1
|
Horne A, Abravan A, Fornacon-Wood I, O’Connor JPB, Price G, McWilliam A, Faivre-Finn C. Mastering CT-based radiomic research in lung cancer: a practical guide from study design to critical appraisal. Br J Radiol 2025; 98:653-668. [PMID: 40100283 PMCID: PMC12012345 DOI: 10.1093/bjr/tqaf051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Revised: 12/18/2024] [Accepted: 02/25/2025] [Indexed: 03/20/2025] Open
Abstract
Radiomics is a health technology that has the potential to extract clinically meaningful biomarkers from standard of care imaging. Despite a wealth of exploratory analysis performed on scans acquired from patients with lung cancer and existing guidelines describing some of the key steps, no radiomic-based biomarker has been widely accepted. This is primarily due to limitations with methodology, data analysis, and interpretation of the available studies. There is currently a lack of guidance relating to the entire radiomic workflow from study design to critical appraisal. This guide, written with early career lung cancer researchers, describes a more complete radiomic workflow. Lung cancer image analysis is the focus due to some of the unique challenges encountered such as patient movement from breathing. The guide will focus on CT imaging as these are the most common scans performed on patients with lung cancer. The aim of this article is to support the production of high-quality research that has the potential to positively impact outcome of patients with lung cancer.
Collapse
Affiliation(s)
- Ashley Horne
- Division of Cancer Sciences, The University of Manchester, Manchester, M13 9NT, United Kingdom
- Department of Thoracic Oncology, The Christie NHS Foundation Trust, Manchester, M20 4BX, United Kingdom
| | - Azadeh Abravan
- Division of Cancer Sciences, The University of Manchester, Manchester, M13 9NT, United Kingdom
| | - Isabella Fornacon-Wood
- Division of Cancer Sciences, The University of Manchester, Manchester, M13 9NT, United Kingdom
| | - James P B O’Connor
- Division of Cancer Sciences, The University of Manchester, Manchester, M13 9NT, United Kingdom
- Division of Radiotherapy and Imaging, Institute of Cancer Research, London, SW7 3RP, United Kingdom
| | - Gareth Price
- Division of Cancer Sciences, The University of Manchester, Manchester, M13 9NT, United Kingdom
| | - Alan McWilliam
- Division of Cancer Sciences, The University of Manchester, Manchester, M13 9NT, United Kingdom
| | - Corinne Faivre-Finn
- Division of Cancer Sciences, The University of Manchester, Manchester, M13 9NT, United Kingdom
- Department of Thoracic Oncology, The Christie NHS Foundation Trust, Manchester, M20 4BX, United Kingdom
| |
Collapse
|
2
|
Zhai W, Li X, Zhou T, Zhou Q, Lin X, Jiang X, Zhang Z, Jin Q, Liu S, Fan L. A machine learning-based 18F-FDG PET/CT multi-modality fusion radiomics model to predict Mediastinal-Hilar lymph node metastasis in NSCLC: a multi-centre study. Clin Radiol 2025; 83:106832. [PMID: 39983386 DOI: 10.1016/j.crad.2025.106832] [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: 08/14/2024] [Revised: 12/09/2024] [Accepted: 01/27/2025] [Indexed: 02/23/2025]
Abstract
AIM To develop and validate a machine learning (ML) model based on positron emission tomography/computed tomography (PET/CT) multi-modality fusion radiomics to improve the prediction efficiency of mediastinal-hilar lymph node metastasis (LNM). MATERIALS AND METHODS Eighty-eight non-small cell lung cancer (NSCLC) patients with 559 LNs from centre 1 were divided into training and internal validation cohorts (7:3 ratio), and 75 patients with 543 LNs from centre 2 were assigned as external validation cohorts. PET and CT images were fused by wavelet transform. Multi-modality fusion radiomics features from six images of lymph nodes were extracted. The multi-modality fusion radiomics (MFR), multi-modality fusion radiomics + metabolic parameters (MFRM), CT, PET and PET + CT models were developed based on the best one among the 11 ML algorithms. The receiver operating characteristic (ROC) curve and the Delong test were used to assess and compare the performance of the models. RESULTS The CatBoost algorithm was chosen, and the MFR, MFRM, CT, PET and PET + CT models were constructed. The MFR and MFRM models showed a high AUC for predicting LNM in centre 1 (AUC = 0.950 and 0.952) and centre 2 (AUC = 0.923 and 0.927), and there were significant differences in centre 2 (P=0.036). The diagnostic efficacy of MFR and MFRM models was significantly higher than CT, PET, PET + CT models and SUVmax≥3.5 (P<0.001). The MFRM prediction was statistically different from the MFR prediction in the hilar/interlobar zone. CONCLUSION Both the MFR and MFRM models based on multi-modality fusion radiomics showed great potential for non-invasively predicting mediastinal-hilar LNM in NSCLC.
Collapse
Affiliation(s)
- W Zhai
- Department of Radiology, Second Affiliated Hospital of Naval Medical University, Shanghai, China; College of Health Sciences and Engineering, University of Shanghai for Science and Technology, Shanghai, China; Department of Nuclear Medicine, Huadong Hospital Affiliated to Fudan University, Shanghai, China
| | - X Li
- Department of Radiology, Second Affiliated Hospital of Naval Medical University, Shanghai, China
| | - T Zhou
- Department of Radiology, Second Affiliated Hospital of Naval Medical University, Shanghai, China; School of Medical Imaging, Weifang Medical University, Weifang, Shandong, China
| | - Q Zhou
- Department of Radiology, Second Affiliated Hospital of Naval Medical University, Shanghai, China; College of Health Sciences and Engineering, University of Shanghai for Science and Technology, Shanghai, China
| | - X Lin
- Department of Radiology, Second Affiliated Hospital of Naval Medical University, Shanghai, China; College of Health Sciences and Engineering, University of Shanghai for Science and Technology, Shanghai, China
| | - X Jiang
- Department of Radiology, Second Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Z Zhang
- Department of Radiology, Second Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Q Jin
- Department of Radiology, Second Affiliated Hospital of Naval Medical University, Shanghai, China
| | - S Liu
- Department of Radiology, Second Affiliated Hospital of Naval Medical University, Shanghai, China
| | - L Fan
- Department of Radiology, Second Affiliated Hospital of Naval Medical University, Shanghai, China.
| |
Collapse
|
3
|
Li Y, Deng J, Ma X, Li W, Wang Z. Diagnostic accuracy of CT and PET/CT radiomics in predicting lymph node metastasis in non-small cell lung cancer. Eur Radiol 2025; 35:1966-1979. [PMID: 39223336 DOI: 10.1007/s00330-024-11036-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 06/09/2024] [Accepted: 08/07/2024] [Indexed: 09/04/2024]
Abstract
OBJECTIVES This study evaluates the accuracy of radiomics in predicting lymph node metastasis in non-small cell lung cancer, which is crucial for patient management and prognosis. METHODS Adhering to PRISMA and AMSTAR guidelines, we systematically reviewed literature from March 2012 to December 2023 using databases including PubMed, Web of Science, and Embase. Radiomics studies utilizing computed tomography (CT) and positron emission tomography (PET)/CT imaging were included. The quality of studies was appraised with QUADAS-2 and RQS tools, and the TRIPOD checklist assessed model transparency. Sensitivity, specificity, and AUC values were synthesized to determine diagnostic performance, with subgroup and sensitivity analyses probing heterogeneity and a Fagan plot evaluating clinical applicability. RESULTS Our analysis incorporated 42 cohorts from 22 studies. CT-based radiomics demonstrated a sensitivity of 0.84 (95% CI: 0.79-0.88, p < 0.01) and specificity of 0.82 (95% CI: 0.75-0.87, p < 0.01), with an AUC of 0.90 (95% CI: 0.87-0.92), indicating no publication bias (p-value = 0.54 > 0.05). PET/CT radiomics showed a sensitivity of 0.82 (95% CI: 0.76-0.86, p < 0.01) and specificity of 0.86 (95% CI: 0.81-0.90, p < 0.01), with an AUC of 0.90 (95% CI: 0.87-0.93), with a slight publication bias (p-value = 0.03 < 0.05). Despite high clinical utility, subgroup analysis did not clarify heterogeneity sources, suggesting influences from possible factors like lymph node location and small subgroup sizes. CONCLUSIONS Radiomics models show accuracy in predicting lung cancer lymph node metastasis, yet further validation with larger, multi-center studies is necessary. CLINICAL RELEVANCE STATEMENT Radiomics models using CT and PET/CT imaging may improve the prediction of lung cancer lymph node metastasis, aiding personalized treatment strategies. RESEARCH REGISTRATION UNIQUE IDENTIFYING NUMBER (UIN) International Prospective Register of Systematic Reviews (PROSPERO), CRD42023494701. This study has been registered on the PROSPERO platform with a registration date of 18 December 2023. https://www.crd.york.ac.uk/prospero/ KEY POINTS: The study explores radiomics for lung cancer lymph node metastasis detection, impacting surgery and prognosis. Radiomics improves the accuracy of lymph node metastasis prediction in lung cancer. Radiomics can aid in the prediction of lymph node metastasis in lung cancer and personalized treatment.
