training ( n = 143) and testing sets ( n = 60). Patients were classified into two groups according to the predominant growth pattern (lower-risk group: lepidic/acinar; higher-risk group: papillary/solid/micropapillary). Preoperative multiphase MDCT and 18 F-FDG PET images were evaluated. The Artificial Intelligence Kit software was used to extract radiomic features. Five predictive models [arterial phase, venous phase, and plain scan (AVP), PET, AVP-PET, clinical, and radiomic-clinical (Rad-Clin) combined model] were developed. The models' performance was assessed using receiver-operating characteristic (ROC) curves and compared using the DeLong test. RESULTS Among the radiomics models (AVP, PET, and AVP-PET), the AVP-PET model [area under ROC curve (AUC) = 0.888] outperformed the PET model (AUC = 0.814; P = 0.015) in predicting the higher-risk growth patterns. The combined Rad-Clin model (AUC = 0.923), which integrates AVP-PET radiomics and five independent clinical predictors (gender, spiculation, long-axis diameter, maximum standardized uptake value, and average standardized uptake value), exhibited superior performance in predicting the higher-risk growth pattern compared with radiomic models ( P = 0.043, vs. AVP-PET; P = 0.016, vs. AVP; P = 0.002, vs. PET) or the clinical model alone (constructing based on five clinical predictors; AUC = 0.793; P < 0.001). CONCLUSION The combined Rad-Clin model can predict the higher-risk growth patterns of invasive adenocarcinoma (IAC). This approach could help determine individual therapeutic strategies for IAC patients by distinguishing predominant growth patterns with high risk.
Collapse
Affiliation(s)
- Yi Luo
- Department of Radiology
- Department of Nuclear Medicine, Daping Hospital, Army Medical University
| | - Xiaoguang Li
- Department of Radiology
- Chongqing Clinical Research Center for Imaging and Nuclear Medicine
| | - Jinju Sun
- Department of Nuclear Medicine, Daping Hospital, Army Medical University
| | | | - Peng Zhong
- Department of Pathology, Daping Hospital, Army Medical University, Chongqing
| | - Huan Liu
- Advanced Application Team, GE Healthcare, Shanghai, China
| | - Xiao Chen
- Department of Nuclear Medicine, Daping Hospital, Army Medical University
| | - Jingqin Fang
- Department of Radiology
- Chongqing Clinical Research Center for Imaging and Nuclear Medicine
| |
Collapse
|
2
|
Janzen I, Ho C, Melosky B, Ye Q, Li J, Wang G, Lam S, MacAulay C, Yuan R. Machine Learning and Computed Tomography Radiomics to Predict Disease Progression to Upfront Pembrolizumab Monotherapy in Advanced Non-Small-Cell Lung Cancer: A Pilot Study. Cancers (Basel) 2024; 17:58. [PMID: 39796687 PMCID: PMC11719007 DOI: 10.3390/cancers17010058] [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: 11/14/2024] [Revised: 12/20/2024] [Accepted: 12/23/2024] [Indexed: 01/13/2025] Open
Abstract
BACKGROUND/OBJECTIVES Pembrolizumab monotherapy is approved in Canada for first-line treatment of advanced NSCLC with PD-L1 ≥ 50% and no EGFR/ALK aberrations. However, approximately 55% of these patients do not respond to pembrolizumab, underscoring the need for the early intervention of non-responders to optimize treatment strategies. Distinguishing the 55% sub-cohort prior to treatment is a real-world dilemma. METHODS In this retrospective study, we analyzed two patient cohorts treated with pembrolizumab monotherapy (training set: n = 97; test set: n = 17). The treatment response was assessed using baseline and follow-up CT scans via RECIST 1.1 criteria. RESULTS A logistic regression model, incorporating pre-treatment CT radiomic features of lung tumors and clinical variables, achieved high predictive accuracy (AUC: 0.85 in training; 0.81 in testing, 95% CI: 0.63-0.99). Notably, radiomic features from the peritumoral region were found to be independent predictors, complementing the standard CT evaluations and other clinical characteristics. CONCLUSIONS This pragmatic model offers a valuable tool to guide first-line treatment decisions in NSCLC patients with high PD-L1 expression and has the potential to advance personalized oncology and improve timely disease management.
Collapse
Affiliation(s)
- Ian Janzen
- Integrative Oncology, BC Cancer Research Institute, 675 West 10th Avenue, Vancouver, BC V5Z Il3, Canada; (I.J.)
- Interdisciplinary Oncology Program, Faculty of Medicine, University of British Columbia, 2329 West Mall, Vancouver, BC V6T IZ4, Canada
| | - Cheryl Ho
- BC Cancer, Vancouver Center, 600 West 10th Avenue, Vancouver, BC V5Z 4E6, Canada
- Department of Medical Oncology, Faculty of Medicine, University of British Columbia, 2329 West Mall, Vancouver, BC V6T IZ4, Canada
| | - Barbara Melosky
- BC Cancer, Vancouver Center, 600 West 10th Avenue, Vancouver, BC V5Z 4E6, Canada
- Department of Medical Oncology, Faculty of Medicine, University of British Columbia, 2329 West Mall, Vancouver, BC V6T IZ4, Canada
| | - Qian Ye
- Department of Statistics, Faculty of Science, University of British Columbia, 2329 West Mall, Vancouver, BC V6T 1Z4, Canada
| | - Jessica Li
- Department of Radiology, Faculty of Medicine, University of British Columbia, 2329 West Mall, Vancouver, BC V6T IZ4, Canada
| | - Gang Wang
- BC Cancer, Vancouver Center, 600 West 10th Avenue, Vancouver, BC V5Z 4E6, Canada
- Department of Pathology, Faculty of Medicine, University of British Columbia, 2329 West Mall, Vancouver, BC V6T IZ4, Canada
| | - Stephen Lam
- Integrative Oncology, BC Cancer Research Institute, 675 West 10th Avenue, Vancouver, BC V5Z Il3, Canada; (I.J.)
- BC Cancer, Vancouver Center, 600 West 10th Avenue, Vancouver, BC V5Z 4E6, Canada
- Department of Respirology, Faculty of Medicine, University of British Columbia, 2329 West Mall, Vancouver, BC V6T IZ4, Canada
| | - Calum MacAulay
- Integrative Oncology, BC Cancer Research Institute, 675 West 10th Avenue, Vancouver, BC V5Z Il3, Canada; (I.J.)
- Department of Pathology, Faculty of Medicine, University of British Columbia, 2329 West Mall, Vancouver, BC V6T IZ4, Canada
| | - Ren Yuan
- BC Cancer, Vancouver Center, 600 West 10th Avenue, Vancouver, BC V5Z 4E6, Canada
- Department of Radiology, Faculty of Medicine, University of British Columbia, 2329 West Mall, Vancouver, BC V6T IZ4, Canada
| |
Collapse
|
3
|
Ouyang Z, Zhang G, He S, Huang Q, Zhang L, Duan X, Zhang X, Liu Y, Ke T, Yang J, Ai C, Lu Y, Liao C. CT and MRI bimodal radiomics for predicting EGFR status in NSCLC patients with brain metastases: A multicenter study. Eur J Radiol 2024; 183:111853. [PMID: 39647269 DOI: 10.1016/j.ejrad.2024.111853] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Revised: 11/01/2024] [Accepted: 11/25/2024] [Indexed: 12/10/2024]
Abstract
BACKGROUND Leveraging the radiomics information from non-small cell lung cancer (NSCLC) primary lesion and brain metastasis (BM) to develop and validate a bimodal radiomics nomogram that can accurately predict epidermal growth factor receptor (EGFR) status. METHODS A total of 309 NSCLC patients with BM from three independent centers were recruited. Among them, the patients of Center I were randomly allocated into the training and internal test cohorts in a 7:3 ratio. Meanwhile, the patients from Center Ⅱ and Center Ⅲ collectively constitute the external test cohort. All chest CT and brain MRI images of each patient were obtained for image registration and sequence combination within a single modality. After image preprocessing, 1037 radiomics features were extracted from each single sequence. Six machine learning algorithms were used to construct radiomics signatures for CT and MRI respectively. The best CT and MRI radiomics signatures were fitted to establish the bimodal radiomics nomogram for predicting the EGFR status. RESULTS The contrast-enhanced (CE) eXtreme gradient boosting (XG Boost) and T2-weighted imaging (T2WI) + T1-weighted contrast-enhanced imaging (T1CE) random forest models were chosen as the radiomics signature representing primary lesion and BM. Both models were found to be independent predictors of EGFR mutation. The bimodal radiomics nomogram, which incorporated CT radiomics signature and MRI radiomics signature, demonstrated a good calibration and discrimination in the internal test cohort [area under curve (AUC), 0.866; 95 % confidence intervals (CI), 0.778-0.950) and the external test cohort (AUC, 0.818; 95 % CI, 0.691-0.938). CONCLUSIONS Our CT and MRI bimodal radiomics nomogram could timely and accurately evaluate the likelihood of EGFR mutation in patients with limited access to necessary materials, thus making up for the shortcoming of plasma sequencing and promoting the advancement of precision medicine.
Collapse
Affiliation(s)
- Zhiqiang Ouyang
- Department of Radiology, Yan an Hospital of Kunming City (Yanan Hospital Affiliated to Kunming Medical University), 245 Renmin East Road, Kunming, Yunnan, China.
| | - Guodong Zhang
- Bidding and Procurement Office, Yunnan Cancer Hospital (The Third Affiliated Hospital of Kunming Medical University), 519 Kunzhou Road, Kunming, Yunnan, China; Department of Chemistry, University of California, 900 University Avenue, Riverside, CA, United States
| | - Shaonan He
- Department of Medical Imaging, The First People's Hospital of Yunnan Province (The Affiliated Hospital of Kunming University of Science and Technology), 157 Jinbi Road, Kunming, Yunnan, China
| | - Qiubo Huang
- Department of Thoracic Surgery, Yunnan Cancer Hospital (The Third Affiliated Hospital of Kunming Medical University), 519 Kunzhou Road, Kunming, Yunnan, China
| | - Liren Zhang
- Department of Thoracic Surgery, The Second Affiliated Hospital of Kunming Medical University, 374 Dianmian Avenue, Kunming, Yunnan, China
| | - Xirui Duan
- Department of Radiology, Yan an Hospital of Kunming City (Yanan Hospital Affiliated to Kunming Medical University), 245 Renmin East Road, Kunming, Yunnan, China
| | - Xuerong Zhang
- Department of Radiology, Yunnan Cancer Hospital (The Third Affiliated Hospital of Kunming Medical University), 519 Kunzhou Road, Kunming, Yunnan, China
| | - Yifan Liu
- Department of Radiology, Yunnan Cancer Hospital (The Third Affiliated Hospital of Kunming Medical University), 519 Kunzhou Road, Kunming, Yunnan, China
| | - Tengfei Ke
- Department of Radiology, Yunnan Cancer Hospital (The Third Affiliated Hospital of Kunming Medical University), 519 Kunzhou Road, Kunming, Yunnan, China
| | - Jun Yang
- Department of Radiology, Yunnan Cancer Hospital (The Third Affiliated Hospital of Kunming Medical University), 519 Kunzhou Road, Kunming, Yunnan, China
| | - Conghui Ai
- Department of Radiology, Yunnan Cancer Hospital (The Third Affiliated Hospital of Kunming Medical University), 519 Kunzhou Road, Kunming, Yunnan, China
| | - Yi Lu
- Department of Radiology, The First Affiliated Hospital of Kunming Medical University, 295 Xichang Road, Kunming, Yunnan, China.
| | - Chengde Liao
- Department of Radiology, Yan an Hospital of Kunming City (Yanan Hospital Affiliated to Kunming Medical University), 245 Renmin East Road, Kunming, Yunnan, China.
| |
Collapse
|
4
|
Yue Q, Zhang M, Jiang W, Gao L, Ye R, Hong J, Li Y. Prognostic value of FDX1, the cuprotosis key gene, and its prediction models across imaging modalities and histology. BMC Cancer 2024; 24:1381. [PMID: 39528953 PMCID: PMC11552402 DOI: 10.1186/s12885-024-13149-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2024] [Accepted: 11/05/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND Cuprotosis has been identified as a novel way of cell death. The key regulator ferredoxin 1 (FDX1) was explored via pan-cancer analysis, and its prediction models were proposed across seven malignancies and two imaging modalities. METHODS The prognostic value of FDX1 was explored via 1654 cases of 33 types of cancer in the Cancer Genome Atlas database. The MRI cohort of hepatocellular carcinoma in the First Affiliated Hospital of Fujian Medical University, and CT and MRI images from the Cancer Imaging Archive, REMBRANDT and Duke databases were exploited to formulate radiomic models to predict FDX1 expression. After segmentation of volumes of interest and feature extraction, the recursive feature elimination algorithm was used to screen features, logistic regression was used to model features, immunohistochemistry staining with FDX1 antibody was performed to test the radiomic model. RESULTS FDX1 was found to be prognostic in various types of cancer. The area under the receiver operating characteristic curve of radiomic models to predict FDX1 expression reached 0.825 (95% CI = 0.739-0.911). Cross-tissue compatibility was confirmed in pan-cancer validation and test cohorts. Mechanistically, the radiomic score was significantly correlated with various immunosuppressive genes and gene mutations. The radiomic score was also found to be an independent prognostic factor, making it a potentially actionable biomarker in the clinical setting. CONCLUSIONS The expression of FDX1 could be non-invasively predicted via radiomics. The radiomic patterns with biological and clinical relevance across histology and modalities could have a broad impact on a larger population of patients.
Collapse
Affiliation(s)
- Qiuyuan Yue
- Department of Radiology, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350004, China
- Department of Radiology, Fujian Cancer Hospital & Fujian Medical University Cancer Hospital, Fuzhou, 350004, China
| | - Mingwei Zhang
- Department of Radiotherapy, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350004, China
- Key Laboratory of Radiation Biology of Fujian Higher Education Institutions, The First Affiliated Hospital, Fujian Medical University, Fuzhou, 350004, China
| | - Wenying Jiang
- Department of Breast Surgery, The Third Affiliated Hospital of Soochow University, Changzhou, 213000, China
- Department of Radiology, The Third Affiliated Hospital of Soochow University, Changzhou, 213000, China
| | - Lanmei Gao
- Key Laboratory of Child Development and Learning Science of Ministry of Education, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China
| | - Rongping Ye
- Department of Radiology, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350004, China
- Department of Radiology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital of Fujian Medical University, Fuzhou, 350212, China
| | - Jinsheng Hong
- Department of Radiotherapy, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350004, China.
- Key Laboratory of Radiation Biology of Fujian Higher Education Institutions, The First Affiliated Hospital, Fujian Medical University, Fuzhou, 350004, China.
| | - Yueming Li
- Department of Radiology, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350004, China.
- Department of Radiology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital of Fujian Medical University, Fuzhou, 350212, China.
| |
Collapse
|
5
|
Wu WF, Lai KM, Chen CH, Wang BC, Chen YJ, Shen CW, Chen KY, Lin EC, Chen CC. Predicting the T790M mutation in non-small cell lung cancer (NSCLC) using brain metastasis MR radiomics: a study with an imbalanced dataset. Discov Oncol 2024; 15:447. [PMID: 39277568 PMCID: PMC11401825 DOI: 10.1007/s12672-024-01333-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2024] [Accepted: 09/10/2024] [Indexed: 09/17/2024] Open
Abstract
BACKGROUND Early detection of T790M mutation in exon 20 of epidermal growth factor receptor (EGFR) in non-small cell lung cancer (NSCLC) patients with brain metastasis is crucial for optimizing treatment strategies. In this study, we developed radiomics models to distinguish NSCLC patients with T790M-positive mutations from those with T790M-negative mutations using multisequence MR images of brain metastasis despite an imbalanced dataset. Various resampling techniques and classifiers were employed to identify the most effective strategy. METHODS Radiomic analyses were conducted on a dataset comprising 125 patients, consisting of 18 with EGFR T790M-positive mutations and 107 with T790M-negative mutations. Seventeen first- and second-order statistical features were selected from CET1WI, T2WI, T2FLAIR, and DWI images. Four classifiers (logistic regression, support vector machine, random forest [RF], and extreme gradient boosting [XGBoost]) were evaluated under 13 different resampling conditions. RESULTS The area under the curve (AUC) value achieved was 0.89, using the SVM-SMOTE oversampling method in combination with the XGBoost classifier. This performance was measured against the AUC reported in the literature, serving as an upper-bound reference. Additionally, comparable results were observed with other oversampling methods paired with RF or XGBoost classifiers. CONCLUSIONS Our study demonstrates that, even when dealing with an imbalanced EGFR T790M dataset, reasonable predictive outcomes can be achieved by employing an appropriate combination of resampling techniques and classifiers. This approach has significant potential for enhancing T790M mutation detection in NSCLC patients with brain metastasis.
Collapse
Affiliation(s)
- Wen-Feng Wu
- Department of Radiology, Ditmanson Medical Foundation Chia-Yi Christian Hospital, Chiayi, 600, Taiwan
| | - Kuan-Ming Lai
- Department of Radiology, Ditmanson Medical Foundation Chia-Yi Christian Hospital, Chiayi, 600, Taiwan
- Central Taiwan University of Science and Technology Institute of Radiological Science, Taichung, 406, Taiwan
| | - Chia-Hung Chen
- Department of Radiology, Ditmanson Medical Foundation Chia-Yi Christian Hospital, Chiayi, 600, Taiwan
- Central Taiwan University of Science and Technology Institute of Radiological Science, Taichung, 406, Taiwan
| | - Bai-Chuan Wang
- Department of Chemistry and Biochemistry, National Chung Cheng University, 168 University Road, Min-Hsiung, Chiayi, 62102, Taiwan
| | - Yi-Jen Chen
- Department of Chemistry and Biochemistry, National Chung Cheng University, 168 University Road, Min-Hsiung, Chiayi, 62102, Taiwan
| | - Chia-Wei Shen
- Department of Chemistry and Biochemistry, National Chung Cheng University, 168 University Road, Min-Hsiung, Chiayi, 62102, Taiwan
| | - Kai-Yan Chen
- Department of Chemistry and Biochemistry, National Chung Cheng University, 168 University Road, Min-Hsiung, Chiayi, 62102, Taiwan
| | - Eugene C Lin
- Department of Chemistry and Biochemistry, National Chung Cheng University, 168 University Road, Min-Hsiung, Chiayi, 62102, Taiwan.
- Center for Nano Bio-Detection, National Chung Cheng University, Chiayi, 621, Taiwan.
| | - Chien-Chin Chen
- Department of Pathology, Ditmanson Medical Foundation Chia-Yi Christian Hospital, No. 539, Zhongxiao Rd., East Dist., Chiayi City, 60002, Taiwan.
- Rong Hsing Research Center for Translational Medicine, National Chung Hsing University, Taichung, 402, Taiwan.
- Department of Biotechnology and Bioindustry Sciences, College of Bioscience and Biotechnology, National Cheng Kung University, Tainan, 701, Taiwan.
- Department of Cosmetic Science, Chia Nan University of Pharmacy and Science, Tainan, 717, Taiwan.
| |
Collapse
|
6
|
Li S, Hu Y, Tian C, Luan J, Zhang X, Wei Q, Li X, Bian Y. Prediction of EGFR-TP53 genes co-mutations in patients with lung adenocarcinoma (LUAD) by 18F-FDG PET/CT radiomics. Clin Transl Oncol 2024:10.1007/s12094-024-03685-0. [PMID: 39251494 DOI: 10.1007/s12094-024-03685-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2024] [Accepted: 08/20/2024] [Indexed: 09/11/2024]
Abstract
PURPOSE This retrospective study was undertaken to assess the predictive efficacy of 18F-FDG PET/CT -derived radiomic features concerning the co-mutation status of epidermal growth factor receptor (EGFR) and TP53 in LUAD. METHODS A cohort of 150 LUAD patients underwent pretreatment 18F-FDG PET/CT scans with known mutation status of EGFR and TP53 were collected. The feature extraction based on their PET/CT images utilized the Pyradiomics package based on the 3D Slicer. The optimal radiomic features were selected through correlation analysis and the Gradient Boosting Decision Tree (GBDT) algorithm, followed by the construction of the radiomic model. The clinical model incorporated meaningful clinical variables, whereas the complex model integrated both the radiomic and clinical models. The area under the receiver operating characteristic curve (AUC) facilitated the comparison of prediction performance across the three models. The DCA gauged the clinical utility of these models. RESULTS The patient cohort was randomly allocated into a training set (n = 105) and a validation set (n = 45) in a 7:3 ratio. Eleven PET and eleven CT optimal radiomic features were selected to construct the radiomic model. The model showed a good ability to discriminate the co-occurrence of EGFR and TP53, with AUC equal to 0.850 in the training set, and 0.748 in the validation set, compared with 0.750 and 0.626 for the clinical model. The complex model exhibited the highest AUC values, with 0.880 and 0.794 in both sets, but there were no significant differences compared to the radiomic model. The DCA revealed favorable clinical value. CONCLUSION
Collapse
Affiliation(s)
- Shuheng Li
- Hebei Medical University, Shijiazhuang, Hebei, China
- Department of Nuclear Medicine, Hebei General Hospital, Shijiazhuang, Hebei, China
- Department of Nuclear Medicine, Affiliated Hospital of Hebei University, Baoding, Hebei, China
| | - Yujing Hu
- Department of Nuclear Medicine, Hebei General Hospital, Shijiazhuang, Hebei, China
| | - Congna Tian
- Department of Nuclear Medicine, Hebei General Hospital, Shijiazhuang, Hebei, China
| | - Jiusong Luan
- Department of Respiratory and Critical Care Medicine, Affiliated Hospital of Hebei University, Baoding, Hebei, China
| | - Xinchao Zhang
- Department of Nuclear Medicine, Hebei General Hospital, Shijiazhuang, Hebei, China
| | - Qiang Wei
- Department of Nuclear Medicine, Hebei General Hospital, Shijiazhuang, Hebei, China
| | - Xiaodong Li
- Department of Nuclear Medicine, Affiliated Hospital of Hebei University, Baoding, Hebei, China
| | - Yanzhu Bian
- Hebei Medical University, Shijiazhuang, Hebei, China.
- Department of Nuclear Medicine, Hebei General Hospital, Shijiazhuang, Hebei, China.
| |
Collapse
|
7
|
Zhuang Z, Lin J, Wan Z, Weng J, Yuan Z, Xie Y, Liu Z, Xie P, Mao S, Wang Z, Wang X, Huang M, Luo Y, Yu H. Radiogenomic profiling of global DNA methylation associated with molecular phenotypes and immune features in glioma. BMC Med 2024; 22:352. [PMID: 39218882 PMCID: PMC11367996 DOI: 10.1186/s12916-024-03573-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Accepted: 08/20/2024] [Indexed: 09/04/2024] Open
Abstract
BACKGROUND The radiogenomic analysis has provided valuable imaging biomarkers with biological insights for gliomas. The radiogenomic markers for molecular profile such as DNA methylation remain to be uncovered to assist the molecular diagnosis and tumor treatment. METHODS We apply the machine learning approaches to identify the magnetic resonance imaging (MRI) features that are associated with molecular profiles in 146 patients with gliomas, and the fitting models for each molecular feature (MoRad) are developed and validated. To provide radiological annotations for the molecular profiles, we devise two novel approaches called radiomic oncology (RO) and radiomic set enrichment analysis (RSEA). RESULTS The generated MoRad models perform well for profiling each molecular feature with radiomic features, including mutational, methylation, transcriptional, and protein profiles. Among them, the MoRad models have a remarkable performance in quantitatively mapping global DNA methylation. With RO and RSEA approaches, we find that global DNA methylation could be reflected by the heterogeneity in volumetric and textural features of enhanced regions in T2-weighted MRI. Finally, we demonstrate the associations of global DNA methylation with clinicopathological, molecular, and immunological features, including histological grade, mutations of IDH and ATRX, MGMT methylation, multiple methylation-high subtypes, tumor-infiltrating lymphocytes, and long-term survival outcomes. CONCLUSIONS Global DNA methylation is highly associated with radiological profiles in glioma. Radiogenomic global methylation is an imaging-based quantitative molecular biomarker that is associated with specific consensus molecular subtypes and immune features.
Collapse
Affiliation(s)
- Zhuokai Zhuang
- Department of General Surgery (Colorectal Surgery), The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
- Guangdong Institute of Gastroenterology, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, 510655, China
- Ministry of Education, Key Laboratory of Human Microbiome and Chronic Diseases (Sun Yat-Sen University), Guangzhou, Guangdong, China
| | - Jinxin Lin
- Department of General Surgery (Colorectal Surgery), The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
- Guangdong Institute of Gastroenterology, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, 510655, China
- Ministry of Education, Key Laboratory of Human Microbiome and Chronic Diseases (Sun Yat-Sen University), Guangzhou, Guangdong, China
| | - Zixiao Wan
- Guangdong Institute of Gastroenterology, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, 510655, China
- Ministry of Education, Key Laboratory of Human Microbiome and Chronic Diseases (Sun Yat-Sen University), Guangzhou, Guangdong, China
| | - Jingrong Weng
- Department of General Surgery (Colorectal Surgery), The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
- Guangdong Institute of Gastroenterology, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, 510655, China
| | - Ze Yuan
- Guangdong Institute of Gastroenterology, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, 510655, China
- Ministry of Education, Key Laboratory of Human Microbiome and Chronic Diseases (Sun Yat-Sen University), Guangzhou, Guangdong, China
| | - Yumo Xie
- Department of General Surgery (Colorectal Surgery), The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
- Guangdong Institute of Gastroenterology, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, 510655, China
| | - Zongchao Liu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Cancer Epidemiology, Peking University Cancer Institute, Beijing, 100142, China
| | - Peiyi Xie
- Department of Radiology, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, 510655, China
| | - Siyue Mao
- Image and Minimally Invasive Intervention Center, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, and Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Zongming Wang
- Department of Neurosurgery and Pituitary Tumor Center, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Xiaolin Wang
- Guangdong Institute of Gastroenterology, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, 510655, China
- Ministry of Education, Key Laboratory of Human Microbiome and Chronic Diseases (Sun Yat-Sen University), Guangzhou, Guangdong, China
| | - Meijin Huang
- Department of General Surgery (Colorectal Surgery), The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
- Guangdong Institute of Gastroenterology, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, 510655, China
- Ministry of Education, Key Laboratory of Human Microbiome and Chronic Diseases (Sun Yat-Sen University), Guangzhou, Guangdong, China
| | - Yanxin Luo
- Department of General Surgery (Colorectal Surgery), The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
- Guangdong Institute of Gastroenterology, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, 510655, China
- Ministry of Education, Key Laboratory of Human Microbiome and Chronic Diseases (Sun Yat-Sen University), Guangzhou, Guangdong, China
| | - Huichuan Yu
- Guangdong Institute of Gastroenterology, Guangzhou, Guangdong, China.
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China.
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, 510655, China.
- Ministry of Education, Key Laboratory of Human Microbiome and Chronic Diseases (Sun Yat-Sen University), Guangzhou, Guangdong, China.
| |
Collapse
|
8
|
Chen Y, Wu J, You J, Gao M, Lu S, Sun C, Shu Y, Wang X. Integrating IASLC grading and radiomics for predicting postoperative outcomes in stage IA invasive lung adenocarcinoma. Med Phys 2024; 51:6513-6524. [PMID: 38781536 DOI: 10.1002/mp.17177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Revised: 05/02/2024] [Accepted: 05/10/2024] [Indexed: 05/25/2024] Open
Abstract
BACKGROUND The International Association for the Study of Lung Cancer (IASLC) Pathology Committee introduced a histologic grading system for invasive lung adenocarcinoma (LUAD) in 2020. The IASLC grading system, hinging on the evaluation of predominant and high-grade histologic patterns, has proven to be practical and prognostic for invasive LUAD. However, there are still limitations in evaluating the prognosis of stage IA LUAD. Radiomics may serve as a valuable complement. PURPOSE To establish a model that integrates IASLC grading and radiomics, aimed at predicting the prognosis of stage IA LUAD. METHODS We conducted a retrospective analysis of 628 patients diagnosed with stage IA LUAD who underwent surgical resection between January 2015 and December 2018 at our institution. The patients were randomly divided into the training set (n = 439) and testing set (n = 189) at a ratio of 7:3. Overall survival (OS) and disease-free survival (DFS) were taken as the end points. Radiomics features were obtained by PyRadiomics. Feature selection was performed using the least absolute shrinkage and selection operator (LASSO). The prediction models for OS and DFS were developed using multivariate Cox regression analysis, and the models were visualized through nomogram plots. The model's performance was evaluated using area under the curves (AUC), concordance index (C-index), calibration curves, and survival decision curve analysis (DCA). RESULTS In total, nine radiomics features were selected for the OS prediction model, and 15 radiomics features were selected for the DFS prediction model. Patients with high radiomics scores were associated with a worse prognosis (p < 0.001). We built separate prediction models using radiomics or IASLC alone, as well as a combined prediction model. In the prediction of OS, we observed that the combined model (C-index: 0.812 ± 0.024, 3 years AUC: 0.692, 5 years AUC: 0.792) achieved superior predictive performance than the radiomics (C-index: 0.743 ± 0.038, 3 years AUC: 0.633, 5 years AUC: 0.768) and IASLC grading (C-index: 0.765 ± 0.042, 3 years AUC: 0.658, 5 years AUC: 0.743) models alone. Similar results were obtained in the models for DFS. CONCLUSION The combination of radiomics and IASLC pathological grading proves to be an effective approach for predicting the prognosis of stage IA LUAD. This has substantial clinical relevance in guiding treatment decisions for early-stage LUAD.
Collapse
Affiliation(s)
- Yong Chen
- First College of Clinical Medicine, Dalian Medical University, Dalian, China
| | - Jun Wu
- Medical College, Yangzhou University, Yangzhou, China
| | - Jie You
- First College of Clinical Medicine, Dalian Medical University, Dalian, China
| | - Mingjun Gao
- First College of Clinical Medicine, Dalian Medical University, Dalian, China
| | - Shichun Lu
- Department of Thoracic Surgery, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou, China
| | - Chao Sun
- Department of Thoracic Surgery, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou, China
| | - Yusheng Shu
- Department of Thoracic Surgery, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou, China
| | - Xiaolin Wang
- Department of Thoracic Surgery, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou, China
| |
Collapse
|
9
|
Lafon M, Cousin S, Alamé M, Nougaret S, Italiano A, Crombé A. Metastatic Lung Adenocarcinomas: Development and Evaluation of Radiomic-Based Methods to Measure Baseline Intra-Patient Inter-Tumor Lesion Heterogeneity. JOURNAL OF IMAGING INFORMATICS IN MEDICINE 2024:10.1007/s10278-024-01163-1. [PMID: 39020153 DOI: 10.1007/s10278-024-01163-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Revised: 06/03/2024] [Accepted: 06/04/2024] [Indexed: 07/19/2024]
Abstract
Radiomics has traditionally focused on individual tumors, often neglecting the integration of metastatic disease, particularly in patients with non-small cell lung cancer. This study sought to examine intra-patient inter-tumor lesion heterogeneity indices using radiomics, exploring their relevance in metastatic lung adenocarcinoma. Consecutive adults newly diagnosed with metastatic lung adenocarcinoma underwent contrast-enhanced CT scans for lesion segmentation and radiomic feature extraction. Three methods were devised to measure distances between tumor lesion profiles within the same patient in radiomic space: centroid to lesion, lesion to lesion, and primitive to lesion, with subsequent calculation of mean, range, and standard deviation of these distances. Associations between HIs, disease control rate, objective response rate to first-line treatment, and overall survival were explored. The study included 167 patients (median age 62.3 years) between 2016 and 2019, divided randomly into experimental (N = 117,546 lesions) and validation (N = 50,232 tumor lesions) cohorts. Patients without disease control/objective response and with poorer survival consistently systematically exhibited values of all heterogeneity indices. Multivariable analyses revealed that the range of primitive-to-lesion distances was associated with disease control in both cohorts and with objective response in the validation cohort. This metrics showed univariable associations with overall survival in the experimental. In conclusion, we proposed original methods to estimate the intra-patient inter-tumor lesion heterogeneity using radiomics that demonstrated correlations with patient outcomes, shedding light on the clinical implications of inter-metastases heterogeneity. This underscores the potential of radiomics in understanding and potentially predicting treatment response and prognosis in metastatic lung adenocarcinoma patients.
Collapse
Affiliation(s)
- Mathilde Lafon
- Department of Medical Oncology, Institut Bergonié, Bordeaux, France
| | - Sophie Cousin
- Department of Medical Oncology, Institut Bergonié, Bordeaux, France
| | - Mélissa Alamé
- Department of Biopathology, Institut Bergonié, Bordeaux, France
| | - Stéphanie Nougaret
- Medical Imaging Department, Montpellier Cancer Institute, Montpellier Cancer Research Institute (U1194), University of Montpellier, Montpellier, France
| | - Antoine Italiano
- Department of Medical Oncology, Institut Bergonié, Bordeaux, France
- SARCOTARGET Team, Bordeaux Research Institute in Oncology (BRIC) INSERM U1312 & University of Bordeaux, Bordeaux, France
| | - Amandine Crombé
- SARCOTARGET Team, Bordeaux Research Institute in Oncology (BRIC) INSERM U1312 & University of Bordeaux, Bordeaux, France.
- Department of Radiology, Institut Bergonié, Bordeaux, France.
- Department of Radiology, Pellegrin University Hospital, Bordeaux, France.
| |
Collapse
|
10
|
Wu J, Meng H, Zhou L, Wang M, Jin S, Ji H, Liu B, Jin P, Du C. Habitat radiomics and deep learning fusion nomogram to predict EGFR mutation status in stage I non-small cell lung cancer: a multicenter study. Sci Rep 2024; 14:15877. [PMID: 38982267 PMCID: PMC11233600 DOI: 10.1038/s41598-024-66751-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2024] [Accepted: 07/03/2024] [Indexed: 07/11/2024] Open
Abstract
Develop a radiomics nomogram that integrates deep learning, radiomics, and clinical variables to predict epidermal growth factor receptor (EGFR) mutation status in patients with stage I non-small cell lung cancer (NSCLC). We retrospectively included 438 patients who underwent curative surgery and completed driver-gene mutation tests for stage I NSCLC from four academic medical centers. Predictive models were established by extracting and analyzing radiomic features in intratumoral, peritumoral, and habitat regions of CT images to identify EGFR mutation status in stage I NSCLC. Additionally, three deep learning models based on the intratumoral region were constructed. A nomogram was developed by integrating representative radiomic signatures, deep learning, and clinical features. Model performance was assessed by calculating the area under the receiver operating characteristic (ROC) curve. The established habitat radiomics features demonstrated encouraging performance in discriminating between EGFR mutant and wild-type, with predictive ability superior to other single models (AUC 0.886, 0.812, and 0.790 for the training, validation, and external test sets, respectively). The radiomics-based nomogram exhibited excellent performance, achieving the highest AUC values of 0.917, 0.837, and 0.809 in the training, validation, and external test sets, respectively. Decision curve analysis (DCA) indicated that the nomogram provided a higher net benefit than other radiomics models, offering valuable information for treatment.
Collapse
Affiliation(s)
- Jingran Wu
- Department of Oncology, General Hospital of Northern Theater Command, Shenyang, 110840, China
| | - Hao Meng
- Department of Thoracic Surgery, General Hospital of Northern Theater Command, Shenyang, 110840, China
| | - Lin Zhou
- Department of Thoracic Surgery, Yuebei People's Hospital Affiliated to Shantou University Medical College, Shaoguan, 512025, China
| | - Meiling Wang
- Department of Oncology, General Hospital of Northern Theater Command, Shenyang, 110840, China
| | - Shanxiu Jin
- Department of Oncology, General Hospital of Northern Theater Command, Shenyang, 110840, China
| | - Hongjuan Ji
- Department of Oncology, General Hospital of Northern Theater Command, Shenyang, 110840, China
| | - Bona Liu
- Department of Oncology, General Hospital of Northern Theater Command, Shenyang, 110840, China.
| | - Peng Jin
- Department of Oncology, The Second Affiliated Hospital of Shandong First Medical University, Taian, 271000, China.
| | - Cheng Du
- Department of Oncology, General Hospital of Northern Theater Command, Shenyang, 110840, China.
| |
Collapse
|
11
|
Kohan A, Hinzpeter R, Kulanthaivelu R, Mirshahvalad SA, Avery L, Tsao M, Li Q, Ortega C, Metser U, Hope A, Veit-Haibach P. Contrast Enhanced CT Radiogenomics in a Retrospective NSCLC Cohort: Models, Attempted Validation of a Published Model and the Relevance of the Clinical Context. Acad Radiol 2024; 31:2953-2961. [PMID: 38383258 DOI: 10.1016/j.acra.2024.01.031] [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: 01/18/2024] [Accepted: 01/23/2024] [Indexed: 02/23/2024]
Abstract
RATIONALE AND OBJECTIVE To develop a radiogenomic predictive model for non-small cell lung cancer (NSCLC) patients studied through contrast enhanced chest computed tomography (CE-CT) targeting the most frequent gene alterations. M&M: A retrospective study of patients with NSCLC imaged with CE-CT before treatment and had their tumor genomics sequenced at our institution was performed. Data was gathered from their imaging studies, their electronic medical records and a web-based database search (cBioPortal.ca). All of the patient data was tabulated for analysis. Two predictive models (M1 & M2) were created using different approaches and a third model was extracted from the literature to also be tested in our population. RESULTS Out of 157 patients, eighty were male (51%) and 124 (79%) had a history of smoking. The three most prevalent genes were KRAS, TP53 and EGFR. The M1 radiomics-only model median AUC were 0.61 (TP53), 0.53 (KRAS) and 0.64 (EGFR) and for M1 radiomics + clinical were 0.61 (TP53), 0.61 (KRAS) and 0.80 (EGFR). The M2 radiomics-only model median AUC were 0.63 (TP53), 0.60 (KRAS) and 0.65 (EGFR) and for M2 radiomics + clinical were 0.64 (TP53), 0.62 (KRAS) and 0.81 (EGFR). The external EGFR radiomic model showed an AUC of 0.69 and 0.86 for the radiomics-only and combined radiomics + clinical respectively. CONCLUSION Our study was able to provide robust predictive radiomics model evaluation for the detection of TP53, KRAS and EGFR. We also compared our performance with an already published model and observed how impactful clinical variables can be on models' performance. CLINICAL RELEVANCE STATEMENT Identifying tumor mutations in patients that can't undergo biopsy is critical for their outcomes. KEYPOINTS • Tumor genomic profiling is critical for treatment selection • CE-CT radiomics produce robust predictive models comparable to those already published • Clinical variables should be considered/included in predictive models.