Collapse
Affiliation(s)
- Yuepeng Li
- Department of Respiratory and Critical Care Medicine, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Respiratory Health and Multimorbidity, West China Hospital, Sichuan University, Chengdu, China
| | - Junyue Deng
- West China School of Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Xuelei Ma
- Department of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, China.
| | - Weimin Li
- Department of Respiratory and Critical Care Medicine, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Respiratory Health and Multimorbidity, West China Hospital, Sichuan University, Chengdu, China
- Institute of Respiratory Health, West China Hospital, Sichuan University, Chengdu, China
- Precision Medicine Center, Precision Medicine Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, China
- The Research Units of West China, Chinese Academy of Medical Sciences, West China Hospital, Chengdu, China
| | - Zhoufeng Wang
- Department of Respiratory and Critical Care Medicine, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Respiratory Health and Multimorbidity, West China Hospital, Sichuan University, Chengdu, China.
- Institute of Respiratory Health, West China Hospital, Sichuan University, Chengdu, China.
- Precision Medicine Center, Precision Medicine Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, China.
- The Research Units of West China, Chinese Academy of Medical Sciences, West China Hospital, Chengdu, China.
| |
Collapse
|
4
|
Ji Q, Zhou G, Sun X. Deep learning signature to predict postoperative anxiety in patients receiving lung cancer surgery. Front Surg 2025; 12:1573370. [PMID: 40196195 PMCID: PMC11973267 DOI: 10.3389/fsurg.2025.1573370] [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: 02/08/2025] [Accepted: 02/20/2025] [Indexed: 04/09/2025] Open
Abstract
This study aims on establishing and validate a deep learning signature based on magnetic resonance imaging (MRI) to predict postoperative anxiety in patients receiving lung cancer surgery. In the current study, 202 patients receiving lung cancer surgery were included. Preoperative MRI-T1WI images were collected to train the deep learning signature utilized the ResNet-152 algorithm. The relationships between clinical variables and postoperative anxiety were explored via Logistic regression and the predictive performances of the developed deep learning signature were evaluated via receiver operating characteristic analysis. Larger tumor size [odds ratio (OR), 2.044; 95% confidence interval (CI), 1.736-3.276; p = 0.002] and occurrence of lymph node metastasis (OR, 2.078; 95% CI, 1.023-3.221; p = 0.043) were revealed as independent predictors for postoperative anxiety. With the increase of deep learning scores, more patients experiencing postoperative anxiety were identified. Moreover, our deep learning signature yielded areas under the curve of 0.865 (95% CI, 0.800-0.930) and 0.822 (95% CI, 0.695-0.950) to predict postoperative anxiety. Therefore, our deep learning signature could help identify lung cancer patients with high risks of postoperative anxiety.
Collapse
Affiliation(s)
- Qingqing Ji
- Shanghai University of Engineering Science, Shanghai, China
| | - Guohua Zhou
- Department of Anesthesiology, Ningbo First Hospital, Ningbo, Zhejiang, China
| | - Xiangxiang Sun
- Department of Thoracic Surgery, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| |
Collapse
|
5
|
Li D, Li S, Zhang H, Xia C, Nan X, Liu H, Chen J. Development and validation of a predictive model for lymph node metastases in peripheral non-small cell lung cancer with a tumor diameter ≤ 2.0 cm and a consolidation-to-tumor ratio > 0.5. Front Oncol 2025; 15:1436771. [PMID: 39911632 PMCID: PMC11794067 DOI: 10.3389/fonc.2025.1436771] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Accepted: 01/07/2025] [Indexed: 02/07/2025] Open
Abstract
Background Precisely predicting lymph node metastasis (LNM) status is critical for the treatment of early non-small5-cell lung cancer (NSCLC). In this study, we developed a LNM prediction tool for peripheral NSCLC with a tumor diameter ≤ 2.0 cm and consolidation-to-tumor ratio (CTR) > 0.5 to identify patients where segmentectomy could be applied. Methods Clinical characteristics were retrospectively collected from 435 patients with NSCLC. Logistic regression analysis of the clinical characteristics of this development cohort was used to estimate independent LNM predictors. A prediction model was then developed and externally validated using a validation cohort at another institution. Results Four independent predictors (tumor size, CTR, pleural indentation, and carcinoembryonic antigen (CEA) values) were identified and entered into the model. The model showed good calibration (Hosmer-Lemeshow (HL) P value = 0.680) with an area under the receiver operating characteristic curve (AUC) = 0.890 (95% confidence interval (CI): 0.808-0.972) in the validation cohort. Conclusions We developed and validated a novel and effective model that predicted the probability of LNM for individual patients with peripheral NSCLC who had a tumor diameter ≤ 2.0 cm and CTR > 0.5. This model could help clinicians make individualized clinical decisions.
Collapse
Affiliation(s)
- Dongyu Li
- Department of Thoracic Surgery, Yuncheng Central Hospital affiliated to Shanxi Medical University, Yuncheng, China
- Department of Lung Cancer Surgery, Tianjin Medical University General Hospital, Tianjin, China
| | - Shaolei Li
- Department of Thoracic Surgery II, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital and Institute, Beijing, China
| | - Hongbing Zhang
- Department of Lung Cancer Surgery, Tianjin Medical University General Hospital, Tianjin, China
| | - Chunqiu Xia
- Department of Lung Cancer Surgery, Tianjin Medical University General Hospital, Tianjin, China
| | - Xiaoyong Nan
- Department of Thoracic Surgery, Yuncheng Central Hospital affiliated to Shanxi Medical University, Yuncheng, China
| | - Hongyu Liu
- Tianjin Key Laboratory of Lung Cancer Metastasis and Tumor Microenvironment, Tianjin Lung Cancer Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Jun Chen
- Department of Lung Cancer Surgery, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Key Laboratory of Lung Cancer Metastasis and Tumor Microenvironment, Tianjin Lung Cancer Institute, Tianjin Medical University General Hospital, Tianjin, China
| |
Collapse
|
6
|
Zhang L, Zhang F, Li G, Xiang X, Liang H, Zhang Y. Predicting lymph node metastasis of clinical T1 non-small cell lung cancer: a brief review of possible methodologies and controversies. Front Oncol 2024; 14:1422623. [PMID: 39720561 PMCID: PMC11667114 DOI: 10.3389/fonc.2024.1422623] [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: 04/24/2024] [Accepted: 11/25/2024] [Indexed: 12/26/2024] Open
Abstract
Non-small cell lung cancer (NSCLC) is a major subtype of lung cancer and poses a serious threat to human health. Due to the advances in lung cancer screening, more and more clinical T1 NSCLC defined as a tumor with a maximum diameter of 3cm surrounded by lung tissue or visceral pleura have been detected and have achieved favorable treatment outcomes, greatly improving the prognosis of NSCLC patients. However, the preoperative lymph node staging and intraoperative lymph node dissection patterns of operable clinical T1 NSCLC are still subject to much disagreement, as well as the heterogeneity between primary tumors and metastatic lymph nodes poses a challenge in designing effective treatment strategies. This article comprehensively describes the clinical risk factors of clinical T1 NSCLC lymph node metastasis, and its invasive and non-invasive prediction, focusing on the genetic heterogeneity between the primary tumor and the metastatic lymph nodes, which is significant for a thoroughly understanding of the biological behavior of early-stage NSCLC.
Collapse
Affiliation(s)
- Li Zhang
- Department of Oncology, the Fifth Affiliated Hospital of Kunming Medical University, Gejiu, China
| | - Feiyue Zhang
- Department of Thoracic Surgery, Yunnan Cancer Center, The Third Affiliated Hospital of Kunming Medical University, Kunming, China
- Department of Oncology, Yuxi City People’s Hospital, The Sixth Affiliated Hospital of Kunming Medical University, Yuxi, China
| | - Gaofeng Li
- Department of Thoracic Surgery, Yunnan Cancer Center, The Third Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Xudong Xiang
- Department of Thoracic Surgery, Yunnan Cancer Center, The Third Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Haifeng Liang
- Department of Oncology, the Fifth Affiliated Hospital of Kunming Medical University, Gejiu, China
| | - Yan Zhang
- Department of Oncology, the Fifth Affiliated Hospital of Kunming Medical University, Gejiu, China
| |
Collapse
|
7
|
Wang L, Zhang C, Li J. A Hybrid CNN-Transformer Model for Predicting N Staging and Survival in Non-Small Cell Lung Cancer Patients Based on CT-Scan. Tomography 2024; 10:1676-1693. [PMID: 39453040 PMCID: PMC11510788 DOI: 10.3390/tomography10100123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 10/03/2024] [Accepted: 10/09/2024] [Indexed: 10/26/2024] Open
Abstract
Accurate assessment of N staging in patients with non-small cell lung cancer (NSCLC) is critical for the development of effective treatment plans, the optimization of therapeutic strategies, and the enhancement of patient survival rates. This study proposes a hybrid model based on 3D convolutional neural networks (CNNs) and transformers for predicting the N-staging and survival rates of NSCLC patients within the NSCLC radiogenomics and Nsclc-radiomics datasets. The model achieved accuracies of 0.805, 0.828, and 0.819 for the training, validation, and testing sets, respectively. By leveraging the strengths of CNNs in local feature extraction and the superior performance of transformers in global information modeling, the model significantly enhances predictive accuracy and efficacy. A comparative analysis with traditional CNN and transformer architectures demonstrates that the CNN-transformer hybrid model outperforms N-staging predictions. Furthermore, this study extracts the one-year survival rate as a feature and employs the Lasso-Cox model for survival predictions at various time intervals (1, 3, 5, and 7 years), with all survival prediction p-values being less than 0.05, illustrating the time-dependent nature of survival analysis. The application of time-dependent ROC curves further validates the model's accuracy and reliability for survival predictions. Overall, this research provides innovative methodologies and new insights for the early diagnosis and prognostic evaluation of NSCLC.