Collapse
Affiliation(s)
- A Kohan
- Joint Department of Medical Imaging, Princess Margaret Hospital, University Health Network, University of Toronto, Toronto, ON M5G 2C1, Canada.
| | - R Hinzpeter
- Joint Department of Medical Imaging, Princess Margaret Hospital, University Health Network, University of Toronto, Toronto, ON M5G 2C1, Canada; Institute of Diagnostic and Interventional Radiology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - R Kulanthaivelu
- Joint Department of Medical Imaging, Princess Margaret Hospital, University Health Network, University of Toronto, Toronto, ON M5G 2C1, Canada
| | - S A Mirshahvalad
- Joint Department of Medical Imaging, Princess Margaret Hospital, University Health Network, University of Toronto, Toronto, ON M5G 2C1, Canada
| | - L Avery
- Department of Biostatistics, Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto, ON M5G 2C1, Canada
| | - M Tsao
- University Health Network, Ontario Cancer Institute/Princess Margaret Cancer Centre, Toronto, ON, Canada
| | - Q Li
- University Health Network, Ontario Cancer Institute/Princess Margaret Cancer Centre, Toronto, ON, Canada
| | - C Ortega
- Joint Department of Medical Imaging, Princess Margaret Hospital, University Health Network, University of Toronto, Toronto, ON M5G 2C1, Canada
| | - U Metser
- Joint Department of Medical Imaging, Princess Margaret Hospital, University Health Network, University of Toronto, Toronto, ON M5G 2C1, Canada
| | - A Hope
- Department of Radiation Oncology, University Health Network, University of Toronto, ON, Canada
| | - P Veit-Haibach
- Joint Department of Medical Imaging, Princess Margaret Hospital, University Health Network, University of Toronto, Toronto, ON M5G 2C1, Canada
| |
Collapse
|
12
|
Zhang G, Man Q, Shang L, Zhang J, Cao Y, Li S, Qian R, Ren J, Pu H, Zhou J, Zhang Z, Kong W. Using Multi-phase CT Radiomics Features to Predict EGFR Mutation Status in Lung Adenocarcinoma Patients. Acad Radiol 2024; 31:2591-2600. [PMID: 38290884 DOI: 10.1016/j.acra.2023.12.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Revised: 12/12/2023] [Accepted: 12/15/2023] [Indexed: 02/01/2024]
Abstract
RATIONALE AND OBJECTIVES This study aimed to non-invasively predict epidermal growth factor receptor (EGFR) mutation status in patients with lung adenocarcinoma using multi-phase computed tomography (CT) radiomics features. MATERIALS AND METHODS A total of 424 patients with lung adenocarcinoma were recruited from two hospitals who underwent preoperative non-enhanced CT (NE-CT) and enhanced CT (including arterial phase CT [AP-CT], and venous phase CT [VP-CT]). Patients were divided into training (n = 297) and external validation (n = 127) cohorts according to hospital. Radiomics features were extracted from the NE-CT, AP-CT, and VP-CT images, respectively. The Wilcoxon test, correlation analysis, and simulated annealing were used for feature screening. A clinical model and eight radiomics models were established. Furthermore, a clinical-radiomics model was constructed by incorporating multi-phase CT features and clinical risk factors. Receiver operating characteristic curves were used to evaluate the predictive performance of the models. RESULTS The predictive performance of multi-phase CT radiomics model (AUC of 0.925 [95% CI, 0.879-0.971] in the validation cohort) was higher than that of NE-CT, AP-CT, VP-CT, and clinical models (AUCs of 0.860 [95% CI,0.794-0.927], 0.792 [95% CI, 0.713-0.871], 0.753 [95% CI, 0.669-0.838], and 0.706 [95% CI, 0.620-0.791] in the validation cohort, respectively) (all P < 0.05). The predictive performance of the clinical-radiomics model (AUC of 0.927 [95% CI, 0.882-0.971] in the validation cohort) was comparable to that of multi-phase CT radiomics model (P > 0.05). CONCLUSION Our multi-phase CT radiomics model showed good performance in identifying the EGFR mutation status in patients with lung adenocarcinoma, which may assist personalized treatment decisions.
Collapse
Affiliation(s)
- Guojin Zhang
- Department of Radiology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China (G.Z., L.S., R.Q., H.P., W.K.)
| | - Qiong Man
- School of Pharmacy, Chengdu Medical College, Chengdu, China (Q.M.)
| | - Lan Shang
- Department of Radiology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China (G.Z., L.S., R.Q., H.P., W.K.)
| | - Jing Zhang
- Department of Radiology, Fifth Affiliated Hospital of Zunyi Medical University, Zhuhai, China (J.Z.)
| | - Yuntai Cao
- Department of Radiology, Affiliated Hospital of Qinghai University, Xining, China (Y.C.)
| | - Shenglin Li
- Department of Radiology, Lanzhou University Second Hospital, Lanzhou, China (S.L., J.Z.)
| | - Rong Qian
- Department of Radiology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China (G.Z., L.S., R.Q., H.P., W.K.)
| | - Jialiang Ren
- ŌGE Healthcare China, Department of Radiology, China (J.R.)
| | - Hong Pu
- Department of Radiology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China (G.Z., L.S., R.Q., H.P., W.K.)
| | - Junlin Zhou
- Department of Radiology, Lanzhou University Second Hospital, Lanzhou, China (S.L., J.Z.)
| | - Zhuoli Zhang
- Department of Radiology and BME, University of California Irvine, Irvine, California, USA (Z.Z.)
| | - Weifang Kong
- Department of Radiology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China (G.Z., L.S., R.Q., H.P., W.K.).
| |
Collapse
|
13
|
Kuang B, Zhang J, Zhang M, Xia H, Qiang G, Zhang J. Advancing NSCLC pathological subtype prediction with interpretable machine learning: a comprehensive radiomics-based approach. Front Med (Lausanne) 2024; 11:1413990. [PMID: 38841579 PMCID: PMC11150591 DOI: 10.3389/fmed.2024.1413990] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Accepted: 05/10/2024] [Indexed: 06/07/2024] Open
Abstract
Objective This research aims to develop and assess the performance of interpretable machine learning models for diagnosing three histological subtypes of non-small cell lung cancer (NSCLC) utilizing CT imaging data. Methods A retrospective cohort of 317 patients diagnosed with NSCLC was included in the study. These individuals were randomly segregated into two groups: a training set comprising 222 patients and a validation set with 95 patients, adhering to a 7:3 ratio. A comprehensive extraction yielded 1,834 radiomic features. For feature selection, statistical methodologies such as the Mann-Whitney U test, Spearman's rank correlation, and one-way logistic regression were employed. To address data imbalance, the Synthetic Minority Over-sampling Technique (SMOTE) was utilized. The study designed three distinct models to predict adenocarcinoma (ADC), squamous cell carcinoma (SCC), and large cell carcinoma (LCC). Six different classifiers, namely Logistic Regression, Support Vector Machine, Decision Tree, Random Forest, eXtreme Gradient Boosting (XGB), and LightGBM, were deployed for model training. Model performance was gauged through accuracy metrics and the area under the receiver operating characteristic (ROC) curves (AUC). To interpret the diagnostic process, the Shapley Additive Explanations (SHAP) approach was applied. Results For the ADC, SCC, and LCC groups, 9, 12, and 8 key radiomic features were selected, respectively. In terms of model performance, the XGB model demonstrated superior performance in predicting SCC and LCC, with AUC values of 0.789 and 0.848, respectively. For ADC prediction, the Random Forest model excelled, showcasing an AUC of 0.748. Conclusion The constructed machine learning models, leveraging CT imaging, exhibited robust predictive capabilities for SCC, LCC, and ADC subtypes of NSCLC. These interpretable models serve as substantial support for clinical decision-making processes.
Collapse
Affiliation(s)
- Bingling Kuang
- Department of Pathology, Affiliated Cancer Hospital and Institution of Guangzhou Medical University, Guangzhou, China
- Nanshan College, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Jingxuan Zhang
- Nanshan College, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Mingqi Zhang
- The Second Clinical School of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Haoming Xia
- School of Clinical Medicine, Tsinghua University, Beijing, China
| | - Guangliang Qiang
- Department of Thoracic Surgery, Peking University Third Hospital, Beijing, China
| | - Jiangyu Zhang
- Department of Pathology, Affiliated Cancer Hospital and Institution of Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
14
|
Li Y, Jin Y, Wang Y, Liu W, Jia W, Wang J. MR-based radiomics predictive modelling of EGFR mutation and HER2 overexpression in metastatic brain adenocarcinoma: a two-centre study. Cancer Imaging 2024; 24:65. [PMID: 38773634 PMCID: PMC11110398 DOI: 10.1186/s40644-024-00709-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Accepted: 05/11/2024] [Indexed: 05/24/2024] Open
Abstract
OBJECTIVES Magnetic resonance (MR)-based radiomics features of brain metastases are utilised to predict epidermal growth factor receptor (EGFR) mutation and human epidermal growth factor receptor 2 (HER2) overexpression in adenocarcinoma, with the aim to identify the most predictive MR sequence. METHODS A retrospective inclusion of 268 individuals with brain metastases from adenocarcinoma across two institutions was conducted. Utilising T1-weighted imaging (T1 contrast-enhanced [T1-CE]) and T2 fluid-attenuated inversion recovery (T2-FLAIR) sequences, 1,409 radiomics features were extracted. These sequences were randomly divided into training and test sets at a 7:3 ratio. The selection of relevant features was done using the least absolute shrinkage selection operator, and the training cohort's support vector classifier model was employed to generate the predictive model. The performance of the radiomics features was evaluated using a separate test set. RESULTS For contrast-enhanced T1-CE cohorts, the radiomics features based on 19 selected characteristics exhibited excellent discrimination. No significant differences in age, sex, and time to metastasis were observed between the groups with EGFR mutations or HER2 + and those with wild-type EGFR or HER2 (p > 0.05). Radiomics feature analysis for T1-CE revealed an area under the curve (AUC) of 0.98, classification accuracy of 0.93, sensitivity of 0.92, and specificity of 0.93 in the training cohort. In the test set, the AUC was 0.82. The 19 radiomics features for the T2-FLAIR sequence showed AUCs of 0.86 in the training set and 0.70 in the test set. CONCLUSIONS This study developed a T1-CE signature that could serve as a non-invasive adjunctive tool to determine the presence of EGFR mutations and HER2 + status in adenocarcinoma, aiding in the direction of treatment plans. CLINICAL RELEVANCE STATEMENT We propose radiomics features based on T1-CE brain MR sequences that are both evidence-based and non-invasive. These can be employed to guide clinical treatment planning in patients with brain metastases from adenocarcinoma.
Collapse
Affiliation(s)
- Yanran Li
- Department of Radiology, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, 830054, China
| | - Yong Jin
- Department of Radiology, Changzhi People's Hospital, Changzhi, 046000, Shanxi Province, China
| | - Yunling Wang
- Department of Radiology, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, 830054, China
| | - Wenya Liu
- Department of Radiology, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, 830054, China
| | - Wenxiao Jia
- Department of Radiology, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, 830054, China
| | - Jian Wang
- Department of Radiology, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, 830054, China.
| |
Collapse
|
15
|
Wu T, Gao C, Lou X, Wu J, Xu M, Wu L. Predictive value of radiomic features extracted from primary lung adenocarcinoma in forecasting thoracic lymph node metastasis: a systematic review and meta-analysis. BMC Pulm Med 2024; 24:246. [PMID: 38762472 PMCID: PMC11102161 DOI: 10.1186/s12890-024-03020-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Accepted: 04/16/2024] [Indexed: 05/20/2024] Open
Abstract
BACKGROUND The application of radiomics in thoracic lymph node metastasis (LNM) of lung adenocarcinoma is increasing, but diagnostic performance of radiomics from primary tumor to predict LNM has not been systematically reviewed. Therefore, this study sought to provide a general overview regarding the methodological quality and diagnostic performance of using radiomic approaches to predict the likelihood of LNM in lung adenocarcinoma. METHODS Studies were gathered from literature databases such as PubMed, Embase, the Web of Science Core Collection, and the Cochrane library. The Radiomic Quality Score (RQS) and the Quality Assessment of Diagnostic Accuracy Studies-2 (QUADAS-2) were both used to assess the quality of each study. The pooled sensitivity, specificity, and area under the curve (AUC) of the best radiomics models in the training and validation cohorts were calculated. Subgroup and meta-regression analyses were also conducted. RESULTS Seventeen studies with 159 to 1202 patients each were enrolled between the years of 2018 to 2022, of which ten studies had sufficient data for the quantitative evaluation. The percentage of RQS was between 11.1% and 44.4% and most of the studies were considered to have a low risk of bias and few applicability concerns in QUADAS-2. Pyradiomics and logistic regression analysis were the most commonly used software and methods for radiomics feature extraction and selection, respectively. In addition, the best prediction models in seventeen studies were mainly based on radiomics features combined with non-radiomics features (semantic features and/or clinical features). The pooled sensitivity, specificity, and AUC of the training cohorts were 0.84 (95% confidence interval (CI) [0.73-0.91]), 0.88 (95% CI [0.81-0.93]), and 0.93(95% CI [0.90-0.95]), respectively. For the validation cohorts, the pooled sensitivity, specificity, and AUC were 0.89 (95% CI [0.82-0.94]), 0.86 (95% CI [0.74-0.93]) and 0.94 (95% CI [0.91-0.96]), respectively. CONCLUSIONS Radiomic features based on the primary tumor have the potential to predict preoperative LNM of lung adenocarcinoma. However, radiomics workflow needs to be standardized to better promote the applicability of radiomics. TRIAL REGISTRATION CRD42022375712.
Collapse
Affiliation(s)
- Ting Wu
- Department of Radiology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), 54 Youdian Road, Hangzhou, China
- The First School of Clinical Medicine of Zhejiang Chinese Medical University, 548 Binwen Road, Hangzhou, China
| | - Chen Gao
- Department of Radiology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), 54 Youdian Road, Hangzhou, China
- The First School of Clinical Medicine of Zhejiang Chinese Medical University, 548 Binwen Road, Hangzhou, China
| | - Xinjing Lou
- Department of Radiology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), 54 Youdian Road, Hangzhou, China
- The First School of Clinical Medicine of Zhejiang Chinese Medical University, 548 Binwen Road, Hangzhou, China
| | - Jun Wu
- Department of Radiology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), 54 Youdian Road, Hangzhou, China
- The First School of Clinical Medicine of Zhejiang Chinese Medical University, 548 Binwen Road, Hangzhou, China
| | - Maosheng Xu
- Department of Radiology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), 54 Youdian Road, Hangzhou, China.
- The First School of Clinical Medicine of Zhejiang Chinese Medical University, 548 Binwen Road, Hangzhou, China.
| | - Linyu Wu
- Department of Radiology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), 54 Youdian Road, Hangzhou, China.
- The First School of Clinical Medicine of Zhejiang Chinese Medical University, 548 Binwen Road, Hangzhou, China.
| |
Collapse
|
16
|
Xu J, Yang Y, Gao Z, Song T, Ma Y, Yu X, Shi C. Distinguishing EGFR mutation molecular subtypes based on MRI radiomics features of lung adenocarcinoma brain metastases. Clin Neurol Neurosurg 2024; 240:108258. [PMID: 38552362 DOI: 10.1016/j.clineuro.2024.108258] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2024] [Accepted: 03/23/2024] [Indexed: 04/29/2024]
Abstract
OBJECTIVE To explore the feasibility of identifying epidermal growth factor receptor (EGFR) mutation molecular subtypes in primary lesions based on the radiomics features of lung adenocarcinoma brain metastases using magnetic resonance imaging (MRI). METHODS We retrospectively analyzed clinical, imaging, and genetic testing data of patients with lung adenocarcinoma with EGFR gene mutations who had brain metastases. Three-dimensional radiomics features were extracted from contrast-enhanced T1-weighted images. The volume of interest was delineated and normalized using Z-score, dimensionality reduction was performed using principal component analysis, feature selection using Relief, and radiomics model construction using adaptive boosting as a classifier. Data were randomly divided into training and testing datasets at an 8:2 ratio. Five-fold cross-validation was conducted in the training set to select the optimal radiomics features and establish a predictive model for distinguishing between exon 19 deletion (19Del) and exon 21 L858R point mutation (21L858R), the two most common EGFR gene mutations. The testing set was used for external validation of the models. Model performance was evaluated using receiver operating characteristic curve and decision curve analyses. RESULTS Overall, 86 patients with 228 brain metastases were included. Patient age was identified as an independent predictor for distinguishing between 19Del and 21L858R. The area under the curve (AUC) values of the radiomics model in the training and testing datasets were 0.895 (95% confidence interval [CI]: 0.850-0.939) and 0.759 (95% CI: 0.0.614-0.903), respectively. The AUC for diagnosis of all cases using a combined model of age and radiomics was 0.888 (95% CI: 0.846-0.930), slightly higher than that of the radiomics model alone (0.866, 95% CI: 0.820-0.913), but without statistical significance (p=0.1626). In the decision curve analysis, both models demonstrated clinical net benefits. CONCLUSIONS The radiomics model based on MRI of lung adenocarcinoma brain metastases could distinguish between EGFR 19Del and 21L858R mutations in the primary lesion.
Collapse
Affiliation(s)
- Jiali Xu
- Department of Radiology, The First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui 233004, China; Department of Medical Imaging Diagnosis, School of Medical Imaging, Bengbu Medical University, Bengbu, Anhui, China.
| | - Yuqiong Yang
- Department of Radiology, The First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui 233004, China; School of Graduate, Bengbu Medical University, Bengbu, Anhui 233030,China
| | - Zhizhen Gao
- Department of Radiology, The First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui 233004, China
| | - Tao Song
- Vascular Surgery Department, the First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui 233004, China
| | - Yichuan Ma
- Department of Radiology, The First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui 233004, China
| | - Xiaojun Yu
- Department of Medical Imaging Center, the First Affiliated Hospital, Jinan University, Guangzhou 510630, China
| | - Changzheng Shi
- Department of Medical Imaging Center, the First Affiliated Hospital, Jinan University, Guangzhou 510630, China
| |
Collapse
|
17
|
Li F, Qi L, Cheng S, Liu J, Chen J, Cui S, Dong S, Wang J. Predicting epidermal growth factor receptor mutations in non-small cell lung cancer through dual-layer spectral CT: a prospective study. Insights Imaging 2024; 15:109. [PMID: 38679659 PMCID: PMC11056350 DOI: 10.1186/s13244-024-01678-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Accepted: 03/22/2024] [Indexed: 05/01/2024] Open
Abstract
OBJECTIVE To determine whether quantitative parameters of detector-derived dual-layer spectral computed tomography (DLCT) can reliably identify epidermal growth factor receptor (EGFR) mutation status in patients with non-small cell lung cancer (NSCLC). METHODS Patients with NSCLC who underwent arterial phase (AP) and venous phase (VP) DLCT between December 2021 and November 2022 were subdivided into the mutated and wild-type EGFR groups following EGFR mutation testing. Their baseline clinical data, conventional CT images, and spectral images were obtained. Iodine concentration (IC), iodine no water (INW), effective atomic number (Zeff), virtual monoenergetic images, the slope of the spectral attenuation curve (λHU), enhancement degree (ED), arterial enhancement fraction (AEF), and normalized AEF (NAEF) were measured for each lesion. RESULTS Ninety-two patients (median age, 61 years, interquartile range [51, 67]; 33 men) were evaluated. The univariate analysis indicated that IC, normalized IC (NIC), INW and ED for the AP and VP, as well as Zeff and λHU for the VP were significantly associated with EGFR mutation status (all p < 0.05). INW(VP) showed the best diagnostic performance (AUC, 0.892 [95% confidence interval {CI}: 0.823, 0.960]). However, neither AEF (p = 0.156) nor NAEF (p = 0.567) showed significant differences between the two groups. The multivariate analysis showed that INW(AP) and NIC(VP) were significant predictors of EGFR mutation status, with the latter showing better performance (p = 0.029; AUC, 0.897 [95% CI: 0.816, 0.951] vs. 0.774 [95% CI: 0.675, 0.855]). CONCLUSION Quantitative parameters of DLCT can help predict EGFR mutation status in patients with NSCLC. CRITICAL RELEVANCE STATEMENT Quantitative parameters of DLCT, especially NIC(VP), can help predict EGFR mutation status in patients with NSCLC, facilitating appropriate and individualized treatment for them. KEY POINTS Determining EGFR mutation status in patients with NSCLC before starting therapy is essential. Quantitative parameters of DLCT can predict EGFR mutation status in NSCLC patients. NIC in venous phase is an important parameter to guide individualized treatment selection for NSCLC patients.