Collapse
Affiliation(s)
| | | | - Jin Li
- College of Intelligent Systems Science and Engineering, Harbin Engineering University, Harbin 150001, China; (L.W.); (C.Z.)
| |
Collapse
|
8
|
Dai M, Wang N, Zhao X, Zhang J, Zhang Z, Zhang J, Wang J, Hu Y, Liu Y, Zhao X, Chen X. Value of Presurgical 18F-FDG PET/CT Radiomics for Predicting Mediastinal Lymph Node Metastasis in Patients with Lung Adenocarcinoma. Cancer Biother Radiopharm 2024; 39:600-610. [PMID: 36342812 DOI: 10.1089/cbr.2022.0038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Objective: The aim of this study was to develop a 18F-fluorodeoxyglucose (18F-FDG) positron emission tomography/computed tomography (PET/CT) radiomic model for predicting mediastinal lymph node metastasis (LNM) in presurgical patients with lung adenocarcinoma. Methods: The study enrolled 320 patients with lung adenocarcinoma (288 internal and 32 external cases) and extracted 190 radiomic features using the LIFEx package. Optimal radiomic features to build a radiomic model were selected using the least absolute shrinkage and selection operator algorithm. Logistic regression was used to build the clinical and complex (combined radiomic and clinical variables) models. Results: Ten radiomic features were selected. In the training group, the area under the receiver operating characteristic curve of the complex model was significantly higher than that of the radiomic and clinical models [0.924 (95% CI: 0.887-0.961) vs. 0.863 (95% CI: 0.814-0.912; p = 0.001) and 0.838 (95% CI: 0.783-0.894; p = 0.000), respectively]. The sensitivity, specificity, accuracy, and positive and negative predictive values of the radiomic model were 0.857, 0.790, 0.811, and 0.651 and 0.924, respectively, which were better than that of visual evaluation (0.539, 0.724, 0.667, and 0.472 and 0.775, respectively) and PET semiquantitative analyses (0.619, 0.732, 0.697, and 0.513 and 0.808, respectively). Conclusions: 18F-FDG PET/CT radiomics showed good predictive performance for LNM and improved the N-stage accuracy of lung adenocarcinoma.
Collapse
Affiliation(s)
- Meng Dai
- Department of Nuclear Medicine, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
- Hebei Provincial Key Laboratory of Tumor Microenvironment and Drug Resistance, Shijiazhuang, China
| | - Na Wang
- Department of Nuclear Medicine, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
- Hebei Provincial Key Laboratory of Tumor Microenvironment and Drug Resistance, Shijiazhuang, China
| | - Xinming Zhao
- Department of Nuclear Medicine, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
- Hebei Provincial Key Laboratory of Tumor Microenvironment and Drug Resistance, Shijiazhuang, China
| | - Jianyuan Zhang
- Department of Nuclear Medicine, Baoding No. 1 Central Hospital, Baoding, China
| | - Zhaoqi Zhang
- Department of Nuclear Medicine, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Jingmian Zhang
- Department of Nuclear Medicine, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Jianfang Wang
- Department of Nuclear Medicine, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Yujing Hu
- Department of Nuclear Medicine, Hebei General Hospital, Shijiazhuang, China
| | - Yunuan Liu
- Department of Nuclear Medicine, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Xiujuan Zhao
- Department of Nuclear Medicine, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Xiaolin Chen
- Department of Nuclear Medicine, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| |
Collapse
|
9
|
Park J, Kim S, Lim JH, Kim CH, You S, Choi JS, Lim JH, Chang JW, Park D, Lee MW, Lee BJ, Shin SC, Cheon YI, Park IS, Han SH, Youn D, Lee HS, Heo J. Development of a multi-modal learning-based lymph node metastasis prediction model for lung cancer. Clin Imaging 2024; 114:110254. [PMID: 39153380 DOI: 10.1016/j.clinimag.2024.110254] [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: 08/05/2024] [Accepted: 08/08/2024] [Indexed: 08/19/2024]
Abstract
PURPOSE This study proposed a three-dimensional (3D) multi-modal learning-based model for the automated prediction and classification of lymph node metastasis in patients with non-small cell lung cancer (NSCLC) using computed tomography (CT) images and clinical information. METHODS We utilized clinical information and CT image data from 4239 patients with NSCLC across multiple institutions. Four deep learning algorithm-based multi-modal models were constructed and evaluated for lymph node classification. To further enhance classification performance, a soft-voting ensemble technique was applied to integrate the outcomes of multiple multi-modal models. RESULTS A comparison of the classification performance revealed that the multi-modal model, which integrated CT images and clinical information, outperformed the single-modal models. Among the four multi-modal models, the Xception model demonstrated the highest classification performance, with an area under the curve (AUC) of 0.756 for the internal test dataset and 0.736 for the external validation dataset. The ensemble model (SEResNet50_DenseNet121_Xception) exhibited even better performance, with an AUC of 0.762 for the internal test dataset and 0.751 for the external validation dataset, surpassing the multi-modal model's performance. CONCLUSIONS Integrating CT images and clinical information improved the performance of the lymph node metastasis prediction models in patients with NSCLC. The proposed 3D multi-modal lymph node prediction model can serve as an auxiliary tool for evaluating lymph node metastasis in patients with non-pretreated NSCLC, aiding in patient screening and treatment planning.
Collapse
Affiliation(s)
- Jeongmin Park
- Department of Radiation Oncology, Ajou University School of Medicine, Suwon, Republic of Korea
| | - Seonhwa Kim
- Department of Radiation Oncology, Ajou University School of Medicine, Suwon, Republic of Korea
| | - June Hyuck Lim
- Department of Radiation Oncology, Ajou University School of Medicine, Suwon, Republic of Korea
| | - Chul-Ho Kim
- Department of Otolaryngology, Ajou University School of Medicine, Suwon, Republic of Korea
| | - Seulgi You
- Department of Radiology, Ajou University School of Medicine, Suwon, Republic of Korea
| | - Jeong-Seok Choi
- Department of Otorhinolaryngology-Head and Neck Surgery Inha University College of Medicine, Incheon, Republic of Korea
| | - Jun Hyeok Lim
- Division of Pulmonology, Department of Internal Medicine, Inha University College of Medicine, Incheon, Republic of Korea
| | - Jae Won Chang
- Department of Otolaryngology-Head and Neck Surgery, Chungnam National University Hospital, Daejeon, Republic of Korea
| | - Dongil Park
- Division of Pulmonary, Allergy and Critical Care Medicine, Critical Care Medicine, Department of Internal Medicine, Chungnam National University Hospital, Daejeon, Republic of Korea
| | - Myung-Won Lee
- Division of Hematology and Oncology, Department of Internal Medicine, Chungnam National University Hospital, Daejeon, Republic of Korea
| | - Byung-Joo Lee
- Department of Otorhinolaryngology - Head and Neck Surgery, College of Medicine, Pusan National University and Biomedical Research Institute, Pusan National University Hospital, Busan, Republic of Korea
| | - Sung-Chan Shin
- Department of Otorhinolaryngology - Head and Neck Surgery, College of Medicine, Pusan National University and Biomedical Research Institute, Pusan National University Hospital, Busan, Republic of Korea
| | - Yong-Il Cheon
- Department of Otorhinolaryngology - Head and Neck Surgery, College of Medicine, Pusan National University and Biomedical Research Institute, Pusan National University Hospital, Busan, Republic of Korea
| | - Il-Seok Park
- Department of Otorhinolaryngology-Head and Neck Surgery, Hallym University Dontan Sacred Heart Hospital, Hallym University College of Medicine, Republic of Korea
| | - Seung Hoon Han
- Department of Otorhinolaryngology-Head and Neck Surgery, Hallym University Dontan Sacred Heart Hospital, Hallym University College of Medicine, Republic of Korea
| | | | | | - Jaesung Heo
- Department of Radiation Oncology, Ajou University School of Medicine, Suwon, Republic of Korea.
| |
Collapse
|
10
|
Zhang L, Li H, Zhao S, Tao X, Li M, Yang S, Zhou L, Liu M, Zhang X, Dong D, Tian J, Wu N. Deep learning model based on primary tumor to predict lymph node status in clinical stage IA lung adenocarcinoma: a multicenter study. JOURNAL OF THE NATIONAL CANCER CENTER 2024; 4:233-240. [PMID: 39281718 PMCID: PMC11401490 DOI: 10.1016/j.jncc.2024.01.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 12/28/2023] [Accepted: 01/22/2024] [Indexed: 09/18/2024] Open
Abstract
Objective To develop a deep learning model to predict lymph node (LN) status in clinical stage IA lung adenocarcinoma patients. Methods This diagnostic study included 1,009 patients with pathologically confirmed clinical stage T1N0M0 lung adenocarcinoma from two independent datasets (699 from Cancer Hospital of Chinese Academy of Medical Sciences and 310 from PLA General Hospital) between January 2005 and December 2019. The Cancer Hospital dataset was randomly split into a training cohort (559 patients) and a validation cohort (140 patients) to train and tune a deep learning model based on a deep residual network (ResNet). The PLA Hospital dataset was used as a testing cohort to evaluate the generalization ability of the model. Thoracic radiologists manually segmented tumors and interpreted high-resolution computed tomography (HRCT) features for the model. The predictive performance was assessed by area under the curves (AUCs), accuracy, precision, recall, and F1 score. Subgroup analysis was performed to evaluate the potential bias of the study population. Results A total of 1,009 patients were included in this study; 409 (40.5%) were male and 600 (59.5%) were female. The median age was 57.0 years (inter-quartile range, IQR: 50.0-64.0). The deep learning model achieved AUCs of 0.906 (95% CI: 0.873-0.938) and 0.893 (95% CI: 0.857-0.930) for predicting pN0 disease in the testing cohort and a non-pure ground glass nodule (non-pGGN) testing cohort, respectively. No significant difference was detected between the testing cohort and the non-pGGN testing cohort (P = 0.622). The precisions of this model for predicting pN0 disease were 0.979 (95% CI: 0.963-0.995) and 0.983 (95% CI: 0.967-0.998) in the testing cohort and the non-pGGN testing cohort, respectively. The deep learning model achieved AUCs of 0.848 (95% CI: 0.798-0.898) and 0.831 (95% CI: 0.776-0.887) for predicting pN2 disease in the testing cohort and the non-pGGN testing cohort, respectively. No significant difference was detected between the testing cohort and the non-pGGN testing cohort (P = 0.657). The recalls of this model for predicting pN2 disease were 0.903 (95% CI: 0.870-0.936) and 0.931 (95% CI: 0.901-0.961) in the testing cohort and the non-pGGN testing cohort, respectively. Conclusions The superior performance of the deep learning model will help to target the extension of lymph node dissection and reduce the ineffective lymph node dissection in early-stage lung adenocarcinoma patients.
Collapse
Affiliation(s)
- Li Zhang
- Department of Diagnostic Radiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Hailin Li
- Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, School of Engineering Medicine, Beihang University, Beijing, China
- CAS Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing, China
| | - Shaohong Zhao
- Department of Radiology, PLA General Hospital, Beijing, China
| | - Xuemin Tao
- Department of Radiology, PLA General Hospital, Beijing, China
| | - Meng Li
- Department of Diagnostic Radiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Shouxin Yang
- Department of Radiology, Peking University Cancer Hospital & Institute, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Beijing, China
| | - Lina Zhou
- Department of Diagnostic Radiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Mengwen 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, Beijing, China
| | - Xue Zhang
- Department of Diagnostic Radiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Di Dong
- School of Artificial Intelligence, University of Chinese Academy of Sciences, Beijing, China
- CAS Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing, China
| | - Jie Tian
- Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, School of Engineering Medicine, Beihang University, Beijing, China
- CAS Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing, China
| | - Ning Wu
- Department of Diagnostic Radiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
11
|
Yan HJ, Zhao JS, Zuo HD, Zhang JJ, Deng ZQ, Yang C, Luo X, Wan JX, Zheng XY, Chen WY, Li SP, Tian D. Dual-Region Computed Tomography Radiomics-Based Machine Learning Predicts Subcarinal Lymph Node Metastasis in Patients with Non-small Cell Lung Cancer. Ann Surg Oncol 2024; 31:5011-5020. [PMID: 38520581 DOI: 10.1245/s10434-024-15197-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Accepted: 03/04/2024] [Indexed: 03/25/2024]
Abstract
BACKGROUND Noninvasively and accurately predicting subcarinal lymph node metastasis (SLNM) for patients with non-small cell lung cancer (NSCLC) remains challenging. This study was designed to develop and validate a tumor and subcarinal lymph nodes (tumor-SLNs) dual-region computed tomography (CT) radiomics model for predicting SLNM in NSCLC. METHODS This retrospective study included NSCLC patients who underwent lung resection and SLNs dissection between January 2017 and December 2020. The radiomic features of the tumor and SLNs were extracted from preoperative CT, respectively. Ninety machine learning (ML) models were developed based on tumor region, SLNs region, and tumor-SLNs dual-region. The model performance was assessed by the area under the curve (AUC) and validated internally by fivefold cross-validation. RESULTS In total, 202 patients were included in this study. ML models based on dual-region radiomics showed good performance for SLNM prediction, with a median AUC of 0.794 (range, 0.686-0.880), which was superior to those of models based on tumor region (median AUC, 0.746; range, 0.630-0.811) and SLNs region (median AUC, 0.700; range, 0.610-0.842). The ML model, which is developed by using the naive Bayes algorithm and dual-region features, had the highest AUC of 0.880 (range of cross-validation, 0.825-0.937) among all ML models. The optimal logistic regression model was inferior to the optimal ML model for predicting SLNM, with an AUC of 0.727. CONCLUSIONS The CT radiomics showed the potential for accurately predicting SLNM in NSCLC patients. The ML model with dual-region radiomic features has better performance than the logistic regression or single-region models.
Collapse
Affiliation(s)
- Hao-Ji Yan
- Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu, China
- Department of General Thoracic Surgery, Juntendo University School of Medicine, Tokyo, Japan
| | - Jia-Sheng Zhao
- College of Clinical Medicine, North Sichuan Medical College, Nanchong, China
| | - Hou-Dong Zuo
- Medical Imaging Key Laboratory of Sichuan Province, Department of Radiology, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Jun-Jie Zhang
- College of Medical Imaging, North Sichuan Medical College, Nanchong, China
| | - Zhi-Qiang Deng
- College of Medical Imaging, North Sichuan Medical College, Nanchong, China
| | - Chen Yang
- College of Medical Imaging, North Sichuan Medical College, Nanchong, China
| | - Xi Luo
- College of Clinical Medicine, North Sichuan Medical College, Nanchong, China
| | - Jia-Xin Wan
- College of Clinical Medicine, North Sichuan Medical College, Nanchong, China
| | - Xiang-Yun Zheng
- Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Wei-Yang Chen
- Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Su-Ping Li
- Department of Nuclear Medicine, Affiliated Hospital of North Sichuan Medical College, North Sichuan Medical College, Nanchong, China.
| | - Dong Tian
- Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|
12
|
Yang M, Li X, Cai C, Liu C, Ma M, Qu W, Zhong S, Zheng E, Zhu H, Jin F, Shi H. [ 18F]FDG PET-CT radiomics signature to predict pathological complete response to neoadjuvant chemoimmunotherapy in non-small cell lung cancer: a multicenter study. Eur Radiol 2024; 34:4352-4363. [PMID: 38127071 DOI: 10.1007/s00330-023-10503-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Revised: 11/07/2023] [Accepted: 11/29/2023] [Indexed: 12/23/2023]
Abstract
OBJECTIVES This study aims to develop and validate a radiomics model based on 18F-fluorodeoxyglucose positron emission tomography-computed tomography ([18F]FDG PET-CT) images to predict pathological complete response (pCR) to neoadjuvant chemoimmunotherapy in non-small cell lung cancer (NSCLC). MATERIALS AND METHODS One hundred eighty-five patients receiving neoadjuvant chemoimmunotherapy for NSCLC at 5 centers from January 2019 to December 2022 were included and divided into a training cohort and a validation cohort. Radiomics models were constructed via the least absolute shrinkage and selection operator (LASSO) method. The performances of models were evaluated by the area under the receiver operating characteristic curve (AUC). In addition, genetic analyses were conducted to reveal the underlying biological basis of the radiomics score. RESULTS After the LASSO process, 9 PET-CT radiomics features were selected for pCR prediction. In the validation cohort, the ability of PET-CT radiomics model to predict pCR was shown to have an AUC of 0.818 (95% confidence interval [CI], 0.711, 0.925), which was better than the PET radiomics model (0.728 [95% CI, 0.610, 0.846]), CT radiomics model (0.732 [95% CI, 0.607, 0.857]), and maximum standard uptake value (0.603 [95% CI, 0.473, 0.733]) (p < 0.05). Moreover, a high radiomics score was related to the upregulation of pathways suppressing tumor proliferation and the infiltration of antitumor immune cell. CONCLUSION The proposed PET-CT radiomics model was capable of predicting pCR to neoadjuvant chemoimmunotherapy in NSCLC patients. CLINICAL RELEVANCE STATEMENT This study indicated that the generated 18F-fluorodeoxyglucose positron emission tomography-computed tomography radiomics model could predict pathological complete response to neoadjuvant chemoimmunotherapy, implying the potential of our radiomics model to personalize the neoadjuvant chemoimmunotherapy in lung cancer patients. KEY POINTS • Recognizing patients potentially benefiting neoadjuvant chemoimmunotherapy is critical for individualized therapy of lung cancer. • [18F]FDG PET-CT radiomics could predict pathological complete response to neoadjuvant immunotherapy in non-small cell lung cancer. • [18F]FDG PET-CT radiomics model could personalize neoadjuvant chemoimmunotherapy in lung cancer patients.