Collapse
Affiliation(s)
- Fenglan Li
- Department of Diagnostic Radiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 17 Panjiayuan Nanli, Beijing, Chaoyang District, 100021, China
| | - Linlin Qi
- Department of Diagnostic Radiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 17 Panjiayuan Nanli, Beijing, Chaoyang District, 100021, China
| | - Sainan Cheng
- Department of Diagnostic Radiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 17 Panjiayuan Nanli, Beijing, Chaoyang District, 100021, China
| | - Jianing Liu
- Department of Diagnostic Radiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 17 Panjiayuan Nanli, Beijing, Chaoyang District, 100021, China
| | - Jiaqi Chen
- Department of Diagnostic Radiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 17 Panjiayuan Nanli, Beijing, Chaoyang District, 100021, China
| | - Shulei Cui
- Department of Diagnostic Radiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 17 Panjiayuan Nanli, Beijing, Chaoyang District, 100021, China
| | - Shushan Dong
- Clinical Science, Philips Healthcare, Beijing, China
| | - Jianwei Wang
- Department of Diagnostic Radiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 17 Panjiayuan Nanli, Beijing, Chaoyang District, 100021, China.
| |
Collapse
|
18
|
Zhen T, Fang J, Hu D, Shen Q, Ruan M. Comparative evaluation of multiparametric lumbar MRI radiomic models for detecting osteoporosis. BMC Musculoskelet Disord 2024; 25:185. [PMID: 38424582 PMCID: PMC10902949 DOI: 10.1186/s12891-024-07309-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2023] [Accepted: 02/24/2024] [Indexed: 03/02/2024] Open
Abstract
BACKGROUND Osteoporosis is a serious global public health issue. Currently, there are few studies that explore the use of multiparametric MRI radiomics for osteoporosis detection. The purpose of this study was to compare the performance of radiomics features from multiple MRI sequences (T1WI, T2WI and T1WI combined with T2WI) for detecting osteoporosis in patients. METHODS A retrospective analysis was performed on 160 patients who had undergone dual-energy X-ray absorptiometry(DXA) and lumbar magnetic resonance imaging (MRI) at our hospital. Among them, 86 patients were diagnosed with abnormal bone mass (osteoporosis or low bone mass), and 74 patients were diagnosed with normal bone mass based on the DXA results. Sagittal T1-and T2-weighted images of all patients were imported into the uAI Research Portal (United Imaging Intelligence) for image delineation and radiomics analysis, where a series of radiomic features were obtained. A radiomic model that included T1WI, T2WI, and T1WI+T2WI was established using features selected by LASSO regression. We used ROC curve analysis to evaluate the predictive efficacy of each model for identifying bone abnormalities and conducted decision curve analysis (DCA) to evaluate the net benefit of each model. Finally, we validated the model in a sample of 35 patients from different health care institution. RESULTS The T1WI + T2WI radiomics model showed better screening performance for patients with abnormal bone mass. In the training group, the sensitivity was 0.758, the specificity was 0.78, and the accuracy was 0.768 (AUC =0.839, 95% CI=0.757-0.901). In the validation group, the sensitivity was 0.792, the specificity was 0.875, and the accuracy was 0.833 (AUC =0.86, 95% CI=0.73-0.943).The DCA also showed that the combined model had better net benefits. In the external validation group, the sensitivity was 0.764, the specificity was 0.833, and the accuracy was 0.8 (AUC =0.824, 95% CI 0.678-0.969). CONCLUSIONS Radiomics-based multiparametric MRI can be used for the quantitative analysis of lumbar MRI and for accurately screening patients with abnormal bone mass.
Collapse
Affiliation(s)
- Tao Zhen
- Department of Radiology, Hangzhou First People's Hospital, No.261, Huansha Road, Hangzhou, Zhejiang, 310006, China.
| | - Jing Fang
- Zhejiang Provincial Hospital of Traditional Chinese medicine, Hangzhou, 310006, China
| | - Dacheng Hu
- Department of Radiology, Hangzhou First People's Hospital, No.261, Huansha Road, Hangzhou, Zhejiang, 310006, China
| | - Qijun Shen
- Department of Radiology, Hangzhou First People's Hospital, No.261, Huansha Road, Hangzhou, Zhejiang, 310006, China
| | - Mei Ruan
- Department of Radiology, Hangzhou First People's Hospital, No.261, Huansha Road, Hangzhou, Zhejiang, 310006, China
| |
Collapse
|
19
|
Zhang X, Zhang G, Qiu X, Yin J, Tan W, Yin X, Yang H, Wang H, Zhang Y. Non-invasive decision support for clinical treatment of non-small cell lung cancer using a multiscale radiomics approach. Radiother Oncol 2024; 191:110082. [PMID: 38195018 DOI: 10.1016/j.radonc.2024.110082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Revised: 12/01/2023] [Accepted: 01/02/2024] [Indexed: 01/11/2024]
Abstract
BACKGROUND Selecting therapeutic strategies for cancer patients is typically based on key target-molecule biomarkers that play an important role in cancer onset, progression, and prognosis. Thus, there is a pressing need for novel biomarkers that can be utilized longitudinally to guide treatment selection. METHODS Using data from 508 non-small cell lung cancer (NSCLC) patients across three institutions, we developed and validated a comprehensive predictive biomarker that distinguishes six genotypes and infiltrative immune phenotypes. These features were analyzed to establish the association between radiological phenotypes and tumor genotypes/immune phenotypes and to create a radiological interpretation of molecular features. In addition, we assessed the sensitivity of the models by evaluating their performance at five different voxel intervals, resulting in improved generalizability of the proposed approach. FINDINGS The radiomics model we developed, which integrates clinical factors and multi-regional features, outperformed the conventional model that only uses clinical and intratumoral features. Our combined model showed significant performance for EGFR, KRAS, ALK, TP53, PIK3CA, and ROS1 mutation status with AUCs of 0.866, 0.874, 0.902, 0.850, 0.860, and 0.900, respectively. Additionally, the predictive performance for PD-1/PD-L1 was 0.852. Although the performance of all models decreased to different degrees at five different voxel space resolutions, the performance advantage of the combined model did not change. CONCLUSIONS We validated multiscale radiomic signatures across tumor genotypes and immunophenotypes in a multi-institutional cohort. This imaging-based biomarker offers a non-invasive approach to select patients with NSCLC who are sensitive to targeted therapies or immunotherapy, which is promising for developing personalized treatment strategies during therapy.
Collapse
Affiliation(s)
- Xingping Zhang
- School of Medical Information Engineering, Gannan Medical University, 341000, Ganzhou, China; Cyberspace Institute of Advanced Technology, Guangzhou University, 510006 Guangzhou, China; Institute for Sustainable Industries and Liveable Cities, Victoria University, 3011, Melbourne, Australia; Department of New Networks, Peng Cheng Laboratory, 518000, Shenzhen, China
| | - Guijuan Zhang
- Department of Respiratory and Critical Care, First Affiliated Hospital of Gannan Medical University, 341000, Ganzhou, China
| | - Xingting Qiu
- Department of Radiology, First Affiliated Hospital of Gannan Medical University, 341000, Ganzhou, China
| | - Jiao Yin
- Institute for Sustainable Industries and Liveable Cities, Victoria University, 3011, Melbourne, Australia
| | - Wenjun Tan
- Key Laboratory of Intelligent Computing in Medical Image, Ministry of Education, Northeastern University, 110189, Shenyang, China
| | - Xiaoxia Yin
- Cyberspace Institute of Advanced Technology, Guangzhou University, 510006 Guangzhou, China
| | - Hong Yang
- Cyberspace Institute of Advanced Technology, Guangzhou University, 510006 Guangzhou, China
| | - Hua Wang
- Institute for Sustainable Industries and Liveable Cities, Victoria University, 3011, Melbourne, Australia.
| | - Yanchun Zhang
- Institute for Sustainable Industries and Liveable Cities, Victoria University, 3011, Melbourne, Australia; School of Computer Science and Technology, Zhejiang Normal University, 321000, Jinhua, China; Department of New Networks, Peng Cheng Laboratory, 518000, Shenzhen, China.
| |
Collapse
|
20
|
LoPiccolo J, Gusev A, Christiani DC, Jänne PA. Lung cancer in patients who have never smoked - an emerging disease. Nat Rev Clin Oncol 2024; 21:121-146. [PMID: 38195910 PMCID: PMC11014425 DOI: 10.1038/s41571-023-00844-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/28/2023] [Indexed: 01/11/2024]
Abstract
Lung cancer is the most common cause of cancer-related deaths globally. Although smoking-related lung cancers continue to account for the majority of diagnoses, smoking rates have been decreasing for several decades. Lung cancer in individuals who have never smoked (LCINS) is estimated to be the fifth most common cause of cancer-related deaths worldwide in 2023, preferentially occurring in women and Asian populations. As smoking rates continue to decline, understanding the aetiology and features of this disease, which necessitate unique diagnostic and treatment paradigms, will be imperative. New data have provided important insights into the molecular and genomic characteristics of LCINS, which are distinct from those of smoking-associated lung cancers and directly affect treatment decisions and outcomes. Herein, we review the emerging data regarding the aetiology and features of LCINS, particularly the genetic and environmental underpinnings of this disease as well as their implications for treatment. In addition, we outline the unique diagnostic and therapeutic paradigms of LCINS and discuss future directions in identifying individuals at high risk of this disease for potential screening efforts.
Collapse
Affiliation(s)
- Jaclyn LoPiccolo
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA.
- The Lowe Center for Thoracic Oncology, Dana-Farber Cancer Institute, Boston, MA, USA.
| | - Alexander Gusev
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- The Eli and Edythe L. Broad Institute, Cambridge, MA, USA
| | - David C Christiani
- Department of Environmental Health, Harvard T. H. Chan School of Public Health, Boston, MA, USA
- Massachusetts General Hospital, Boston, MA, USA
| | - Pasi A Jänne
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- The Lowe Center for Thoracic Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| |
Collapse
|
21
|
Hou S, Wang H, Wang X, Chen H, Zhou B, Meng R, Sha X, Chang S, Wang H, Jiang W. Tumor-liver interface in MRI of liver metastasis enables prediction of EGFR mutation in patients with lung cancer: A proof-of-concept study. Med Phys 2024; 51:1083-1091. [PMID: 37408393 DOI: 10.1002/mp.16581] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Revised: 04/19/2023] [Accepted: 06/05/2023] [Indexed: 07/07/2023] Open
Abstract
BACKGROUND Preoperative prediction of the epidermal growth factor receptor (EGFR) status in non-small-cell lung cancer (NSCLC) patients with liver metastasis (LM) may have potential clinical values for assisting in treatment decision-making. PURPOSE To explore the value of tumor-liver interface (TLI)-based magnetic resonance imaging (MRI) radiomics for detecting the EGFR mutation in NSCLC patients with LM. METHODS This retrospective study included 123 and 44 patients from hospital 1 (between Feb. 2018 and Dec. 2021) and hospital 2 (between Nov. 2015 and Aug. 2022), respectively. The patients received contrast-enhanced T1-weighted (CET1) and T2-weighted (T2W) liver MRI scans before treatment. Radiomics features were extracted from MRI images of TLI and the whole tumor region, separately. The least absolute shrinkage and selection operator (LASSO) regression was used to screen the features and establish radiomics signatures (RSs) based on TLI (RS-TLI) and the whole tumor (RS-W). The RSs were evaluated by the receiver operating characteristic (ROC) curve analysis. RESULTS A total of 5 and 6 features were identified highly correlated with the EGFR mutation status from TLI and the whole tumor, respectively. The RS-TLI showed better prediction performance than RS-W in the training (AUCs, RS-TLI vs. RS-W, 0.842 vs. 0.797), internal validation (AUCs, RS-TLI vs. RS-W, 0.771 vs. 0.676) and external validation (AUCs, RS-TLI vs. RS-W, 0.733 vs. 0.679) cohort. CONCLUSION Our study demonstrated that TLI-based radiomics can improve prediction performance of the EGFR mutation in lung cancer patients with LM. The established multi-parametric MRI radiomics models may be used as new markers that can potentially assist in personalized treatment planning.
Collapse
Affiliation(s)
- Shaoping Hou
- School of Intelligent Medicine, China Medical University, Shenyang, Liaoning, P.R. China
| | - Hongbo Wang
- Department of Radiology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, P.R. China
| | - Xiaoyu Wang
- Department of Radiology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang, P.R. China
| | - Huanhuan Chen
- Department of Oncology, Shengjing Hospital, Shenyang, Liaoning, P.R. China
| | - Boyu Zhou
- School of Intelligent Medicine, China Medical University, Shenyang, Liaoning, P.R. China
| | - Ruiqing Meng
- School of Intelligent Medicine, China Medical University, Shenyang, Liaoning, P.R. China
| | - Xianzheng Sha
- School of Intelligent Medicine, China Medical University, Shenyang, Liaoning, P.R. China
| | - Shijie Chang
- School of Intelligent Medicine, China Medical University, Shenyang, Liaoning, P.R. China
| | - Huan Wang
- Radiation Oncology Department of Thoracic Cancer, Liaoning Cancer Hospital and Institute, Shenyang, Liaoning, P.R. China
| | - Wenyan Jiang
- Department of Scientific Research and Academic, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang, Liaoning, P.R. China
| |
Collapse
|
22
|
Nguyen HS, Ho DKN, Nguyen NN, Tran HM, Tam KW, Le NQK. Predicting EGFR Mutation Status in Non-Small Cell Lung Cancer Using Artificial Intelligence: A Systematic Review and Meta-Analysis. Acad Radiol 2024; 31:660-683. [PMID: 37120403 DOI: 10.1016/j.acra.2023.03.040] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 03/25/2023] [Accepted: 03/28/2023] [Indexed: 05/01/2023]
Abstract
RATIONALE AND OBJECTIVES Recent advancements in artificial intelligence (AI) render a substantial promise for epidermal growth factor receptor (EGFR) mutation status prediction in non-small cell lung cancer (NSCLC). We aimed to evaluate the performance and quality of AI algorithms that use radiomics features in predicting EGFR mutation status in patient with NSCLC. MATERIALS AND METHODS We searched PubMed (Medline), EMBASE, Web of Science, and IEEExplore for studies published up to February 28, 2022. Studies utilizing an AI algorithm (either conventional machine learning [cML] and deep learning [DL]) for predicting EGFR mutations in patients with NSLCL were included. We extracted binary diagnostic accuracy data and constructed a bivariate random-effects model to obtain pooled sensitivity, specificity, and 95% confidence interval. This study is registered with PROSPERO, CRD42021278738. RESULTS Our search identified 460 studies, of which 42 were included. Thirty-five studies were included in the meta-analysis. The AI algorithms exhibited an overall area under the curve (AUC) value of 0.789 and pooled sensitivity and specificity levels of 72.2% and 73.3%, respectively. The DL algorithms outperformed cML in terms of AUC (0.822 vs. 0.775) and sensitivity (80.1% vs. 71.1%), but had lower specificity (70.0% vs. 73.8%, p-value < 0.001) compared to cML. Subgroup analysis revealed that the use of positron-emission tomography/computed tomography, additional clinical information, deep feature extraction, and manual segmentation can improve diagnostic performance. CONCLUSION DL algorithms can serve as a novel method for increasing predictive accuracy and thus have considerable potential for use in predicting EGFR mutation status in patient with NSCLC. We also suggest that guidelines on using AI algorithms in medical image analysis should be developed with a focus on oncologic radiomics.
Collapse
Affiliation(s)
- Hung Song Nguyen
- International Ph.D. Program in Medicine, College of Medicine, Taipei Medical University, Taipei City, Taiwan (H.S.N., N.N.N.); Department of Pediatrics, Pham Ngoc Thach University of Medicine, Ho Chi Minh City, Viet Nam (H.S.N.); Intensive Care Unit Department, Children's Hospital 1, Ho Chi Minh City, Viet Nam (H.S.N.)
| | - Dang Khanh Ngan Ho
- School of Nutrition and Health Sciences, College of Nutrition, Taipei Medical University, Taipei, Taiwan (D.K.N.H.)
| | - Nam Nhat Nguyen
- International Ph.D. Program in Medicine, College of Medicine, Taipei Medical University, Taipei City, Taiwan (H.S.N., N.N.N.)
| | - Huy Minh Tran
- Department of Neurosurgery, Faculty of Medicine, University of Medicine and Pharmacy at Ho Chi Minh City, Ho Chi Minh City, Viet Nam (H.M.T.)
| | - Ka-Wai Tam
- Center for Evidence-based Health Care, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan (K.-W.T.); Cochrane Taiwan, Taipei Medical University, Taipei City, Taiwan (K.-W.T.); Division of General Surgery, Department of Surgery, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan (K.-W.T.); Division of General Surgery, Department of Surgery, School of Medicine, College of Medicine, Taipei Medical University, Taipei City, Taiwan (K.-W.T.)
| | - Nguyen Quoc Khanh Le
- Professional Master Program in Artificial Intelligence in Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan (N.Q.K.L.); Research Center for Artificial Intelligence in Medicine, Taipei Medical University, Taipei 110, Taiwan (N.Q.K.L.); AIBioMed Research Group, Taipei Medical University, Taipei 110, Taiwan (N.Q.K.L.); Translational Imaging Research Center, Taipei Medical University Hospital, Taipei 110, Taiwan (N.Q.K.L.).
| |
Collapse
|
23
|
Lu J, Ji X, Liu X, Jiang Y, Li G, Fang P, Li W, Zuo A, Guo Z, Yang S, Ji Y, Lu D. Machine learning-based radiomics strategy for prediction of acquired EGFR T790M mutation following treatment with EGFR-TKI in NSCLC. Sci Rep 2024; 14:446. [PMID: 38172228 PMCID: PMC10764785 DOI: 10.1038/s41598-023-50984-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2023] [Accepted: 12/28/2023] [Indexed: 01/05/2024] Open
Abstract
The epidermal growth factor receptor (EGFR) Thr790 Met (T790M) mutation is responsible for approximately half of the acquired resistance to EGFR-tyrosine kinase inhibitor (TKI) in non-small-cell lung cancer (NSCLC) patients. Identifying patients at diagnosis who are likely to develop this mutation after first- or second-generation EGFR-TKI treatment is crucial for better treatment outcomes. This study aims to develop and validate a radiomics-based machine learning (ML) approach to predict the T790M mutation in NSCLC patients at diagnosis. We collected retrospective data from 210 positive EGFR mutation NSCLC patients, extracting 1316 radiomics features from CT images. Using the LASSO algorithm, we selected 10 radiomics features and 2 clinical features most relevant to the mutations. We built models with 7 ML approaches and assessed their performance through the receiver operating characteristic (ROC) curve. The radiomics model and combined model, which integrated radiomics features and relevant clinical factors, achieved an area under the curve (AUC) of 0.80 (95% confidence interval [CI] 0.79-0.81) and 0.86 (0.87-0.88), respectively, in predicting the T790M mutation. Our study presents a convenient and noninvasive radiomics-based ML model for predicting this mutation at the time of diagnosis, aiding in targeted treatment planning for NSCLC patients with EGFR mutations.
Collapse
Affiliation(s)
- Jiameng Lu
- Department of Respiratory, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Shandong Institute of Respiratory Diseases, Shandong Institute of Anesthesia and Respiratory Critical Medicine, 16766 Jingshilu, Lixia, Jinan, 250014, Shandong, People's Republic of China
- School of Microelectronics, Shandong University, Jinan, 250100, Shandong, People's Republic of China
| | - Xiaoqing Ji
- Department of Nursing, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, 250014, Shandong, People's Republic of China
| | - Xinyi Liu
- Graduate School of Shandong First Medical University, Jinan, 250000, Shandong, People's Republic of China
| | - Yunxiu Jiang
- Graduate School of Shandong First Medical University, Jinan, 250000, Shandong, People's Republic of China
| | - Gang Li
- Department of Radiology, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Shandong Medicine and Health Key Laboratory of Abdominal Medicine Imaging, Shandong Lung Cancer Institute, Shandong Institute of Neuroimmunology, Jinan, 250000, Shandong, China
| | - Ping Fang
- Department of Blood Transfusion, The First Affiliated Hospital of Shandong First Medical University and Shandong Province Qianfoshan Hospital, Jinan, 250014, Shandong, China
| | - Wei Li
- Department of Radiology, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Shandong Medicine and Health Key Laboratory of Abdominal Medicine Imaging, Shandong Lung Cancer Institute, Shandong Institute of Neuroimmunology, Jinan, 250000, Shandong, China
| | - Anli Zuo
- Graduate School of Shandong First Medical University, Jinan, 250000, Shandong, People's Republic of China
| | - Zihan Guo
- Graduate School of Shandong First Medical University, Jinan, 250000, Shandong, People's Republic of China
| | - Shuran Yang
- Graduate School of Shandong First Medical University, Jinan, 250000, Shandong, People's Republic of China
| | - Yanbo Ji
- Department of Nursing, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, 250014, Shandong, People's Republic of China
| | - Degan Lu
- Department of Respiratory, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Shandong Institute of Respiratory Diseases, Shandong Institute of Anesthesia and Respiratory Critical Medicine, 16766 Jingshilu, Lixia, Jinan, 250014, Shandong, People's Republic of China.
| |
Collapse
|
24
|
Liu F, Xiang Z, Li Q, Fang X, Zhou J, Yang X, Lin H, Yang Q. 18F-FDG PET/CT-based radiomics model for predicting the degree of pathological differentiation in non-small cell lung cancer: a multicentre study. Clin Radiol 2024; 79:e147-e155. [PMID: 37884401 DOI: 10.1016/j.crad.2023.09.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 09/18/2023] [Accepted: 09/20/2023] [Indexed: 10/28/2023]
Abstract
AIM To explore the value of 2-[18F]-fluoro-2-deoxy-d-glucose (FDG) positron-emission tomography (PET)/computed tomography (CT)-based radiomics model for predicting the degree of pathological differentiation in non-small-cell lung cancer (NSCLC). MATERIALS AND METHODS Clinical characteristics of 182 NSCLC patients from four centres were collected, and radiomics features were extracted from 18F-FDG PET/CT images. Three logistic regression prediction models were established: clinical model; radiomics model; and nomogram combining radiomics signatures and clinical features. The predictive ability of the models was assessed using receiver operating characteristics curve analysis. RESULTS Patients from centre 1 were assigned randomly to the training and internal validation cohorts (7:3 ratio); patients from centres 2-4 served as the external validation cohort. The area under the curve (AUC) values for the clinical model in the training, internal validation, and external validation cohort were 0.74 (95% confidence interval [CI] = 0.64-0.84), 0.64 (95% CI = 0.46-0.81), and 0.74 (95% CI = 0.60-0.88), respectively. In the training (AUC: 0.84 [95% CI = 0.77-0.92]), internal validation (AUC: 0.81 [95% CI = 0.67-0.95]), and external validation cohorts (AUC: 0.74 [95% CI = 0.58-0.89]), the radiomics model showed good predictive ability for differentiation. Compared to the clinical and radiomics models, the nomogram has relatively better diagnostic performance, and the AUC values for nomogram in the training, internal validation, and external validation cohort were 0.86 (95% CI = 0.78-0.93), 0.83 (95% CI = 0.70-0.96), and 0.77 (95% CI = 0.62-0.92), respectively. CONCLUSIONS The 18F-FDG PET/CT-based radiomics model showed good ability for predicting the degree of differentiation of NSCLC. The nomogram combining the radiomics signature and clinical features has relatively better diagnostic performance.
Collapse
Affiliation(s)
- F Liu
- Department of Radiology, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Z Xiang
- Department of Radiology, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Q Li
- Department of Radiology, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - X Fang
- Department of Radiology, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China.
| | - J Zhou
- The Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu 212001, China
| | - X Yang
- Sichuan Science City Hospital, Mianyang, Sichuan 621000, China
| | - H Lin
- Department of Pharmaceutical Diagnosis, GE Healthcare, Changsha 410005, China
| | - Q Yang
- Center for Molecular Imaging Probe, Hunan Province Key Laboratory of Tumour Cellular and Molecular Pathology, Cancer Research Institute, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China.
| |
Collapse
|
25
|
Gong J, Fu F, Ma X, Wang T, Ma X, You C, Zhang Y, Peng W, Chen H, Gu Y. Hybrid deep multi-task learning radiomics approach for predicting EGFR mutation status of non-small cell lung cancer in CT images. Phys Med Biol 2023; 68:245021. [PMID: 37972417 DOI: 10.1088/1361-6560/ad0d43] [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: 07/09/2023] [Accepted: 11/15/2023] [Indexed: 11/19/2023]
Abstract
Objective.Epidermal growth factor receptor (EGFR) mutation genotyping plays a pivotal role in targeted therapy for non-small cell lung cancer (NSCLC). We aimed to develop a computed tomography (CT) image-based hybrid deep radiomics model to predict EGFR mutation status in NSCLC and investigate the correlations between deep image and quantitative radiomics features.Approach.First, we retrospectively enrolled 818 patients from our centre and 131 patients from The Cancer Imaging Archive database to establish a training cohort (N= 654), an independent internal validation cohort (N= 164) and an external validation cohort (N= 131). Second, to predict EGFR mutation status, we developed three CT image-based models, namely, a multi-task deep neural network (DNN), a radiomics model and a feature fusion model. Third, we proposed a hybrid loss function to train the DNN model. Finally, to evaluate the model performance, we computed the areas under the receiver operating characteristic curves (AUCs) and decision curve analysis curves of the models.Main results.For the two validation cohorts, the feature fusion model achieved AUC values of 0.86 ± 0.03 and 0.80 ± 0.05, which were significantly higher than those of the single-task DNN and radiomics models (allP< 0.05). There was no significant difference between the feature fusion and the multi-task DNN models (P> 0.8). The binary prediction scores showed excellent prognostic value in predicting disease-free survival (P= 0.02) and overall survival (P< 0.005) for validation cohort 2.Significance.The results demonstrate that (1) the feature fusion and multi-task DNN models achieve significantly higher performance than that of the conventional radiomics and single-task DNN models, (2) the feature fusion model can decode the imaging phenotypes representing NSCLC heterogeneity related to both EGFR mutation and patient NSCLC prognosis, and (3) high correlations exist between some deep image and radiomics features.
Collapse
Affiliation(s)
- Jing Gong
- Department of Radiology, Fudan University Shanghai Cancer Center, 270 Dongan Road, Shanghai, 20003, People's Republic of China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, People's Republic of China
| | - Fangqiu Fu
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, People's Republic of China
- Department of Thoracic Surgery and State key Laboratory of Genetic Engineering, Fudan University Shanghai Cancer Center, Shanghai, 200032, People's Republic of China
| | - Xiaowen Ma
- Department of Radiology, Fudan University Shanghai Cancer Center, 270 Dongan Road, Shanghai, 20003, People's Republic of China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, People's Republic of China
| | - Ting Wang
- Department of Radiology, Fudan University Shanghai Cancer Center, 270 Dongan Road, Shanghai, 20003, People's Republic of China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, People's Republic of China
| | - Xiangyi Ma
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, People's Republic of China
- Department of Thoracic Surgery and State key Laboratory of Genetic Engineering, Fudan University Shanghai Cancer Center, Shanghai, 200032, People's Republic of China
| | - Chao You
- Department of Radiology, Fudan University Shanghai Cancer Center, 270 Dongan Road, Shanghai, 20003, People's Republic of China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, People's Republic of China
| | - Yang Zhang
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, People's Republic of China
- Department of Thoracic Surgery and State key Laboratory of Genetic Engineering, Fudan University Shanghai Cancer Center, Shanghai, 200032, People's Republic of China
| | - Weijun Peng
- Department of Radiology, Fudan University Shanghai Cancer Center, 270 Dongan Road, Shanghai, 20003, People's Republic of China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, People's Republic of China
| | - Haiquan Chen
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, People's Republic of China
- Department of Thoracic Surgery and State key Laboratory of Genetic Engineering, Fudan University Shanghai Cancer Center, Shanghai, 200032, People's Republic of China
| | - Yajia Gu
- Department of Radiology, Fudan University Shanghai Cancer Center, 270 Dongan Road, Shanghai, 20003, People's Republic of China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, People's Republic of China
| |
Collapse
|
26
|
Çalışkan M, Tazaki K. AI/ML advances in non-small cell lung cancer biomarker discovery. Front Oncol 2023; 13:1260374. [PMID: 38148837 PMCID: PMC10750392 DOI: 10.3389/fonc.2023.1260374] [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: 07/17/2023] [Accepted: 11/16/2023] [Indexed: 12/28/2023] Open
Abstract
Lung cancer is the leading cause of cancer deaths among both men and women, representing approximately 25% of cancer fatalities each year. The treatment landscape for non-small cell lung cancer (NSCLC) is rapidly evolving due to the progress made in biomarker-driven targeted therapies. While advancements in targeted treatments have improved survival rates for NSCLC patients with actionable biomarkers, long-term survival remains low, with an overall 5-year relative survival rate below 20%. Artificial intelligence/machine learning (AI/ML) algorithms have shown promise in biomarker discovery, yet NSCLC-specific studies capturing the clinical challenges targeted and emerging patterns identified using AI/ML approaches are lacking. Here, we employed a text-mining approach and identified 215 studies that reported potential biomarkers of NSCLC using AI/ML algorithms. We catalogued these studies with respect to BEST (Biomarkers, EndpointS, and other Tools) biomarker sub-types and summarized emerging patterns and trends in AI/ML-driven NSCLC biomarker discovery. We anticipate that our comprehensive review will contribute to the current understanding of AI/ML advances in NSCLC biomarker research and provide an important catalogue that may facilitate clinical adoption of AI/ML-derived biomarkers.
Collapse
Affiliation(s)
- Minal Çalışkan
- Translational Science Department, Precision Medicine Function, Daiichi Sankyo, Inc., Basking Ridge, NJ, United States
| | - Koichi Tazaki
- Translational Science Department I, Precision Medicine Function, Daiichi Sankyo, Tokyo, Japan
| |
Collapse
|
27
|
Shang Y, Chen W, Li G, Huang Y, Wang Y, Kui X, Li M, Zheng H, Zhao W, Liu J. Computed Tomography-derived intratumoral and peritumoral radiomics in predicting EGFR mutation in lung adenocarcinoma. LA RADIOLOGIA MEDICA 2023; 128:1483-1496. [PMID: 37749461 PMCID: PMC10700425 DOI: 10.1007/s11547-023-01722-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/15/2023] [Accepted: 09/04/2023] [Indexed: 09/27/2023]
Abstract
OBJECTIVE To investigate the value of Computed Tomography (CT) radiomics derived from different peritumoral volumes of interest (VOIs) in predicting epidermal growth factor receptor (EGFR) mutation status in lung adenocarcinoma patients. MATERIALS AND METHODS A retrospective cohort of 779 patients who had pathologically confirmed lung adenocarcinoma were enrolled. 640 patients were randomly divided into a training set, a validation set, and an internal testing set (3:1:1), and the remaining 139 patients were defined as an external testing set. The intratumoral VOI (VOI_I) was manually delineated on the thin-slice CT images, and seven peritumoral VOIs (VOI_P) were automatically generated with 1, 2, 3, 4, 5, 10, and 15 mm expansion along the VOI_I. 1454 radiomic features were extracted from each VOI. The t-test, the least absolute shrinkage and selection operator (LASSO), and the minimum redundancy maximum relevance (mRMR) algorithm were used for feature selection, followed by the construction of radiomics models (VOI_I model, VOI_P model and combined model). The performance of the models were evaluated by the area under the curve (AUC). RESULTS 399 patients were classified as EGFR mutant (EGFR+), while 380 were wild-type (EGFR-). In the training and validation sets, internal and external testing sets, VOI4 (intratumoral and peritumoral 4 mm) model achieved the best predictive performance, with AUCs of 0.877, 0.727, and 0.701, respectively, outperforming the VOI_I model (AUCs of 0.728, 0.698, and 0.653, respectively). CONCLUSIONS Radiomics extracted from peritumoral region can add extra value in predicting EGFR mutation status of lung adenocarcinoma patients, with the optimal peritumoral range of 4 mm.
Collapse
Affiliation(s)
- Youlan Shang
- Department of Radiology, The Second Xiangya Hospital, Central South University, No. 139 Middle Renmin Road, Changsha, 410011, Hunan, People's Republic of China
| | - Weidao Chen
- Infervision, Chaoyang District, Beijing, 100025, China
| | - Ge Li
- Department of Radiology, Xiangya Hospital, Central South University, No. 87 Xiangya Rd, Changsha, 410008, Hunan, People's Republic of China
| | - Yijie Huang
- Department of Radiology, The Second Xiangya Hospital, Central South University, No. 139 Middle Renmin Road, Changsha, 410011, Hunan, People's Republic of China
| | - Yisong Wang
- Department of Radiology, The Second Xiangya Hospital, Central South University, No. 139 Middle Renmin Road, Changsha, 410011, Hunan, People's Republic of China
| | - Xiaoyan Kui
- School of Computer Science and Engineering, Central South University, Changsha, 410083, Hunan, People's Republic of China
| | - Ming Li
- Department of Radiology, Huadong Hospital Affiliated to Fudan University, Shanghai, People's Republic of China
| | - Hairong Zheng
- Paul C. Lauterbur Research Center for Biomedical Imaging, Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, People's Republic of China
| | - Wei Zhao
- Department of Radiology, The Second Xiangya Hospital, Central South University, No. 139 Middle Renmin Road, Changsha, 410011, Hunan, People's Republic of China.
- Paul C. Lauterbur Research Center for Biomedical Imaging, Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, People's Republic of China.
- Clinical Research Center for Medical Imaging in Hunan Province, Changsha, Hunan Province, People's Republic of China.
| | - Jun Liu
- Department of Radiology, The Second Xiangya Hospital, Central South University, No. 139 Middle Renmin Road, Changsha, 410011, Hunan, People's Republic of China.
- Clinical Research Center for Medical Imaging in Hunan Province, Changsha, Hunan Province, People's Republic of China.
| |
Collapse
|
28
|
Ma T, Zhang Y, Zhao M, Wang L, Wang H, Ye Z. A machine learning-based radiomics model for prediction of tumor mutation burden in gastric cancer. Front Genet 2023; 14:1283090. [PMID: 38028587 PMCID: PMC10657897 DOI: 10.3389/fgene.2023.1283090] [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: 08/25/2023] [Accepted: 10/23/2023] [Indexed: 12/01/2023] Open
Abstract
Purpose: To evaluate the potential of machine learning (ML)-based radiomics approach for predicting tumor mutation burden (TMB) in gastric cancer (GC). Methods: The contrast enhanced CT (CECT) images with corresponding clinical information of 256 GC patients were retrospectively collected. Patients were separated into training set (n = 180) and validation set (n = 76). A total of 3,390 radiomics features were extracted from three phases images of CECT. The least absolute shrinkage and selection operator (LASSO) model was used for feature screening. Seven machine learning (ML) algorithms were employed to find the optimal classifier. The predictive ability of radiomics model (RM) was evaluated with receiver operating characteristic. The correlation between RM and TMB values was evaluated using Spearman's correlation coefficient. The explainability of RM was assessed by the Shapley Additive explanations (SHAP) method. Results: Logistic regression algorithm was chosen for model construction. The RM showed good predictive ability of TMB status with AUCs of 0.89 [95% confidence interval (CI): 0.85-0.94] and 0.86 (95% CI: 0.74-0.98) in the training and validation sets. The correlation analysis revealed a good correlation between RM and TMB levels (correlation coefficient: 0.62, p < 0.001). The RM also showed favorable and stable predictive accuracy within the cutoff value range 6-16 mut/Mb in both sets. Conclusion: The ML-based RM offered a promising image biomarker for predicting TMB status in GC patients.