Collapse
Affiliation(s)
- Minglei Yang
- Department of Thoracic Surgery, Ningbo No. 2 Hospital, Ningbo, China
| | - Xiaoxiao Li
- Shanghai Universal Cloud Medical Imaging Diagnostic Center, Shanghai, China
| | - Chuang Cai
- School of Computer Science and Communication Engineering, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Chunli Liu
- Shanghai Universal Cloud Medical Imaging Diagnostic Center, Shanghai, China
| | - Minjie Ma
- Department of Thoracic Surgery, The First Hospital of Lanzhou University, Lanzhou, Gansu, China
| | - Wendong Qu
- Department of Thoracic Surgery, The Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China.
| | | | - Enkuo Zheng
- Department of Thoracic Surgery, Ningbo No. 2 Hospital, Ningbo, China
| | - Huangkai Zhu
- Department of Thoracic Surgery, Ningbo No. 2 Hospital, Ningbo, China
| | - Feng Jin
- Shandong Key Laboratory of Infectious Respiratory Diseases, Shandong Public Health Clinical Center, Shandong University, Shandong, China.
| | - Huazheng Shi
- Shanghai Universal Cloud Medical Imaging Diagnostic Center, Shanghai, China.
| |
Collapse
|
13
|
Ruan D, Fang J, Teng X. Efficient 18F-fluorodeoxyglucose positron emission tomography/computed tomography-based machine learning model for predicting epidermal growth factor receptor mutations in non-small cell lung cancer. THE QUARTERLY JOURNAL OF NUCLEAR MEDICINE AND MOLECULAR IMAGING : OFFICIAL PUBLICATION OF THE ITALIAN ASSOCIATION OF NUCLEAR MEDICINE (AIMN) [AND] THE INTERNATIONAL ASSOCIATION OF RADIOPHARMACOLOGY (IAR), [AND] SECTION OF THE SOCIETY OF... 2024; 68:70-83. [PMID: 35420272 DOI: 10.23736/s1824-4785.22.03441-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
BACKGROUND Beyond the human eye's limitations, radiomics provides more information that can be used for diagnosis. We develop a personalized and efficient model based on 18F-fluorodeoxyglucose (18F-FDG) positron emission tomography/computed tomography (PET/CT) to predict epidermal growth factor receptor (EGFR) mutations to help identify which non-small cell cancer (NSCLC) patients are candidates for EGFR-tyrosine kinase inhibitors (TKIs) therapy. METHODS We retrospectively included 100 patients with NSCLC and randomized them according to 70 patients in the training group and 30 patients in the validation group. The least absolute shrinkage and selection operator logistic regression (LLR) algorithm and support vector machine (SVM) classifier were used to build the models and predict whether EGFR is mutated or not. The predictive efficacy of the LLR algorithm-based model and the SVM classifier-based model was evaluated by plotting the receiver operating characteristic (ROC) curves and calculating the area under the curve (AUC). RESULTS The AUC, sensitivity and specificity of our radiomics model by LLR algorithm were 0.792, 0.967, and 0.600 for the training group and 0.643, 1.00, and 0.378 for the validation group, respectively, in predicting EGFR mutations. The AUC was 0.838 for the training group and 0.696 for the validation group after combining radiomics features with clinical features. The prediction results based on the SVM classifier showed that the validation group had the best performance when based on radial kernel function with AUC, sensitivity, and specificity of 0.741, 0.667, and 0.825, respectively. CONCLUSIONS Radiomics models based on 18F-FDG PET/CT modeled with different machine learning algorithms can improve the predictive efficacy of the models. Models that combine clinical features are more clinically valuable.
Collapse
Affiliation(s)
- Dan Ruan
- Department of Nuclear Medicine, Xiamen Branch, Zhongshan Hospital, Fudan University, Fujian, China -
| | - Janyao Fang
- Department of Nuclear Medicine, Xiamen Branch, Zhongshan Hospital, Fudan University, Fujian, China
| | - Xinyu Teng
- Department of Nuclear Medicine, Xiamen Branch, Zhongshan Hospital, Fudan University, Fujian, China
| |
Collapse
|
14
|
Song X, Duan X, He X, Wang Y, Li K, Deng B, Chen X, Wang Y, Li M, Shan H. Computer-aided diagnosis of distal metastasis in non-small cell lung cancer by low-dose CT based radiomics and deep learning signatures. LA RADIOLOGIA MEDICA 2024; 129:239-251. [PMID: 38214839 DOI: 10.1007/s11547-024-01770-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Accepted: 01/03/2024] [Indexed: 01/13/2024]
Abstract
BACKGROUND This study aimed to develop and validate radiomics and deep learning (DL) signatures for predicting distal metastasis (DM) of non-small cell lung cancer (NSCLC) in low-dose computed tomography (LDCT). METHODS Images and clinical data were retrospectively collected for 381 NSCLC patients and prospectively collected for 114 patients at the Fifth Affiliated Hospital of Sun Yat-Sen University. Additionally, we enrolled 179 patients from the Jiangmen Central Hospital to externally validate the signatures. Machine-learning algorithms were employed to develop radiomics signature while the DL signature was developed using neural architecture search. The diagnostic efficiency was primarily quantified with the area under receiver operating characteristic curve (AUC). We interpreted the reasoning process of the radiomics signature and DL signature by radiomics voxel mapping and attention weight tracking. RESULTS A total of 674 patients with pathologically-confirmed NSCLC were included from two institutions, with 143 of them having DM. The radiomics signature achieved AUCs of 0.885, 0.854, and 0.733 in the internal validation, prospective validation, and external validation while those for DL signature were 0.893, 0.786, and 0.780. The proposed signatures achieved a promising performance in predicting the DM of NSCLC and outperformed the approaches proposed in previous studies. Interpretability analysis revealed that both radiomics and DL signatures could detect the variations among voxels inside tumors, which helped in identifying the DM of NSCLC. CONCLUSIONS Our study demonstrates the potential of LDCT-based radiomics and DL signatures for predicting DM in NSCLC. These signatures could help improve lung cancer screening regarding further diagnostic tests and treatment strategies.
Collapse
Affiliation(s)
- Xiaoyi Song
- Guangdong Provincial Engineering Research Center of Molecular Imaging, the Fifth Affiliated Hospital, Sun Yat-Sen University, Zhuhai, 519000, Guangdong Province, China
- Guangdong-Hong Kong-Macao University Joint Laboratory of Interventional Medicine, the Fifth Affiliated Hospital, Sun Yat-Sen University, Zhuhai, 519000, China
| | - Xiaobei Duan
- Department of Nuclear Medicine, Jiangmen Central Hospital, Jiangmen, 529030, China
| | - Xinghua He
- Department of Nuclear Medicine, the Fifth Affiliated Hospital, Sun Yat-Sen University, Zhuhai, 519000, Guangdong Province, China
| | - Yubo Wang
- Department of Nuclear Medicine, the Fifth Affiliated Hospital, Sun Yat-Sen University, Zhuhai, 519000, Guangdong Province, China
| | - Kunwei Li
- Department of Radiology, the Fifth Affiliated Hospital, Sun Yat-Sen University, Zhuhai, 519000, China
| | - Bangxuan Deng
- Department of Nuclear Medicine, the Fifth Affiliated Hospital, Sun Yat-Sen University, Zhuhai, 519000, Guangdong Province, China
| | - Xiangmeng Chen
- Department of Radiology, Jiangmen Central Hospital, Jiangmen, 529030, China
| | - Ying Wang
- Department of Nuclear Medicine, the Fifth Affiliated Hospital, Sun Yat-Sen University, Zhuhai, 519000, Guangdong Province, China.
| | - Man Li
- Guangdong Provincial Engineering Research Center of Molecular Imaging, the Fifth Affiliated Hospital, Sun Yat-Sen University, Zhuhai, 519000, Guangdong Province, China.
- Guangdong-Hong Kong-Macao University Joint Laboratory of Interventional Medicine, the Fifth Affiliated Hospital, Sun Yat-Sen University, Zhuhai, 519000, China.
- Department of Information Technology and Data Center, the Fifth Affiliated Hospital, Sun Yat-Sen University, Zhuhai, 519000, China.
- Biobank of the Fifth Affiliated Hospital, Sun Yat-Sen University, Zhuhai, 519000, China.
| | - Hong Shan
- Guangdong Provincial Engineering Research Center of Molecular Imaging, the Fifth Affiliated Hospital, Sun Yat-Sen University, Zhuhai, 519000, Guangdong Province, China.
- Guangdong-Hong Kong-Macao University Joint Laboratory of Interventional Medicine, the Fifth Affiliated Hospital, Sun Yat-Sen University, Zhuhai, 519000, China.
- Department of Interventional Medicine, the Fifth Affiliated Hospital, Sun Yat-Sen University, Zhuhai, 519000, China.
| |
Collapse
|
15
|
Tian W, Yan Q, Huang X, Feng R, Shan F, Geng D, Zhang Z. Predicting occult lymph node metastasis in solid-predominantly invasive lung adenocarcinoma across multiple centers using radiomics-deep learning fusion model. Cancer Imaging 2024; 24:8. [PMID: 38216999 PMCID: PMC10785418 DOI: 10.1186/s40644-024-00654-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Accepted: 01/02/2024] [Indexed: 01/14/2024] Open
Abstract
BACKGROUND In solid-predominantly invasive lung adenocarcinoma (SPILAC), occult lymph node metastasis (OLNM) is pivotal for determining treatment strategies. This study seeks to develop and validate a fusion model combining radiomics and deep learning to predict OLNM preoperatively in SPILAC patients across multiple centers. METHODS In this study, 1325 cT1a-bN0M0 SPILAC patients from six hospitals were retrospectively analyzed and divided into pathological nodal positive (pN+) and negative (pN-) groups. Three predictive models for OLNM were developed: a radiomics model employing decision trees and support vector machines; a deep learning model using ResNet-18, ResNet-34, ResNet-50, DenseNet-121, and Swin Transformer, initialized randomly or pre-trained on large-scale medical data; and a fusion model integrating both approaches using addition and concatenation techniques. The model performance was evaluated by the area under the receiver operating characteristic (ROC) curve (AUC). RESULTS All patients were assigned to four groups: training set (n = 470), internal validation set (n = 202), and independent test set 1 (n = 227) and 2 (n = 426). Among the 1325 patients, 478 (36%) had OLNM (pN+). The fusion model, combining radiomics with pre-trained ResNet-18 features via concatenation, outperformed others with an average AUC (aAUC) of 0.754 across validation and test sets, compared to aAUCs of 0.715 for the radiomics model and 0.676 for the deep learning model. CONCLUSION The radiomics-deep learning fusion model showed promising ability to generalize in predicting OLNM from CT scans, potentially aiding personalized treatment for SPILAC patients across multiple centers.
Collapse
Affiliation(s)
- Weiwei Tian
- Academy for Engineering and Technology, Fudan University, No. 220 Handan Road, Shanghai, 200433, China
| | - Qinqin Yan
- Department of Radiology, Ruijin Hospital, Shanghai Jiao Tong University of Medicine, No. 197 Ruijin Er Road, Shanghai, 200025, China
| | - Xinyu Huang
- School of Computer Science, Shanghai Key Laboratory of Intelligent Information Processing, Fudan University, No. 2005 Songhu Road, Shanghai, 200433, China
| | - Rui Feng
- School of Computer Science, Shanghai Key Laboratory of Intelligent Information Processing, Fudan University, No. 2005 Songhu Road, Shanghai, 200433, China.
- Fudan University, No. 220 Handan Road, Shanghai, 200433, China.
| | - Fei Shan
- Department of Radiology, Shanghai Public Health Clinical Center, Fudan University, No. 2501 Caolang Road, Shanghai, 201508, China.
| | - Daoying Geng
- Academy for Engineering and Technology, Fudan University, No. 220 Handan Road, Shanghai, 200433, China
| | - Zhiyong Zhang
- Department of Radiology, Shanghai Public Health Clinical Center, Fudan University, No. 2501 Caolang Road, Shanghai, 201508, China
- Department of Radiology, Zhongshan Hospital, Fudan University, No. 180 Fenglin Road, Shanghai, 200032, China
- Fudan University, No. 220 Handan Road, Shanghai, 200433, China
| |
Collapse
|
16
|
Meng N, Feng P, Yu X, Wu Y, Fu F, Li Z, Luo Y, Tan H, Yuan J, Yang Y, Wang Z, Wang M. An [ 18F]FDG PET/3D-ultrashort echo time MRI-based radiomics model established by machine learning facilitates preoperative assessment of lymph node status in non-small cell lung cancer. Eur Radiol 2024; 34:318-329. [PMID: 37530809 DOI: 10.1007/s00330-023-09978-2] [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: 11/14/2022] [Revised: 04/09/2023] [Accepted: 04/21/2023] [Indexed: 08/03/2023]
Abstract
OBJECTIVES To develop an [18F]FDG PET/3D-UTE model based on clinical factors, three-dimensional ultrashort echo time (3D-UTE), and PET radiomics features via machine learning for the assessment of lymph node (LN) status in non-small cell lung cancer (NSCLC). METHODS A total of 145 NSCLC patients (training, 101 cases; test, 44 cases) underwent whole-body [18F]FDG PET/CT and chest [18F]FDG PET/MRI were enrolled. Preoperative clinical factors and 3D-UTE, CT, and PET radiomics features were analyzed. The Mann-Whitney U test, LASSO regression, and SelectKBest were used for feature extraction. Five machine learning algorithms were used to establish prediction models, which were evaluated by the area under receiver-operator characteristic (ROC), DeLong test, calibration curves, and decision curve analysis (DCA). RESULTS A prediction model based on random forest, consisting of four clinical factors, six 3D-UTE, and six PET radiomics features, was used as the final model for PET/3D-UTE. The AUCs of this model were 0.912 and 0.791 in the training and test sets, respectively, which not only showed different degrees of improvement over individual models such as clinical, 3D-UTE, and PET (AUC-training = 0.838, 0.834, and 0.828, AUC-test = 0.756, 0.745, and 0.768, respectively) but also achieved the similar diagnostic efficacy as the optimal PET/CT model (AUC-training = 0.890, AUC-test = 0.793). The calibration curves and DCA indicated good consistency (C-index, 0.912) and clinical utility of this model, respectively. CONCLUSION The [18F]FDG PET/3D-UTE model based on clinical factors, 3D-UTE, and PET radiomics features using machine learning methods could noninvasively assess the LN status of NSCLC. CLINICAL RELEVANCE STATEMENT A machine learning model of 18F-fluorodeoxyglucose positron emission tomography/ three-dimensional ultrashort echo time could noninvasively assess the lymph node status of non-small cell lung cancer, which provides a novel method with less radiation burden for clinical practice. KEY POINTS • The 3D-UTE radiomics model using the PLS-DA classifier was significantly associated with LN status in NSCLC and has similar diagnostic performance as the clinical, CT, and PET models. • The [18F]FDG PET/3D-UTE model based on clinical factors, 3D-UTE, and PET radiomics features using the RF classifier could noninvasively assess the LN status of NSCLC and showed improved diagnostic performance compared to the clinical, 3D-UTE, and PET models. • In the assessment of LN status in NSCLC, the [18F]FDG PET/3D-UTE model has similar diagnostic efficacy as the [18F]FDG PET/CT model that incorporates clinical factors and CT and PET radiomics features.
Collapse
Affiliation(s)
- Nan Meng
- Department of Medical Imaging, Henan Provincial People's Hospital & Zhengzhou University People's Hospital, 7 Weiwu Road, Zhengzhou, 450000, China
- Laboratory of Brain Science and Brain-Like Intelligence Technology, Biomedical Research Institute, Henan Academy of Science, Zhengzhou, China
- Academy of Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Pengyang Feng
- Department of Medical Imaging, Henan Provincial People's Hospital & Zhengzhou University People's Hospital, 7 Weiwu Road, Zhengzhou, 450000, China
- Department of Medical Imaging, Henan University People's Hospital & Henan Provincial People's Hospital, Zhengzhou, China
| | - Xuan Yu
- Department of Medical Imaging, Henan Provincial People's Hospital & Zhengzhou University People's Hospital, 7 Weiwu Road, Zhengzhou, 450000, China
| | - Yaping Wu
- Department of Medical Imaging, Henan Provincial People's Hospital & Zhengzhou University People's Hospital, 7 Weiwu Road, Zhengzhou, 450000, China
| | - Fangfang Fu
- Department of Medical Imaging, Henan Provincial People's Hospital & Zhengzhou University People's Hospital, 7 Weiwu Road, Zhengzhou, 450000, China
| | - Ziqiang Li
- Department of Medical Imaging, Xinxiang Medical University People's Hospital & Henan Provincial People's Hospital, Zhengzhou, China
| | - Yu Luo
- Department of Medical Imaging, Henan Provincial People's Hospital & Zhengzhou University People's Hospital, 7 Weiwu Road, Zhengzhou, 450000, China
| | - Hongna Tan
- Department of Medical Imaging, Henan Provincial People's Hospital & Zhengzhou University People's Hospital, 7 Weiwu Road, Zhengzhou, 450000, China
| | - Jianmin Yuan
- Central Research Institute, United Imaging Healthcare Group, Shanghai, China
| | - Yang Yang
- Beijing United Imaging Research Institute of Intelligent Imaging, United Imaging Healthcare Group, Beijing, China
| | - Zhe Wang
- Central Research Institute, United Imaging Healthcare Group, Shanghai, China
| | - Meiyun Wang
- Department of Medical Imaging, Henan Provincial People's Hospital & Zhengzhou University People's Hospital, 7 Weiwu Road, Zhengzhou, 450000, China.
- Laboratory of Brain Science and Brain-Like Intelligence Technology, Biomedical Research Institute, Henan Academy of Science, Zhengzhou, China.
- Academy of Medical Sciences, Zhengzhou University, Zhengzhou, China.
| |
Collapse
|
17
|
Pan Z, Zhang Y, Zhang L, Wang L, Zhao K, Li Q, Wang A, Hu Y, Xie X. Detection, measurement, and diagnosis of lung nodules by ultra-low-dose CT in lung cancer screening: a systematic review. BJR Open 2024; 6:tzae041. [PMID: 39665102 PMCID: PMC11634541 DOI: 10.1093/bjro/tzae041] [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: 06/26/2024] [Revised: 09/24/2024] [Accepted: 11/16/2024] [Indexed: 12/13/2024] Open
Abstract
Objective There is a lack of recent meta-analyses and systematic reviews on the use of ultra-low-dose CT (ULDCT) for the detection, measurement, and diagnosis of lung nodules. This review aims to summarize the latest advances of ULDCT in these areas. Methods A systematic review of studies in PubMed and Web of Science was conducted, using search terms specific to ULDCT and lung nodules. The included studies were published in the last 5 years (January 2019-August 2024). Two reviewers independently selected articles, extracted data, and assessed the risk of bias and concerns using the Quality Assessment of Diagnostic Accuracy Studies-II (QUADAS-II) tool. The standard-dose, low-dose, or contrast-enhanced CT served as the reference-standard CT to evaluate ULDCT. Results The literature search yielded 15 high-quality articles on a total of 1889 patients, of which 10, 3, and 2 dealt with the detection, measurement, and diagnosis of lung nodules. QUADAS-II showed a generally low risk of bias. The mean radiation dose for ULDCT was 0.22 ± 0.10 mSv (7.7%) against 2.84 ± 1.80 mSv for reference-standard CT. Nodule detection rates ranged from 86.1% to 100%. The variability of diameter measurements ranged from 2.1% to 14.4% against contrast-enhanced CT and from 3.1% to 8.29% against standard CT. The diagnosis rate of malignant nodules ranged from 75% to 91%. Conclusions ULDCT proves effective in detecting lung nodules while substantially reducing radiation exposure. However, the use of ULDCT for the measurement and diagnosis of lung nodules remains challenging and requires further research. Advances in knowledge When ULDCT reduces radiation exposure to 7.7%, it detects lung nodules at a rate of 86.1%-100%, with a measurement variance of 2.1%-14.4% and a diagnostic accuracy for malignancy of 75%-91%, suggesting the potential for safe and effective lung cancer screening.