Collapse
Affiliation(s)
- Tingting Ma
- Department of Radiology, Tianjin Cancer Hospital Airport Hospital, Tianjin, China
- Department of Radiology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- National Clinical Research Center for Cancer, Tianjin, China
- Tianjin’s Clinical Research Center for Cancer, Tianjin, China
- The Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - Yuwei Zhang
- Department of Radiology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- National Clinical Research Center for Cancer, Tianjin, China
- Tianjin’s Clinical Research Center for Cancer, Tianjin, China
- The Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - Mengran Zhao
- Department of Radiology, Tianjin Cancer Hospital Airport Hospital, Tianjin, China
- Department of Radiology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- National Clinical Research Center for Cancer, Tianjin, China
- Tianjin’s Clinical Research Center for Cancer, Tianjin, China
- The Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - Lingwei Wang
- Department of Radiology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- National Clinical Research Center for Cancer, Tianjin, China
- Tianjin’s Clinical Research Center for Cancer, Tianjin, China
- The Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - Hua Wang
- Department of Radiology, Tianjin Cancer Hospital Airport Hospital, Tianjin, China
- Department of Radiology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- National Clinical Research Center for Cancer, Tianjin, China
- Tianjin’s Clinical Research Center for Cancer, Tianjin, China
- The Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - Zhaoxiang Ye
- Department of Radiology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- National Clinical Research Center for Cancer, Tianjin, China
- Tianjin’s Clinical Research Center for Cancer, Tianjin, China
- The Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| |
Collapse
|
29
|
Tang X, Li Y, Shen LT, Yan WF, Qian WL, Yang ZG. CT Radiomics Predict EGFR-T790M Resistance Mutation in Advanced Non-Small Cell Lung Cancer Patients After Progression on First-line EGFR-TKI. Acad Radiol 2023; 30:2574-2587. [PMID: 36941156 DOI: 10.1016/j.acra.2023.01.040] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 01/25/2023] [Accepted: 01/31/2023] [Indexed: 03/23/2023]
Abstract
RATIONALE AND OBJECTIVES We aim to explore the value of chest CT radiomics in predicting the epidermal growth factor receptor (EGFR)-T790M resistance mutation of advanced non-small cell lung cancer (NSCLC) patients after the failure of first-line EGFR-tyrosine kinase inhibitor (EGFR-TKI). MATERIALS AND METHODS A total of 211 and 135 advanced NSCLC patients with tumor tissue-based (Cohort-1) or circulating tumor DNA (ctDNA)-based (Cohort-2) EGFR-T790M testing were included, respectively. Cohort-1 was used for modeling and Cohort-2 was for models' validation. Radiomic features were extracted from tumor lesions on chest nonenhanced CT (NECT) and/or contrast-enhanced CT (CECT). We used eight feature selectors and eight classifier algorithms to establish radiomic models. Models were evaluated by area under the receiver operating characteristic curve (AUC), calibration curve, and decision curve analysis (DCA). RESULTS CT morphological manifestations of peripheral location and pleural indentation sign were associated with EGFR-T790M. For NECT, CECT, and NECT+CECT radiomic features, the feature selector and classifier algorithms of LASSO and Stepwise logistic regression, Boruta and SVM, and LASSO and SVM were chosen to develop the optimal model, respectively (AUC: 0.844, 0.811, and 0.897). All models performed well in calibration curves and DCA. Independent validation of models in Cohort-2 revealed that both NECT and CECT models individually had limited power for predicting EGFR-T790M mutation detected by ctDNA (AUC: 0.649, 0.675), while the NECT+CECT radiomic model had a satisfactory AUC (0.760). CONCLUSION This study proved the feasibility of using CT radiomic features to predict the EGFR-T790M resistance mutation, which could be helpful in guiding personalized therapeutic strategies.
Collapse
Affiliation(s)
- Xin Tang
- Department of Radiology, Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yuan Li
- Department of Radiology, Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Li-Ting Shen
- Department of Radiology, Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Wei-Feng Yan
- Department of Radiology, Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Wen-Lei Qian
- Department of Radiology, Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Zhi-Gang Yang
- Department of Radiology, Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
| |
Collapse
|
30
|
Rinaldi L, Guerini Rocco E, Spitaleri G, Raimondi S, Attili I, Ranghiero A, Cammarata G, Minotti M, Lo Presti G, De Piano F, Bellerba F, Funicelli G, Volpe S, Mora S, Fodor C, Rampinelli C, Barberis M, De Marinis F, Jereczek-Fossa BA, Orecchia R, Rizzo S, Botta F. Association between Contrast-Enhanced Computed Tomography Radiomic Features, Genomic Alterations and Prognosis in Advanced Lung Adenocarcinoma Patients. Cancers (Basel) 2023; 15:4553. [PMID: 37760521 PMCID: PMC10527057 DOI: 10.3390/cancers15184553] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Revised: 09/11/2023] [Accepted: 09/12/2023] [Indexed: 09/29/2023] Open
Abstract
Non-invasive methods to assess mutational status, as well as novel prognostic biomarkers, are warranted to foster therapy personalization of patients with advanced non-small cell lung cancer (NSCLC). This study investigated the association of contrast-enhanced Computed Tomography (CT) radiomic features of lung adenocarcinoma lesions, alone or integrated with clinical parameters, with tumor mutational status (EGFR, KRAS, ALK alterations) and Overall Survival (OS). In total, 261 retrospective and 48 prospective patients were enrolled. A Radiomic Score (RS) was created with LASSO-Logistic regression models to predict mutational status. Radiomic, clinical and clinical-radiomic models were trained on retrospective data and tested (Area Under the Curve, AUC) on prospective data. OS prediction models were trained and tested on retrospective data with internal cross-validation (C-index). RS significantly predicted each alteration at training (radiomic and clinical-radiomic AUC 0.95-0.98); validation performance was good for EGFR (AUC 0.86), moderate for KRAS and ALK (AUC 0.61-0.65). RS was also associated with OS at univariate and multivariable analysis, in the latter with stage and type of treatment. The validation C-index was 0.63, 0.79, and 0.80 for clinical, radiomic, and clinical-radiomic models. The study supports the potential role of CT radiomics for non-invasive identification of gene alterations and prognosis prediction in patients with advanced lung adenocarcinoma, to be confirmed with independent studies.
Collapse
Affiliation(s)
- Lisa Rinaldi
- Radiation Research Unit, IEO European Institute of Oncology IRCCS, Via Ripamonti 435, 20141 Milan, Italy;
| | - Elena Guerini Rocco
- Division of Pathology, European Institute of Oncology IRCCS, 20141 Milan, Italy; (E.G.R.); (A.R.); (M.B.)
- Department of Oncology and Hemato-Oncology, University of Milan, Via Festa del Perdono 7, 20122 Milan, Italy; (S.V.)
| | - Gianluca Spitaleri
- Division of Thoracic Oncology, IEO European Institute of Oncology IRCCS, Via Ripamonti 435, 20141 Milan, Italy; (G.S.); (I.A.); (F.D.M.)
| | - Sara Raimondi
- Department of Experimental Oncology, IEO European Institute of Oncology IRCCS, Via Ripamonti 435, 20141 Milan, Italy (F.B.)
| | - Ilaria Attili
- Division of Thoracic Oncology, IEO European Institute of Oncology IRCCS, Via Ripamonti 435, 20141 Milan, Italy; (G.S.); (I.A.); (F.D.M.)
| | - Alberto Ranghiero
- Division of Pathology, European Institute of Oncology IRCCS, 20141 Milan, Italy; (E.G.R.); (A.R.); (M.B.)
| | - Giulio Cammarata
- Department of Experimental Oncology, IEO European Institute of Oncology IRCCS, Via Ripamonti 435, 20141 Milan, Italy (F.B.)
| | - Marta Minotti
- Division of Radiology, IEO European Institute of Oncology IRCCS, Via Ripamonti 435, 20141 Milan, Italy; (M.M.); (C.R.); (R.O.)
| | - Giuliana Lo Presti
- Department of Experimental Oncology, IEO European Institute of Oncology IRCCS, Via Ripamonti 435, 20141 Milan, Italy (F.B.)
| | - Francesca De Piano
- Division of Radiology, IEO European Institute of Oncology IRCCS, Via Ripamonti 435, 20141 Milan, Italy; (M.M.); (C.R.); (R.O.)
| | - Federica Bellerba
- Department of Experimental Oncology, IEO European Institute of Oncology IRCCS, Via Ripamonti 435, 20141 Milan, Italy (F.B.)
| | - Gianluigi Funicelli
- Division of Radiology, IEO European Institute of Oncology IRCCS, Via Ripamonti 435, 20141 Milan, Italy; (M.M.); (C.R.); (R.O.)
| | - Stefania Volpe
- Department of Oncology and Hemato-Oncology, University of Milan, Via Festa del Perdono 7, 20122 Milan, Italy; (S.V.)
- Department of Radiation Oncology, IEO European Institute of Oncology IRCCS, Via Ripamonti 435, 20141 Milan, Italy
| | - Serena Mora
- Data Management Unit, IEO European Institute of Oncology IRCCS, Via Ripamonti 435, 20141 Milan, Italy; (S.M.); (C.F.)
| | - Cristiana Fodor
- Data Management Unit, IEO European Institute of Oncology IRCCS, Via Ripamonti 435, 20141 Milan, Italy; (S.M.); (C.F.)
| | - Cristiano Rampinelli
- Division of Radiology, IEO European Institute of Oncology IRCCS, Via Ripamonti 435, 20141 Milan, Italy; (M.M.); (C.R.); (R.O.)
| | - Massimo Barberis
- Division of Pathology, European Institute of Oncology IRCCS, 20141 Milan, Italy; (E.G.R.); (A.R.); (M.B.)
| | - Filippo De Marinis
- Division of Thoracic Oncology, IEO European Institute of Oncology IRCCS, Via Ripamonti 435, 20141 Milan, Italy; (G.S.); (I.A.); (F.D.M.)
| | - Barbara Alicja Jereczek-Fossa
- Department of Oncology and Hemato-Oncology, University of Milan, Via Festa del Perdono 7, 20122 Milan, Italy; (S.V.)
- Department of Radiation Oncology, IEO European Institute of Oncology IRCCS, Via Ripamonti 435, 20141 Milan, Italy
| | - Roberto Orecchia
- Division of Radiology, IEO European Institute of Oncology IRCCS, Via Ripamonti 435, 20141 Milan, Italy; (M.M.); (C.R.); (R.O.)
- Scientific Direction, IEO European Institute of Oncology IRCCS, Via Ripamonti 435, 20141 Milan, Italy
| | - Stefania Rizzo
- Clinica di Radiologia EOC, Istituto Imaging della Svizzera Italiana (IIMSI), Via Tesserete 46, 6900 Lugano, Switzerland;
- Faculty of Biomedical Sciences, Università della Svizzera italiana, Via G. Buffi 13, 6900 Lugano, Switzerland
| | - Francesca Botta
- Medical Physics Unit, IEO European Institute of Oncology IRCCS, Via Ripamonti 435, 20141 Milan, Italy;
| |
Collapse
|
31
|
Lin P, Lin YQ, Gao RZ, Wan WJ, He Y, Yang H. Integrative radiomics and transcriptomics analyses reveal subtype characterization of non-small cell lung cancer. Eur Radiol 2023; 33:6414-6425. [PMID: 36826501 DOI: 10.1007/s00330-023-09503-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 12/15/2022] [Accepted: 01/30/2023] [Indexed: 02/25/2023]
Abstract
OBJECTIVES To assess whether integrative radiomics and transcriptomics analyses could provide novel insights for radiomic features' molecular annotation and effective risk stratification in non-small cell lung cancer (NSCLC). METHODS A total of 627 NSCLC patients from three datasets were included. Radiomics features were extracted from segmented 3-dimensional tumour volumes and were z-score normalized for further analysis. In transcriptomics level, 186 pathways and 28 types of immune cells were assessed by using the Gene Set Variation Analysis (GSVA) algorithm. NSCLC patients were categorized into subgroups based on their radiomic features and pathways enrichment scores using consensus clustering. Subgroup-specific radiomics features were used to validate clustering performance and prognostic value. Kaplan-Meier survival analysis with the log-rank test and univariable and multivariable Cox analyses were conducted to explore survival differences among the subgroups. RESULTS Three radiotranscriptomics subtypes (RTSs) were identified based on the radiomics and pathways enrichment profiles. The three RTSs were characterized as having specific molecular hallmarks: RTS1 (proliferation subtype), RTS2 (metabolism subtype), and RTS3 (immune activation subtype). RTS3 showed increased infiltration of most immune cells. The RTS stratification strategy was validated in a validation cohort and showed significant prognostic value. Survival analysis demonstrated that the RTS strategy could stratify NSCLC patients according to prognosis (p = 0.009), and the RTS strategy remained an independent prognostic indicator after adjusting for other clinical parameters. CONCLUSIONS This radiotranscriptomics study provides a stratification strategy for NSCLC that could provide information for radiomics feature molecular annotation and prognostic prediction. KEY POINTS • Radiotranscriptomics subtypes (RTSs) could be used to stratify molecularly heterogeneous patients. • RTSs showed relationships between molecular phenotypes and radiomics features. • The RTS algorithm could be used to identify patients with poor prognosis.
Collapse
Affiliation(s)
- Peng Lin
- Department of Medical Ultrasound, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, People's Republic of China
| | - Yi-Qun Lin
- Department of Radiology, The 909th Hospital. School of Medicine, Xiamen University, Fujian, Zhangzhou, People's Republic of China
| | - Rui-Zhi Gao
- Department of Medical Ultrasound, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, People's Republic of China
| | - Wei-Jun Wan
- Department of Medical Ultrasound, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, People's Republic of China
| | - Yun He
- Department of Medical Ultrasound, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, People's Republic of China.
| | - Hong Yang
- Department of Medical Ultrasound, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, People's Republic of China.
| |
Collapse
|
32
|
Shao J, Feng J, Li J, Liang S, Li W, Wang C. Novel tools for early diagnosis and precision treatment based on artificial intelligence. CHINESE MEDICAL JOURNAL PULMONARY AND CRITICAL CARE MEDICINE 2023; 1:148-160. [PMID: 39171128 PMCID: PMC11332840 DOI: 10.1016/j.pccm.2023.05.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Indexed: 08/23/2024]
Abstract
Lung cancer has the highest mortality rate among all cancers in the world. Hence, early diagnosis and personalized treatment plans are crucial to improving its 5-year survival rate. Chest computed tomography (CT) serves as an essential tool for lung cancer screening, and pathology images are the gold standard for lung cancer diagnosis. However, medical image evaluation relies on manual labor and suffers from missed diagnosis or misdiagnosis, and physician heterogeneity. The rapid development of artificial intelligence (AI) has brought a whole novel opportunity for medical task processing, demonstrating the potential for clinical application in lung cancer diagnosis and treatment. AI technologies, including machine learning and deep learning, have been deployed extensively for lung nodule detection, benign and malignant classification, and subtype identification based on CT images. Furthermore, AI plays a role in the non-invasive prediction of genetic mutations and molecular status to provide the optimal treatment regimen, and applies to the assessment of therapeutic efficacy and prognosis of lung cancer patients, enabling precision medicine to become a reality. Meanwhile, histology-based AI models assist pathologists in typing, molecular characterization, and prognosis prediction to enhance the efficiency of diagnosis and treatment. However, the leap to extensive clinical application still faces various challenges, such as data sharing, standardized label acquisition, clinical application regulation, and multimodal integration. Nevertheless, AI holds promising potential in the field of lung cancer to improve cancer care.
Collapse
Affiliation(s)
- Jun Shao
- Department of Pulmonary and Critical Care Medicine, Med-X Center for Manufacturing, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Jiaming Feng
- West China School of Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Jingwei Li
- Department of Pulmonary and Critical Care Medicine, Med-X Center for Manufacturing, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Shufan Liang
- Department of Pulmonary and Critical Care Medicine, Med-X Center for Manufacturing, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Weimin Li
- Department of Pulmonary and Critical Care Medicine, Med-X Center for Manufacturing, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Chengdi Wang
- Department of Pulmonary and Critical Care Medicine, Med-X Center for Manufacturing, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| |
Collapse
|
33
|
Chen M, Copley SJ, Viola P, Lu H, Aboagye EO. Radiomics and artificial intelligence for precision medicine in lung cancer treatment. Semin Cancer Biol 2023; 93:97-113. [PMID: 37211292 DOI: 10.1016/j.semcancer.2023.05.004] [Citation(s) in RCA: 39] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 04/14/2023] [Accepted: 05/17/2023] [Indexed: 05/23/2023]
Abstract
Lung cancer is the leading cause of cancer-related deaths worldwide. It exhibits, at the mesoscopic scale, phenotypic characteristics that are generally indiscernible to the human eye but can be captured non-invasively on medical imaging as radiomic features, which can form a high dimensional data space amenable to machine learning. Radiomic features can be harnessed and used in an artificial intelligence paradigm to risk stratify patients, and predict for histological and molecular findings, and clinical outcome measures, thereby facilitating precision medicine for improving patient care. Compared to tissue sampling-driven approaches, radiomics-based methods are superior for being non-invasive, reproducible, cheaper, and less susceptible to intra-tumoral heterogeneity. This review focuses on the application of radiomics, combined with artificial intelligence, for delivering precision medicine in lung cancer treatment, with discussion centered on pioneering and groundbreaking works, and future research directions in the area.
Collapse
Affiliation(s)
- Mitchell Chen
- Department of Surgery and Cancer, The Commonwealth Building, Du Cane Road, Hammersmith Campus, Imperial College, London W12 0NN, UK; Imperial College Healthcare NHS Trust, Hammersmith Hospital, Du Cane Road, London W12 0HS, UK
| | - Susan J Copley
- Department of Surgery and Cancer, The Commonwealth Building, Du Cane Road, Hammersmith Campus, Imperial College, London W12 0NN, UK; Imperial College Healthcare NHS Trust, Hammersmith Hospital, Du Cane Road, London W12 0HS, UK
| | - Patrizia Viola
- North West London Pathology, Charing Cross Hospital, Fulham Palace Rd, London W6 8RF, UK
| | - Haonan Lu
- Department of Surgery and Cancer, The Commonwealth Building, Du Cane Road, Hammersmith Campus, Imperial College, London W12 0NN, UK
| | - Eric O Aboagye
- Department of Surgery and Cancer, The Commonwealth Building, Du Cane Road, Hammersmith Campus, Imperial College, London W12 0NN, UK.
| |
Collapse
|
34
|
Felfli M, Liu Y, Zerka F, Voyton C, Thinnes A, Jacques S, Iannessi A, Bodard S. Systematic Review, Meta-Analysis and Radiomics Quality Score Assessment of CT Radiomics-Based Models Predicting Tumor EGFR Mutation Status in Patients with Non-Small-Cell Lung Cancer. Int J Mol Sci 2023; 24:11433. [PMID: 37511192 PMCID: PMC10380456 DOI: 10.3390/ijms241411433] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2023] [Revised: 07/11/2023] [Accepted: 07/12/2023] [Indexed: 07/30/2023] Open
Abstract
Assessment of the quality and current performance of computed tomography (CT) radiomics-based models in predicting epidermal growth factor receptor (EGFR) mutation status in patients with non-small-cell lung carcinoma (NSCLC). Two medical literature databases were systematically searched, and articles presenting original studies on CT radiomics-based models for predicting EGFR mutation status were retrieved. Forest plots and related statistical tests were performed to summarize the model performance and inter-study heterogeneity. The methodological quality of the selected studies was assessed via the Radiomics Quality Score (RQS). The performance of the models was evaluated using the area under the curve (ROC AUC). The range of the Risk RQS across the selected articles varied from 11 to 24, indicating a notable heterogeneity in the quality and methodology of the included studies. The average score was 15.25, which accounted for 42.34% of the maximum possible score. The pooled Area Under the Curve (AUC) value was 0.801, indicating the accuracy of CT radiomics-based models in predicting the EGFR mutation status. CT radiomics-based models show promising results as non-invasive alternatives for predicting EGFR mutation status in NSCLC patients. However, the quality of the studies using CT radiomics-based models varies widely, and further harmonization and prospective validation are needed before the generalization of these models.
Collapse
Affiliation(s)
- Mehdi Felfli
- Median Technologies, F-06560 Valbonne, France; (M.F.); (Y.L.); (F.Z.); (C.V.); (A.T.); (S.J.); (A.I.)
| | - Yan Liu
- Median Technologies, F-06560 Valbonne, France; (M.F.); (Y.L.); (F.Z.); (C.V.); (A.T.); (S.J.); (A.I.)
| | - Fadila Zerka
- Median Technologies, F-06560 Valbonne, France; (M.F.); (Y.L.); (F.Z.); (C.V.); (A.T.); (S.J.); (A.I.)
| | - Charles Voyton
- Median Technologies, F-06560 Valbonne, France; (M.F.); (Y.L.); (F.Z.); (C.V.); (A.T.); (S.J.); (A.I.)
| | - Alexandre Thinnes
- Median Technologies, F-06560 Valbonne, France; (M.F.); (Y.L.); (F.Z.); (C.V.); (A.T.); (S.J.); (A.I.)
| | - Sebastien Jacques
- Median Technologies, F-06560 Valbonne, France; (M.F.); (Y.L.); (F.Z.); (C.V.); (A.T.); (S.J.); (A.I.)
| | - Antoine Iannessi
- Median Technologies, F-06560 Valbonne, France; (M.F.); (Y.L.); (F.Z.); (C.V.); (A.T.); (S.J.); (A.I.)
- Centre Antoine Lacassagne, F-06100 Nice, France
| | - Sylvain Bodard
- AP-HP, Service d’Imagerie Adulte, Hôpital Necker Enfants Malades, Université de Paris Cité, F-75015 Paris, France
- CNRS UMR 7371, INSERM U 1146, Laboratoire d’Imagerie Biomédicale, Sorbonne Université, F-75006 Paris, France
| |
Collapse
|
35
|
Genova C. The Long Run towards Personalized Therapy in Non-Small-Cell Lung Cancer: Current State and Future Directions. Int J Mol Sci 2023; 24:ijms24098212. [PMID: 37175919 PMCID: PMC10178998 DOI: 10.3390/ijms24098212] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Accepted: 04/26/2023] [Indexed: 05/15/2023] Open
Abstract
Non-small-cell lung cancer (NSCLC) is the major cause of cancer-related deaths worldwide, due to its high incidence and mortality [...].
Collapse
Affiliation(s)
- Carlo Genova
- Dipartimento di Medicina Interna e Specialità Mediche, Università degli Studi di Genova, 16132 Genova, Italy
- UO Clinica di Oncologia Medica, IRCCS Ospedale Policlinico San Martino, 16132 Genova, Italy
| |
Collapse
|
36
|
Zhang Z, Wei X. Artificial intelligence-assisted selection and efficacy prediction of antineoplastic strategies for precision cancer therapy. Semin Cancer Biol 2023; 90:57-72. [PMID: 36796530 DOI: 10.1016/j.semcancer.2023.02.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 01/12/2023] [Accepted: 02/13/2023] [Indexed: 02/16/2023]
Abstract
The rapid development of artificial intelligence (AI) technologies in the context of the vast amount of collectable data obtained from high-throughput sequencing has led to an unprecedented understanding of cancer and accelerated the advent of a new era of clinical oncology with a tone of precision treatment and personalized medicine. However, the gains achieved by a variety of AI models in clinical oncology practice are far from what one would expect, and in particular, there are still many uncertainties in the selection of clinical treatment options that pose significant challenges to the application of AI in clinical oncology. In this review, we summarize emerging approaches, relevant datasets and open-source software of AI and show how to integrate them to address problems from clinical oncology and cancer research. We focus on the principles and procedures for identifying different antitumor strategies with the assistance of AI, including targeted cancer therapy, conventional cancer therapy, and cancer immunotherapy. In addition, we also highlight the current challenges and directions of AI in clinical oncology translation. Overall, we hope this article will provide researchers and clinicians with a deeper understanding of the role and implications of AI in precision cancer therapy, and help AI move more quickly into accepted cancer guidelines.
Collapse
Affiliation(s)
- Zhe Zhang
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610041, PR China; State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu 610041, PR China
| | - Xiawei Wei
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610041, PR China.
| |
Collapse
|
37
|
Zhou C, Hou L, Tang X, Liu C, Meng Y, Jia H, Yang H, Zhou S. CT-based radiomics nomogram may predict who can benefit from adaptive radiotherapy in patients with local advanced-NSCLC patients. Radiother Oncol 2023; 183:109637. [PMID: 36963440 DOI: 10.1016/j.radonc.2023.109637] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 02/14/2023] [Accepted: 03/17/2023] [Indexed: 03/26/2023]
Abstract
BACKGROUND Although adaptive radiotherapy (ART) has many advantages, ART is not universal in the clinical appliance due to the consumption of a lot of labor, and economic burden. It is necessary to explore a CT stimulation-based radiomics model for screening who can get more benefits from ART in locally advanced non-small cell lung cancer (NSCLC) patients. METHOD 183 cases of NSCLC patients receiving concurrent chemoradiotherapy with an adaptive approach were enrolled as a primary cohort, while 28 cases from another hospital served as an independent external validation cohort. Tumor regression assessment was conducted based on GTV reduction (Criteria A) or according to RECIST Version 1.1(Criteria B). The radiomics features were extracted by the "PyRadiomics" package and further screened by the LASSO method. Then, logistic regression was used to establish the model. Bootstrap and external validation were applied to verify the stability of the model. The receiver operating characteristic (ROC) curve was delineated to assess the predictive efficacy of the radiomics model. Dose-volume histograms were quantitatively compared between the initial and composite ART plans. Clinical endpoints included overall survival (OS) and progression-free survival (PFS). RESULT There were no significant differences in clinical features between tumor regression-resistant (RR) and tumor regression-sensitivity (RS) groups. The AUC values of the Criteria A model and Criteria B model were 0.767 and 0.771, respectively. Bootstrapping validation and external validation confirmed the stability of models. In all patients, there was a significant benefit of ART in the lung, heart, cord, and esophagus compared to non-ART, particularly in RS patients. Furthermore, PFS and OS from ART were significantly longer in RS as defined by Criterion B than in RR patients with the same ART application. CONCLUSION CT-based radiomics can screen out the patients who can gain more benefits from ART, which contribute to guiding and popularizing the application of ART strategy in the clinic within economic benefits and feasibility.
Collapse
Affiliation(s)
- Chao Zhou
- From Department of Radiation Oncology, Taizhou Hospital Affiliated to Wenzhou Medical University, Zhejiang Province 317000, China
| | - Liqiao Hou
- From Department of Radiation Oncology, Taizhou Hospital Affiliated to Wenzhou Medical University, Zhejiang Province 317000, China
| | - Xingni Tang
- From Department of Radiation Oncology, Taizhou Hospital Affiliated to Wenzhou Medical University, Zhejiang Province 317000, China
| | - Changxing Liu
- Department of Epidemiology and Biostatistics, School of Public Health, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, China
| | - Yinnan Meng
- From Department of Radiation Oncology, Taizhou Hospital Affiliated to Wenzhou Medical University, Zhejiang Province 317000, China
| | - Haijian Jia
- Department of Radiation Oncology, Enze Hospital Affiliated Hospital of Hangzhou Medical College, Zhejiang Province 317000, China
| | - Haihua Yang
- Department of Radiation Oncology, Xi'an No.3 Hospital, the Affiliated Hospital of Northwest University, Xi'an, Shaanxi 710018, P.R. China.
| | - Suna Zhou
- From Department of Radiation Oncology, Taizhou Hospital Affiliated to Wenzhou Medical University, Zhejiang Province 317000, China; Department of Radiation Oncology, Xi'an No.3 Hospital, the Affiliated Hospital of Northwest University, Xi'an, Shaanxi 710018, P.R. China.
| |
Collapse
|
38
|
Chen C, Geng Q, Song G, Zhang Q, Wang Y, Sun D, Zeng Q, Dai Z, Wang G. A comprehensive nomogram combining CT-based radiomics with clinical features for differentiation of benign and malignant lung subcentimeter solid nodules. Front Oncol 2023; 13:1066360. [PMID: 37007065 PMCID: PMC10064794 DOI: 10.3389/fonc.2023.1066360] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Accepted: 02/13/2023] [Indexed: 03/09/2023] Open
Abstract
ObjectiveTo establish a nomogram based on non-enhanced computed tomography(CT) imaging radiomics and clinical features for use in predicting the malignancy of sub-centimeter solid nodules (SCSNs).Materials and methodsRetrospective analysis was performed of records for 198 patients with SCSNs that were surgically resected and examined pathologically at two medical institutions between January 2020 and June 2021. Patients from Center 1 were included in the training cohort (n = 147), and patients from Center 2 were included in the external validation cohort (n = 52). Radiomic features were extracted from chest CT images. The least absolute shrinkage and selection operator (LASSO) regression model was used for radiomic feature extraction and computation of radiomic scores. Clinical features, subjective CT findings, and radiomic scores were used to build multiple predictive models. Model performance was examined by evaluating the area under the receiver operating characteristic curve (AUC). The best model was selected for efficacy evaluation in a validation cohort, and column line plots were created.ResultsPulmonary malignant nodules were significantly associated with vascular alterations in both the training (p < 0.001) and external validation (p < 0.001) cohorts. Eleven radiomic features were selected after a dimensionality reduction to calculate the radiomic scores. Based on these findings, three prediction models were constructed: subjective model (Model 1), radiomic score model (Model 2), and comprehensive model (Model 3), with AUCs of 0.672, 0.888, and 0.930, respectively. The optimal model with an AUC of 0.905 was applied to the validation cohort, and decision curve analysis indicated that the comprehensive model column line plot was clinically useful.ConclusionPredictive models constructed based on CT-based radiomics with clinical features can help clinicians diagnose pulmonary nodules and guide clinical decision making.
Collapse
Affiliation(s)
- Chengyu Chen
- Department of Thoracic Surgery, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
- Department of Thoracic Surgery, The First Affiliated Hospital of Shandong First Medical University, Jinan, China
| | - Qun Geng
- Department of Ultrasound, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Gesheng Song
- Department of Radiology, The First Affiliated Hospital of Shandong First Medical Unversity, Jinan, China
| | - Qian Zhang
- Department of General Surgery, The First Affiliated Hospital of Shandong First Medical University, Jinan, China
| | - Youruo Wang
- Elite Class of 2017, Shandong First Medical University, Jinan, China
| | - Dongfeng Sun
- Department of Thoracic Surgery, The First Affiliated Hospital of Shandong First Medical University, Jinan, China
| | - Qingshi Zeng
- Department of Radiology, The First Affiliated Hospital of Shandong First Medical Unversity, Jinan, China
| | - Zhengjun Dai
- Scientific Research Department, Huiying Medical Technology Co., Ltd, Beijing, China
| | - Gongchao Wang
- Department of Thoracic Surgery, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
- *Correspondence: Gongchao Wang,
| |
Collapse
|
39
|
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
|
40
|
Fan Y, Li W, Nie W, Yao H, Ren Y, Wang M, Nie H, Gu C, Liu J, An B. Novel Dual-Target Kinase Inhibitors of EGFR and ALK Were Designed, Synthesized, and Induced Cell Apoptosis in Non-Small Cell Lung Cancer. Molecules 2023; 28:molecules28052006. [PMID: 36903251 PMCID: PMC10004195 DOI: 10.3390/molecules28052006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Revised: 02/07/2023] [Accepted: 02/14/2023] [Indexed: 02/25/2023] Open
Abstract
ALK-positive NSCLC coexisting with EGFR mutations is a frequently occurring clinical phenomenon. Targeting ALK and EGFR simultaneously may be an effective way to treat these cancer patients. In this study, we designed and synthesized ten new dual-target EGFR/ALK inhibitors. Among them, the optimal compound 9j exhibited good activity with IC50 values of 0.07829 ± 0.03 μM and 0.08183 ± 0.02 μM against H1975 (EGFR T790M/L858R) and H2228 (EML4-ALK) cells, respectively. Immunofluorescence assays indicated that the compound could simultaneously inhibit the expression of phosphorylated EGFR and ALK proteins. A kinase assay demonstrated that compound 9j could inhibit both EGFR and ALK kinases; thus, exerting an antitumor effect. Additionally, compound 9j induced apoptosis in a dose-dependent manner and inhibited the invasion and migration of tumor cells. All of these results indicate that 9j is worthy of further study.
Collapse
Affiliation(s)
- Yangyang Fan
- School of Pharmacy, Binzhou Medical University, Yantai 264003, China
| | - Wei Li
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Wenyan Nie
- School of Pharmacy, Binzhou Medical University, Yantai 264003, China
| | - Han Yao
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Yuanyuan Ren
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Mengxuan Wang
- School of Pharmacy, Binzhou Medical University, Yantai 264003, China
| | - Haoran Nie
- School of Pharmacy, Binzhou Medical University, Yantai 264003, China
| | - Chenxi Gu
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Jiadai Liu
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Baijiao An
- School of Pharmacy, Binzhou Medical University, Yantai 264003, China
- Shandong Technology Innovation Center of Molecular Targeting and Intelligent Diagnosis and Treatment, Yantai 264003, China
- Correspondence:
| |
Collapse
|
41
|
Xi C, Du R, Wang R, Wang Y, Hou L, Luan M, Zheng X, Huang H, Liang Z, Ding X, Luo Q, Shen C. AI‐BRAF
V600E
: A deep convolutional neural network for BRAF
V600E
mutation status prediction of thyroid nodules using ultrasound images. VIEW 2023. [DOI: 10.1002/viw.20220057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Affiliation(s)
- Chuang Xi
- Department of Nuclear Medicine Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine Shanghai China
| | - Ruiqi Du
- School of Computer Engineering and Science Shanghai University Shanghai China
| | - Ren Wang
- Department of Ultrasound Medicine Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine Shanghai China
| | - Yang Wang
- Department of Nuclear Medicine Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine Shanghai China
| | - Liying Hou
- Department of Nuclear Medicine Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine Shanghai China
| | - Mengqi Luan
- Department of Ultrasound Ruijin Hospital Shanghai Jiaotong University School of Medicine Shanghai China
| | - Xuan Zheng
- Department of Ultrasound Nanjing First Hospital Nanjing Medical University Nanjing China
| | - Hongyan Huang
- Department of Ultrasound Guangdong Second Provincial General Hospital Guangzhou China
| | - Zhixin Liang
- Department of Nuclear Medicine Jinshazhou Hospital Guangzhou University of Chinese Medicine Guangzhou China
| | - Xuehai Ding
- School of Computer Engineering and Science Shanghai University Shanghai China
| | - Quanyong Luo
- Department of Nuclear Medicine Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine Shanghai China
| | - Chentian Shen
- Department of Nuclear Medicine Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine Shanghai China
| |
Collapse
|
42
|
Gao L, Jiang W, Yue Q, Ye R, Li Y, Hong J, Zhang M. Radiomic model to predict the expression of PD-1 and overall survival of patients with ovarian cancer. Int Immunopharmacol 2022; 113:109335. [DOI: 10.1016/j.intimp.2022.109335] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2022] [Revised: 09/29/2022] [Accepted: 10/09/2022] [Indexed: 11/05/2022]
|
43
|
Lin Q, Wu HJ, Song QS, Tang YK. CT-based radiomics in predicting pathological response in non-small cell lung cancer patients receiving neoadjuvant immunotherapy. Front Oncol 2022; 12:937277. [PMID: 36267975 PMCID: PMC9577189 DOI: 10.3389/fonc.2022.937277] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Accepted: 08/30/2022] [Indexed: 11/13/2022] Open
Abstract
Objectives In radiomics, high-throughput algorithms extract objective quantitative features from medical images. In this study, we evaluated CT-based radiomics features, clinical features, in-depth learning features, and a combination of features for predicting a good pathological response (GPR) in non-small cell lung cancer (NSCLC) patients receiving immunotherapy-based neoadjuvant therapy (NAT). Materials and methods We reviewed 62 patients with NSCLC who received surgery after immunotherapy-based NAT and collected clinicopathological data and CT images before and after immunotherapy-based NAT. A series of image preprocessing was carried out on CT scanning images: tumor segmentation, conventional radiomics feature extraction, deep learning feature extraction, and normalization. Spearman correlation coefficient, principal component analysis (PCA), and least absolute shrinkage and selection operator (LASSO) were used to screen features. The pretreatment traditional radiomics combined with clinical characteristics (before_rad_cil) model and pretreatment deep learning characteristics (before_dl) model were constructed according to the data collected before treatment. The data collected after NAT created the after_rad_cil model and after_dl model. The entire model was jointly constructed by all clinical features, conventional radiomics features, and deep learning features before and after neoadjuvant treatment. Finally, according to the data obtained before and after treatment, the before_nomogram and after_nomogram were constructed. Results In the before_rad_cil model, four traditional radiomics features ("original_shape_flatness," "wavelet hhl_firer_skewness," "wavelet hlh_firer_skewness," and "wavelet lll_glcm_correlation") and two clinical features ("gender" and "N stage") were screened out to predict a GPR. The average prediction accuracy (ACC) after modeling with k-nearest neighbor (KNN) was 0.707. In the after_rad_cil model, nine features predictive of GPR were obtained after feature screening, among which seven were traditional radiomics features: "exponential_firer_skewness," "exponential_glrlm_runentropy," "log- sigma-5-0-mm-3d_firer_kurtosis," "logarithm_skewness," "original_shape_elongation," "original_shape_brilliance," and "wavelet llh_glcm_clustershade"; two were clinical features: "after_CRP" and "after lymphocyte percentage." The ACC after modeling with support vector machine (SVM) was 0.682. The before_dl model and after_dl model were modeled by SVM, and the ACC was 0.629 and 0.603, respectively. After feature screening, the entire model was constructed by multilayer perceptron (MLP), and the ACC of the GPR was the highest, 0.805. The calibration curve showed that the predictions of the GPR by the before_nomogram and after_nomogram were in consensus with the actual GPR. Conclusion CT-based radiomics has a good predictive ability for a GPR in NSCLC patients receiving immunotherapy-based NAT. Among the radiomics features combined with the clinicopathological information model, deep learning feature model, and the entire model, the entire model had the highest prediction accuracy.
Collapse
Affiliation(s)
| | - Hai Jun Wu
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, China
| | | | | |
Collapse
|
44
|
Zhang R, Huo X, Wang Q, Zhang J, Duan S, Zhang Q, Zhang S. Prediction of TTF-1 expression in non-small-cell lung cancer using machine learning-based radiomics. J Cancer Res Clin Oncol 2022:10.1007/s00432-022-04357-8. [PMID: 36151427 DOI: 10.1007/s00432-022-04357-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Accepted: 09/10/2022] [Indexed: 11/26/2022]
Abstract
PURPOSE To explore the feasibility and performance of machine learning-based radiomics models in predicting thyroid transcription factor-1 (TTF-1) expression in non-small cell lung cancer (NSCLC). METHODS A total of 227 NSCLC patients were included in this retrospective study and divided into the training set and test set with a ratio of 8:2 randomly. Lung tumors on CT images were semi-automatic segmented utilizing 3D Slicer. Radiomic features quantifying tumor intensity, shape, texture, and transformed wavelet were extracted using a Python toolkit. Variance threshold (VT), principal component analysis (PCA), and least absolute shrinkage selection operator (LASSO) were used to reduce features; logistic regression (LR), random forest (RF), and support vector machine (SVM) were used to develop classifier, respectively. The performance of the models was evaluated by areas under the curves (AUC) of receiver operating characteristic (ROC) curves. Different models were compared by the Delong test to determine the optimal algorithms. RESULTS Total 1968 radiomic features were extracted from the lung tumors images, and then 13, 15, and 13 stable features were selected by VT, PCA, and LASSO, respectively. Each classifier could discriminate against the TTF-1-positive groups with average AUC ranging from 0.601 to 0.784 in the training set. Among the models, three models constructed by the LASSO method showed satisfactory performance in the test set with AUC ranging from 0.715 to 0.787. The Delong test showed no significant difference between the LASSO models (P > 0.05). CONCLUSION Machine learning-based radiomics model could predict the expression of TTF-1 in NSCLC patients.