Collapse
Affiliation(s)
- Zhijie Pan
- Radiology Department, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| | - Yaping Zhang
- Radiology Department, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| | - Lu Zhang
- Radiology Department, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| | - Lingyun Wang
- Radiology Department, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| | - Keke Zhao
- Radiology Department, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| | - Qingyao Li
- Radiology Department, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
- Radiology Department, Shanghai General Hospital, University of Shanghai for Science and Technology, Shanghai 200093, China
| | - Ai Wang
- Radiology Department, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| | - Yanfei Hu
- Radiology Department, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| | - Xueqian Xie
- Radiology Department, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| |
Collapse
|
18
|
Yan H, Yang H, Jiang P, Dong L, Zhang Z, Zhou Y, Zeng Q, Li P, Sun Y, Zhu S. A radiomics model based on T2WI and clinical indexes for prediction of lateral lymph node metastasis in rectal cancer. Asian J Surg 2024; 47:450-458. [PMID: 37833219 DOI: 10.1016/j.asjsur.2023.09.156] [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: 06/26/2023] [Revised: 09/19/2023] [Accepted: 09/28/2023] [Indexed: 10/15/2023] Open
Abstract
OBJECTIVE The aim of this study was to explore the clinical value of a radiomics prediction model based on T2-weighted imaging (T2WI) and clinical indexes in predicting lateral lymph node (LLN) metastasis in rectal cancer patients. METHODS This was a retrospective analysis of 106 rectal cancer patients who had undergone LLN dissection. The clinical risk factors for LLN metastasis were selected by multivariable logistic regression analysis of the clinical indicators of the patients. The LLN radiomics features were extracted from the pelvic T2WI of the patients. The least absolute shrinkage and selection operator algorithm and backward stepwise regression method were adopted for feature selection. Three LLN metastasis prediction models were established through logistic regression analysis based on the clinical risk factors and radiomics features. Model performance was assessed in terms of discriminability and decision curve analysis in the training, verification and test sets. RESULTS The model based on the combined T2WI radiomics features and clinical risk factors demonstrated the highest accuracy, surpassing the models based solely on either T2WI radiomics features or clinical risk factors. Specifically, the model achieved an AUC value of 0.836 in the test set. Decision curve analysis revealed that this model had the greatest clinical utility for the vast majority of the threshold probability range from 0.4 to 1.0. CONCLUSION Combining T2WI radiomics features with clinical risk factors holds promise for the noninvasive assessment of the biological characteristics of the LLNs in rectal cancer, potentially aiding in therapeutic decision-making and optimizing patient outcomes.
Collapse
Affiliation(s)
- Hao Yan
- Department of Oncology, Tianjin Union Medical Center, Nankai University, Tianjin, 300121, China
| | - Hongjie Yang
- Nankai University, Tianjin, 300071, China; The Institute of Translational Medicine, Tianjin Union Medical Center of Nankai University, Tianjin, 300121, China; Department of Colorectal Surgery, Tianjin Union Medical Center, Tianjin, 300121, China
| | | | - Longchun Dong
- Department of Radiology, Tianjin Union Medical Center, Tianjin, 300121, China
| | - Zhichun Zhang
- The Institute of Translational Medicine, Tianjin Union Medical Center of Nankai University, Tianjin, 300121, China; Department of Colorectal Surgery, Tianjin Union Medical Center, Tianjin, 300121, China
| | - Yuanda Zhou
- The Institute of Translational Medicine, Tianjin Union Medical Center of Nankai University, Tianjin, 300121, China; Department of Colorectal Surgery, Tianjin Union Medical Center, Tianjin, 300121, China
| | - Qingsheng Zeng
- The Institute of Translational Medicine, Tianjin Union Medical Center of Nankai University, Tianjin, 300121, China; Department of Colorectal Surgery, Tianjin Union Medical Center, Tianjin, 300121, China
| | - Peng Li
- The Institute of Translational Medicine, Tianjin Union Medical Center of Nankai University, Tianjin, 300121, China; Department of Colorectal Surgery, Tianjin Union Medical Center, Tianjin, 300121, China
| | - Yi Sun
- Nankai University, Tianjin, 300071, China; The Institute of Translational Medicine, Tianjin Union Medical Center of Nankai University, Tianjin, 300121, China; Department of Colorectal Surgery, Tianjin Union Medical Center, Tianjin, 300121, China.
| | - Siwei Zhu
- Department of Oncology, Tianjin Union Medical Center, Nankai University, Tianjin, 300121, China; Nankai University, Tianjin, 300071, China; The Institute of Translational Medicine, Tianjin Union Medical Center of Nankai University, Tianjin, 300121, China.
| |
Collapse
|
19
|
Zhang X, Zhang G, Qiu X, Yin J, Tan W, Yin X, Yang H, Liao L, Wang H, Zhang Y. Radiomics under 2D regions, 3D regions, and peritumoral regions reveal tumor heterogeneity in non-small cell lung cancer: a multicenter study. LA RADIOLOGIA MEDICA 2023; 128:1079-1092. [PMID: 37486526 DOI: 10.1007/s11547-023-01676-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Accepted: 06/29/2023] [Indexed: 07/25/2023]
Abstract
PURPOSE Lung cancer has significant genetic and phenotypic heterogeneity, leading to poor prognosis. Radiomic features have emerged as promising predictors of the tumor phenotype. However, the role of underlying information surrounding the cancer remains unclear. MATERIALS AND METHODS We conducted a retrospective study of 508 patients with NSCLC from three institutions. Radiomics models were built using features from six tumor regions and seven classifiers to predict three prognostically significant tumor phenotypes. The models were evaluated and interpreted by the mean area under the receiver operating characteristic curve (AUC) under nested cross-validation and Shapley values. The best-performing predictive models corresponding to six tumor regions and three tumor phenotypes were identified for further comparative analysis. In addition, we designed five experiments with different voxel spacing to assess the sensitivity of the experimental results to the spatial resolution of the voxels. RESULTS Our results demonstrated that models based on 2D, 3D, and peritumoral region features yielded mean AUCs and 95% confidence intervals of 0.759 and [0.747-0.771] for lymphovascular invasion, 0.889 and [0.882-0.896] for pleural invasion, and 0.839 and [0.829-0.849] for T-staging in the testing cohort, which was significantly higher than all other models. Similar results were obtained for the model combining the three regional features at five voxel spacings. CONCLUSION Our study revealed the predictive role of the developed methods with multi-regional features for the preoperative assessment of prognostic factors in NSCLC. The analysis of different voxel spacing and model interpretability strengthens the experimental findings and contributes to understanding the biological significance of the radiological phenotype.
Collapse
Affiliation(s)
- Xingping Zhang
- Cyberspace Institute of Advanced Technology, Guangzhou University, Guangzhou, 510006, China
- Institute for Sustainable Industries and Liveable Cities, Victoria University, Melbourne, VIC, 3011, Australia
- Department of New Networks, Peng Cheng Laboratory, Shenzhen, 518000, China
| | - Guijuan Zhang
- Department of Respiratory and Critical Care, First Affiliated Hospital of Gannan Medical University, Ganzhou, 341000, China
| | - Xingting Qiu
- Department of Radiology, First Affiliated Hospital of Gannan Medical University, Ganzhou, 341000, China
| | - Jiao Yin
- Institute for Sustainable Industries and Liveable Cities, Victoria University, Melbourne, VIC, 3011, Australia
| | - Wenjun Tan
- Key Laboratory of Intelligent Computing in Medical Image, Ministry of Education, Northeastern University, Shenyang, 110189, China
| | - Xiaoxia Yin
- Cyberspace Institute of Advanced Technology, Guangzhou University, Guangzhou, 510006, China
| | - Hong Yang
- Cyberspace Institute of Advanced Technology, Guangzhou University, Guangzhou, 510006, China
| | - Liefa Liao
- School of Information Engineering, Jiangxi University of Science and Technology, Ganzhou, 341000, China
| | - Hua Wang
- Institute for Sustainable Industries and Liveable Cities, Victoria University, Melbourne, VIC, 3011, Australia.
| | - Yanchun Zhang
- Cyberspace Institute of Advanced Technology, Guangzhou University, Guangzhou, 510006, China.
- Institute for Sustainable Industries and Liveable Cities, Victoria University, Melbourne, VIC, 3011, Australia.
- Department of New Networks, Peng Cheng Laboratory, Shenzhen, 518000, China.
| |
Collapse
|
20
|
Zeng C, Zhang W, Liu M, Liu J, Zheng Q, Li J, Wang Z, Sun G. Efficacy of radiomics model based on the concept of gross tumor volume and clinical target volume in predicting occult lymph node metastasis in non-small cell lung cancer. Front Oncol 2023; 13:1096364. [PMID: 37293586 PMCID: PMC10246750 DOI: 10.3389/fonc.2023.1096364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2022] [Accepted: 05/09/2023] [Indexed: 06/10/2023] Open
Abstract
Objective This study aimed to establish a predictive model for occult lymph node metastasis (LNM) in patients with clinical stage I-A non-small cell lung cancer (NSCLC) based on contrast-enhanced CT. Methods A total of 598 patients with stage I-IIA NSCLC from different hospitals were randomized into the training and validation group. The "Radiomics" tool kit of AccuContour software was employed to extract the radiomics features of GTV and CTV from chest-enhanced CT arterial phase pictures. Then, the least absolute shrinkage and selection operator (LASSO) regression analysis was applied to reduce the number of variables and develop GTV, CTV, and GTV+CTV models for predicting occult lymph node metastasis (LNM). Results Eight optimal radiomics features related to occult LNM were finally identified. The receiver operating characteristic (ROC) curves of the three models showed good predictive effects. The area under the curve (AUC) value of GTV, CTV, and GTV+CTV model in the training group was 0.845, 0.843, and 0.869, respectively. Similarly, the corresponding AUC values in the validation group were 0.821, 0.812, and 0.906. The combined GTV+CTV model exhibited a better predictive performance in the training and validation group by the Delong test (p<0.05). Moreover, the decision curve showed that the combined GTV+CTV predictive model was superior to the GTV or CTV model. Conclusion The radiomics prediction models based on GTV and CTV can predict occult LNM in patients with clinical stage I-IIA NSCLC preoperatively, and the combined GTV+CTV model is the optimal strategy for clinical application.