Collapse
Affiliation(s)
- Ruijie Zhang
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin, 300052, China
- Department of Radiology, Qilu Hospital of Shandong University Dezhou Hospital, Dezhou, 253000, Shandong, China
| | - Xiankai Huo
- Department of Radiology, Qilu Hospital of Shandong University Dezhou Hospital, Dezhou, 253000, Shandong, China
| | - Qian Wang
- Department of Radiology, Qilu Hospital of Shandong University Dezhou Hospital, Dezhou, 253000, Shandong, China
| | - Juntao Zhang
- Precision Health Institution, GE Healthcare, Pudong new town, Shanghai, 210000, China
| | - Shaofeng Duan
- Precision Health Institution, GE Healthcare, Pudong new town, Shanghai, 210000, China
| | - Quan Zhang
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin, 300052, China.
| | - Shicai Zhang
- Department of Radiology, Qilu Hospital of Shandong University Dezhou Hospital, Dezhou, 253000, Shandong, China.
| |
Collapse
|
45
|
Yin X, Liao H, Yun H, Lin N, Li S, Xiang Y, Ma X. Artificial intelligence-based prediction of clinical outcome in immunotherapy and targeted therapy of lung cancer. Semin Cancer Biol 2022; 86:146-159. [PMID: 35963564 DOI: 10.1016/j.semcancer.2022.08.002] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 08/06/2022] [Accepted: 08/08/2022] [Indexed: 11/26/2022]
Abstract
Lung cancer accounts for the main proportion of malignancy-related deaths and most patients are diagnosed at an advanced stage. Immunotherapy and targeted therapy have great advances in application in clinics to treat lung cancer patients, yet the efficacy is unstable. The response rate of these therapies varies among patients. Some biomarkers have been proposed to predict the outcomes of immunotherapy and targeted therapy, including programmed cell death-ligand 1 (PD-L1) expression and oncogene mutations. Nevertheless, the detection tests are invasive, time-consuming, and have high demands on tumor tissue. The predictive performance of conventional biomarkers is also unsatisfactory. Therefore, novel biomarkers are needed to effectively predict the outcomes of immunotherapy and targeted therapy. The application of artificial intelligence (AI) can be a possible solution, as it has several advantages. AI can help identify features that are unable to be used by humans and perform repetitive tasks. By combining AI methods with radiomics, pathology, genomics, transcriptomics, proteomics, and clinical data, the integrated model has shown predictive value in immunotherapy and targeted therapy, which significantly improves the precision treatment of lung cancer patients. Herein, we reviewed the application of AI in predicting the outcomes of immunotherapy and targeted therapy in lung cancer patients, and discussed the challenges and future directions in this field.
Collapse
Affiliation(s)
- Xiaomeng Yin
- Division of Biotherapy, Cancer Center, West China Hospital and State Key Laboratory of Biotherapy, Sichuan University, No. 37 GuoXue Alley, Chengdu 610041, China
| | - Hu Liao
- Department of Thoracic Surgery, West China Hospital, Sichuan University, No. 37 GuoXue Alley, Chengdu 610041, China
| | - Hong Yun
- Division of Biotherapy, Cancer Center, West China Hospital and State Key Laboratory of Biotherapy, Sichuan University, No. 37 GuoXue Alley, Chengdu 610041, China
| | - Nan Lin
- Division of Biotherapy, Cancer Center, West China Hospital and State Key Laboratory of Biotherapy, Sichuan University, No. 37 GuoXue Alley, Chengdu 610041, China
| | - Shen Li
- West China School of Medicine, West China Hospital, Sichuan University, No. 37 GuoXue Alley, Chengdu 610041, China
| | - Yu Xiang
- Division of Biotherapy, Cancer Center, West China Hospital and State Key Laboratory of Biotherapy, Sichuan University, No. 37 GuoXue Alley, Chengdu 610041, China
| | - Xuelei Ma
- Division of Biotherapy, Cancer Center, West China Hospital and State Key Laboratory of Biotherapy, Sichuan University, No. 37 GuoXue Alley, Chengdu 610041, China.
| |
Collapse
|
46
|
Yang H, Wang L, Shao G, Dong B, Wang F, Wei Y, Li P, Chen H, Chen W, Zheng Y, He Y, Zhao Y, Du X, Sun X, Wang Z, Wang Y, Zhou X, Lai X, Feng W, Shen L, Qiu G, Ji Y, Chen J, Jiang Y, Liu J, Zeng J, Wang C, Zhao Q, Yang X, Hu X, Ma H, Chen Q, Chen M, Jiang H, Xu Y. A combined predictive model based on radiomics features and clinical factors for disease progression in early-stage non-small cell lung cancer treated with stereotactic ablative radiotherapy. Front Oncol 2022; 12:967360. [PMID: 35982975 PMCID: PMC9380646 DOI: 10.3389/fonc.2022.967360] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2022] [Accepted: 07/05/2022] [Indexed: 12/04/2022] Open
Abstract
Purpose To accurately assess disease progression after Stereotactic Ablative Radiotherapy (SABR) of early-stage Non-Small Cell Lung Cancer (NSCLC), a combined predictive model based on pre-treatment CT radiomics features and clinical factors was established. Methods This study retrospectively analyzed the data of 96 patients with early-stage NSCLC treated with SABR. Clinical factors included general information (e.g. gender, age, KPS, Charlson score, lung function, smoking status), pre-treatment lesion status (e.g. diameter, location, pathological type, T stage), radiation parameters (biological effective dose, BED), the type of peritumoral radiation-induced lung injury (RILI). Independent risk factors were screened by logistic regression analysis. Radiomics features were extracted from pre-treatment CT. The minimum Redundancy Maximum Relevance (mRMR) and the Least Absolute Shrinkage and Selection Operator (LASSO) were adopted for the dimensionality reduction and feature selection. According to the weight coefficient of the features, the Radscore was calculated, and the radiomics model was constructed. Multiple logistic regression analysis was applied to establish the combined model based on radiomics features and clinical factors. Receiver Operating Characteristic (ROC) curve, DeLong test, Hosmer-Lemeshow test, and Decision Curve Analysis (DCA) were used to evaluate the model’s diagnostic efficiency and clinical practicability. Results With the median follow-up of 59.1 months, 29 patients developed progression and 67 remained good controlled within two years. Among the clinical factors, the type of peritumoral RILI was the only independent risk factor for progression (P< 0.05). Eleven features were selected from 1781 features to construct a radiomics model. For predicting disease progression after SABR, the Area Under the Curve (AUC) of training and validation cohorts in the radiomics model was 0.88 (95%CI 0.80-0.96) and 0.80 (95%CI 0.62-0.98), and AUC of training and validation cohorts in the combined model were 0.88 (95%CI 0.81-0.96) and 0.81 (95%CI 0.62-0.99). Both the radiomics and the combined models have good prediction efficiency in the training and validation cohorts. Still, DeLong test shows that there is no difference between them. Conclusions Compared with the clinical model, the radiomics model and the combined model can better predict the disease progression of early-stage NSCLC after SABR, which might contribute to individualized follow-up plans and treatment strategies.
Collapse
Affiliation(s)
- Hong Yang
- Department of Radiology, Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, China
| | - Lin Wang
- Shaoxing University School of Medicine, Shaoxing, China
| | - Guoliang Shao
- Department of Radiology, Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, China
| | - Baiqiang Dong
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University, Guangzhou, China
| | - Fang Wang
- Department of Radiology, Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, China
| | - Yuguo Wei
- Precision Health Institution, General Electric (GE) Healthcare, Hangzhou, China
| | - Pu Li
- Department of Radiation Physics, Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, China
| | - Haiyan Chen
- Department of Radiology, Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, China
| | - Wujie Chen
- Department of Radiology, Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, China
| | - Yao Zheng
- Department of Radiology, Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, China
| | - Yiwei He
- Department of Radiology, Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, China
| | - Yankun Zhao
- Department of Radiology, Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, China
| | - Xianghui Du
- Department of Radiation Oncology, Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, China
| | - Xiaojiang Sun
- Department of Radiation Oncology, Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, China
| | - Zhun Wang
- Department of Radiation Oncology, Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, China
| | - Yuezhen Wang
- Department of Radiation Oncology, Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, China
| | - Xia Zhou
- Department of Radiation Oncology, Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, China
| | - Xiaojing Lai
- Department of Radiation Oncology, Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, China
| | - Wei Feng
- Department of Radiation Oncology, Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, China
| | - Liming Shen
- Department of Radiation Oncology, Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, China
| | - Guoqing Qiu
- Department of Radiation Oncology, Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, China
| | - Yongling Ji
- Department of Radiation Oncology, Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, China
| | - Jianxiang Chen
- Department of Radiation Oncology, Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, China
| | - Youhua Jiang
- Department of Thoracic Surgery, Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, China
| | - Jinshi Liu
- Department of Thoracic Surgery, Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, China
| | - Jian Zeng
- Department of Thoracic Surgery, Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, China
| | - Changchun Wang
- Department of Thoracic Surgery, Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, China
| | - Qiang Zhao
- Department of Thoracic Surgery, Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, China
| | - Xun Yang
- Department of Thoracic Surgery, Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, China
| | - Xiao Hu
- Department of Radiation Oncology, Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, China
| | - Honglian Ma
- Department of Radiation Oncology, Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, China
| | - Qixun Chen
- Department of Thoracic Surgery, Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, China
| | - Ming Chen
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University, Guangzhou, China
| | - Haitao Jiang
- Department of Radiology, Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, China
- *Correspondence: Haitao Jiang, ; Yujin Xu,
| | - Yujin Xu
- Department of Radiation Oncology, Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, China
- *Correspondence: Haitao Jiang, ; Yujin Xu,
| |
Collapse
|
47
|
Yang X, Fang C, Li C, Gong M, Yi X, Lin H, Li K, Yu X. Can CT Radiomics Detect Acquired T790M Mutation and Predict Prognosis in Advanced Lung Adenocarcinoma With Progression After First- or Second-Generation EGFR TKIs? Front Oncol 2022; 12:904983. [PMID: 35875167 PMCID: PMC9300753 DOI: 10.3389/fonc.2022.904983] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2022] [Accepted: 06/06/2022] [Indexed: 12/03/2022] Open
Abstract
Objective To explore the potential of CT radiomics in detecting acquired T790M mutation and predicting prognosis in patients with advanced lung adenocarcinoma with progression after first- or second-generation epidermal growth factor receptor (EGFR) tyrosine kinase inhibitor (TKI) therapy. Materials and Methods Contrast-enhanced thoracic CT was collected from 250 lung adenocarcinoma patients (with acquired T790M mutation, n = 146, without mutation, n = 104) after progression on first- or second-generation TKIs. Radiomic features were extracted from each volume of interest. The maximum relevance minimum redundancy and the least absolute shrinkage and selection operator (LASSO) regression method were used to select the optimized features in detecting acquired T790M mutation. Univariate Cox regression and LASSO Cox regression were used to establish the radiomics model to predict the progression-free survival of osimertinib treatment. Finally, nomograms (which) combined clinical factors with radscore to predict the acquired T790M mutation and prognosis were built separately. In addition, the two nomograms were validated by the concordance index (C-index), decision curve analysis (DCA), and calibration curve analysis where appropriate. Results Clinical factors including the progression-free survival of first-line EGFR TKIs, EGFR mutation, and N stage and 12 radiomic features were useful in predicting the acquired T790M mutation. The area under the receiver operating characteristic curves (AUC) of clinical, radiomics, and nomogram models were 0.70, 0.74, and 0.78 in the training set and 0.71, 0.71, and 0.76 in the validation set, respectively. The DCA and calibration curve analysis demonstrated a good performance of the nomogram model. Clinical factors including age and first-generation EGFR TKIs and 12 radiomic features were useful in patients’ outcome prediction. The C-index of the combined nomogram was 0.686 in the training set and 0.630 in the validation set, respectively. Calibration curves demonstrated a relatively poor performance of the nomogram model. Conclusion Nomogram combined clinical factors with radiomic features might be helpful to detect whether patients developed acquired T790M mutation or not after progression on first- or second-generation EGFR TKIs. Nomogram prognostic model combined clinical factors with radiomic features might have a limited value in predicting the survival of patients harboring acquired T790M mutation treated with osimertinib.
Collapse
Affiliation(s)
- Xiaohuang Yang
- Department of Radiology, Hunan Cancer Hospital, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Chao Fang
- Department of Clinical Pharmaceutical Research, Hunan Cancer Hospital, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Congrui Li
- Department of Radiology, Hunan Cancer Hospital, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Min Gong
- Department of Radiology, Hunan Cancer Hospital, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Xiaochun Yi
- Department of Radiology, Hunan Cancer Hospital, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Huashan Lin
- Department of Pharmaceutical Diagnosis, General Electric (GE) Healthcare, Changsha, China
| | - Kunyan Li
- Department of Clinical Pharmaceutical Research, Hunan Cancer Hospital, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Xiaoping Yu
- Department of Radiology, Hunan Cancer Hospital, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| |
Collapse
|
48
|
Zheng X, He B, Hu Y, Ren M, Chen Z, Zhang Z, Ma J, Ouyang L, Chu H, Gao H, He W, Liu T, Li G. Diagnostic Accuracy of Deep Learning and Radiomics in Lung Cancer Staging: A Systematic Review and Meta-Analysis. Front Public Health 2022; 10:938113. [PMID: 35923964 PMCID: PMC9339706 DOI: 10.3389/fpubh.2022.938113] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2022] [Accepted: 06/15/2022] [Indexed: 12/24/2022] Open
Abstract
BackgroundArtificial intelligence has far surpassed previous related technologies in image recognition and is increasingly used in medical image analysis. We aimed to explore the diagnostic accuracy of the models based on deep learning or radiomics for lung cancer staging.MethodsStudies were systematically reviewed using literature searches from PubMed, EMBASE, Web of Science, and Wanfang Database, according to PRISMA guidelines. Studies about the diagnostic accuracy of radiomics and deep learning, including the identifications of lung cancer, tumor types, malignant lung nodules and lymph node metastase, were included. After identifying the articles, the methodological quality was assessed using the QUADAS-2 checklist. We extracted the characteristic of each study; the sensitivity, specificity, and AUROC for lung cancer diagnosis were summarized for subgroup analysis.ResultsThe systematic review identified 19 eligible studies, of which 14 used radiomics models and 5 used deep learning models. The pooled AUROC of 7 studies to determine whether patients had lung cancer was 0.83 (95% CI 0.78–0.88). The pooled AUROC of 9 studies to determine whether patients had NSCLC was 0.78 (95% CI 0.73–0.83). The pooled AUROC of the 6 studies that determined patients had malignant lung nodules was 0.79 (95% CI 0.77–0.82). The pooled AUROC of the other 6 studies that determined whether patients had lymph node metastases was 0.74 (95% CI 0.66–0.82).ConclusionThe models based on deep learning or radiomics have the potential to improve diagnostic accuracy for lung cancer staging.Systematic Review Registrationhttps://inplasy.com/inplasy-2022-3-0167/, identifier: INPLASY202230167.
Collapse
Affiliation(s)
- Xiushan Zheng
- Department of Thoracic Surgery, The 3rd Affiliated Hospital of Chengdu Medical College, Pidu District People's Hospital, Chengdu, China
| | - Bo He
- Department of Thoracic Surgery, The 3rd Affiliated Hospital of Chengdu Medical College, Pidu District People's Hospital, Chengdu, China
| | - Yunhai Hu
- Department of Thoracic Surgery, The 3rd Affiliated Hospital of Chengdu Medical College, Pidu District People's Hospital, Chengdu, China
| | - Min Ren
- Department of Thoracic Surgery, The 3rd Affiliated Hospital of Chengdu Medical College, Pidu District People's Hospital, Chengdu, China
| | - Zhiyuan Chen
- Department of Thoracic Surgery, The 3rd Affiliated Hospital of Chengdu Medical College, Pidu District People's Hospital, Chengdu, China
| | - Zhiguang Zhang
- Department of Thoracic Surgery, The 3rd Affiliated Hospital of Chengdu Medical College, Pidu District People's Hospital, Chengdu, China
| | - Jun Ma
- Department of Thoracic Surgery, The 3rd Affiliated Hospital of Chengdu Medical College, Pidu District People's Hospital, Chengdu, China
| | - Lanwei Ouyang
- Department of Thoracic Surgery, The 3rd Affiliated Hospital of Chengdu Medical College, Pidu District People's Hospital, Chengdu, China
| | - Hongmei Chu
- Department of Thoracic Surgery, The 3rd Affiliated Hospital of Chengdu Medical College, Pidu District People's Hospital, Chengdu, China
| | - Huan Gao
- Department of Thoracic Surgery, The 3rd Affiliated Hospital of Chengdu Medical College, Pidu District People's Hospital, Chengdu, China
| | - Wenjing He
- School of Electronic Engineering, Chengdu University of Technology, Chengdu, China
| | - Tianhu Liu
- Department of Cardiology, The 3rd Affiliated Hospital of Chengdu Medical College, Pidu District People's Hospital, Chengdu, China
- *Correspondence: Tianhu Liu
| | - Gang Li
- Department of Cardiology, The 3rd Affiliated Hospital of Chengdu Medical College, Pidu District People's Hospital, Chengdu, China
- Gang Li
| |
Collapse
|
49
|
Prediction Model of Hemorrhage Transformation in Patient with Acute Ischemic Stroke Based on Multiparametric MRI Radiomics and Machine Learning. Brain Sci 2022; 12:brainsci12070858. [PMID: 35884664 PMCID: PMC9313447 DOI: 10.3390/brainsci12070858] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Revised: 06/23/2022] [Accepted: 06/27/2022] [Indexed: 12/13/2022] Open
Abstract
Intravenous thrombolysis is the most commonly used drug therapy for patients with acute ischemic stroke, which is often accompanied by complications of intracerebral hemorrhage transformation (HT). This study proposed to build a reliable model for pretreatment prediction of HT. Specifically, 5400 radiomics features were extracted from 20 regions of interest (ROIs) of multiparametric MRI images of 71 patients. Furthermore, a minimal set of all-relevant features were selected by LASSO from all ROIs and used to build a radiomics model through the random forest (RF). To explore the significance of normal ROIs, we built a model only based on abnormal ROIs. In addition, a model combining clinical factors and radiomics features was further built. Finally, the models were tested on an independent validation cohort. The radiomics model with 14 All-ROIs features achieved pretreatment prediction of HT (AUC = 0.871, accuracy = 0.848), which significantly outperformed the model with only 14 Abnormal-ROIs features (AUC = 0.831, accuracy = 0.818). Besides, combining clinical factors with radiomics features further benefited the prediction performance (AUC = 0.911, accuracy = 0.894). So, we think that the combined model can greatly assist doctors in diagnosis. Furthermore, we find that even if there were no lesions in the normal ROIs, they also provide characteristic information for the prediction of HT.
Collapse
|
50
|
When artificial intelligence meets PD-1/PD-L1 inhibitors: Population screening, response prediction and efficacy evaluation. Comput Biol Med 2022; 145:105499. [DOI: 10.1016/j.compbiomed.2022.105499] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 03/26/2022] [Accepted: 04/03/2022] [Indexed: 02/07/2023]
|