Collapse
Affiliation(s)
- Chao Zeng
- Hebei Key Laboratory of Medical-industrial Integration Precision Medicine, Clinical Medicine College, Affiliated Hospital, North China University of Science and Technology, Tangshan, Hebei, China
| | - Wei Zhang
- Department of Radiotherapy, Yantai Yuhuangding Hospital, The Affiliated Hospital of Qingdao University, Yantai, Shandong, China
| | - Meiyue Liu
- Hebei Key Laboratory of Medical-industrial Integration Precision Medicine, Clinical Medicine College, Affiliated Hospital, North China University of Science and Technology, Tangshan, Hebei, China
| | - Jianping Liu
- Department of Chemoradiation, Tangshan People’s Hospital, Tangshan, Hebei, China
| | - Qiangxin Zheng
- Hebei Key Laboratory of Medical-industrial Integration Precision Medicine, Clinical Medicine College, Affiliated Hospital, North China University of Science and Technology, Tangshan, Hebei, China
| | - Jianing Li
- Hebei Key Laboratory of Medical-industrial Integration Precision Medicine, Clinical Medicine College, Affiliated Hospital, North China University of Science and Technology, Tangshan, Hebei, China
| | - Zhiwu Wang
- Department of Chemoradiation, Tangshan People’s Hospital, Tangshan, Hebei, China
| | - Guogui Sun
- Hebei Key Laboratory of Medical-industrial Integration Precision Medicine, Clinical Medicine College, Affiliated Hospital, North China University of Science and Technology, Tangshan, Hebei, China
| |
Collapse
|
21
|
Mu J, Kuang K, Ao M, Li W, Dai H, Ouyang Z, Li J, Huang J, Guo S, Yang J, Yang L. Deep learning predicts malignancy and metastasis of solid pulmonary nodules from CT scans. Front Med (Lausanne) 2023; 10:1145846. [PMID: 37275359 PMCID: PMC10235703 DOI: 10.3389/fmed.2023.1145846] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Accepted: 04/10/2023] [Indexed: 06/07/2023] Open
Abstract
In the clinic, it is difficult to distinguish the malignancy and aggressiveness of solid pulmonary nodules (PNs). Incorrect assessments may lead to delayed diagnosis and an increased risk of complications. We developed and validated a deep learning-based model for the prediction of malignancy as well as local or distant metastasis in solid PNs based on CT images of primary lesions during initial diagnosis. In this study, we reviewed the data from multiple patients with solid PNs at our institution from 1 January 2019 to 30 April 2022. The patients were divided into three groups: benign, Ia-stage lung cancer, and T1-stage lung cancer with metastasis. Each cohort was further split into training and testing groups. The deep learning system predicted the malignancy and metastasis status of solid PNs based on CT images, and then we compared the malignancy prediction results among four different levels of clinicians. Experiments confirmed that human-computer collaboration can further enhance diagnostic accuracy. We made a held-out testing set of 134 cases, with 689 cases in total. Our convolutional neural network model reached an area under the ROC (AUC) of 80.37% for malignancy prediction and an AUC of 86.44% for metastasis prediction. In observer studies involving four clinicians, the proposed deep learning method outperformed a junior respiratory clinician and a 5-year respiratory clinician by considerable margins; it was on par with a senior respiratory clinician and was only slightly inferior to a senior radiologist. Our human-computer collaboration experiment showed that by simply adding binary human diagnosis into model prediction probabilities, model AUC scores improved to 81.80-88.70% when combined with three out of four clinicians. In summary, the deep learning method can accurately diagnose the malignancy of solid PNs, improve its performance when collaborating with human experts, predict local or distant metastasis in patients with T1-stage lung cancer, and facilitate the application of precision medicine.
Collapse
Affiliation(s)
- Junhao Mu
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Kaiming Kuang
- Dianei Technology, Shanghai, China
- University of California, San Diego, San Diego, CA, United States
| | - Min Ao
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Weiyi Li
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Haiyun Dai
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Zubin Ouyang
- Department of Radiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jingyu Li
- Dianei Technology, Shanghai, China
- School of Computer Science, Wuhan University, Wuhan, China
| | - Jing Huang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Shuliang Guo
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jiancheng Yang
- Dianei Technology, Shanghai, China
- Shanghai Jiao Tong University, Shanghai, China
- École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Li Yang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
22
|
Zheng X, Li R, Fan L, Ge Y, Li W, Feng F. Prognostic predictors of radical resection of stage I-IIIB non-small cell lung cancer: the role of preoperative CT texture features, conventional imaging features, and clinical features in a retrospectively analyzed. BMC Pulm Med 2023; 23:122. [PMID: 37060067 PMCID: PMC10105471 DOI: 10.1186/s12890-023-02422-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2022] [Accepted: 04/03/2023] [Indexed: 04/16/2023] Open
Abstract
BACKGROUND To investigate the value of preoperative computed tomography (CT) texture features, routine imaging features, and clinical features in the prognosis of non-small cell lung cancer (NSCLC) after radical resection. METHODS Demographic parameters and clinically features were analyzed in 107 patients with stage I-IIIB NSCLC, while 73 of these patients received CT scanning and radiomic characteristics for prognosis assessment. Texture analysis features include histogram, gray size area matrix and gray co-occurrence matrix features. The clinical risk features were identified using univariate and multivariate logistic analyses. By incorporating the radiomics score (Rad-score) and clinical risk features with multivariate cox regression, a combined nomogram was built. The nomogram performance was assessed by its calibration, clinical usefulness and Harrell's concordance index (C-index). The 5-year OS between the dichotomized subgroups was compared using Kaplan-Meier (KM) analysis and the log-rank test. RESULTS Consisting of 4 selected features, the radiomics signature showed a favorable discriminative performance for prognosis, with an AUC of 0.91 (95% CI: 0.84 ~ 0.97). The nomogram, consisting of the radiomics signature, N stage, and tumor size, showed good calibration. The nomogram also exhibited prognostic ability with a C-index of 0.91 (95% CI, 0.86-0.95) for OS. The decision curve analysis indicated that the nomogram was clinically useful. According to the KM survival curves, the low-risk group had higher 5-year survival rate compared to high-risk. CONCLUSION The as developed nomogram, combining with preoperative radiomics evidence, N stage, and tumor size, has potential to preoperatively predict the prognosis of NSCLC with a high accuracy and could assist to treatment for the NSCLC patients in the clinic.
Collapse
Affiliation(s)
- Xingxing Zheng
- Department of Radiology, Xi'an Jiaotong University, Xi'an, 710049, China
- Department of Radiology, Baoji Central Hospital, Baoji, 721000, China
- Department of Radiology, Affiliated Tumor Hospital of Nantong University, No. 30 Tongyangbei Road, Tongzhou District, Nantong, 226361, China
| | - Rui Li
- Department of Radiology, Affiliated Tumor Hospital of Nantong University, No. 30 Tongyangbei Road, Tongzhou District, Nantong, 226361, China
| | - Lihua Fan
- Department of Radiology, Affiliated Hospital of Shaanxi University of Traditional Chinese Medicine, Xianyang, 712000, China
| | - Yaqiong Ge
- GE Healthcare China, Shanghai, 210000, China
| | - Wei Li
- Department of Radiology, Baoji Central Hospital, Baoji, 721000, China
| | - Feng Feng
- Department of Radiology, Affiliated Tumor Hospital of Nantong University, No. 30 Tongyangbei Road, Tongzhou District, Nantong, 226361, China.
| |
Collapse
|
23
|
Ma X, Xia L, Chen J, Wan W, Zhou W. Development and validation of a deep learning signature for predicting lymph node metastasis in lung adenocarcinoma: comparison with radiomics signature and clinical-semantic model. Eur Radiol 2023; 33:1949-1962. [PMID: 36169691 DOI: 10.1007/s00330-022-09153-z] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 07/23/2022] [Accepted: 09/08/2022] [Indexed: 11/04/2022]
Abstract
OBJECTIVE To develop and validate a deep learning (DL) signature for predicting lymph node (LN) metastasis in patients with lung adenocarcinoma. METHODS A total of 612 patients with pathologically-confirmed lung adenocarcinoma were retrospectively enrolled and were randomly divided into training cohort (n = 489) and internal validation cohort (n = 123). Besides, 108 patients were enrolled and constituted an independent test cohort (n = 108). Patients' clinical characteristics and CT semantic features were collected. The radiomics features were derived from contrast-enhanced CT images. The clinical-semantic model and radiomics signature were built to predict LN metastasis. Furthermore, Swin Transformer was adopted to develop a DL signature predictive of LN metastasis. Model performance was evaluated by area under the receiver operating characteristic curve (AUC), sensitivity, specificity, calibration curve, and decision curve analysis. The comparisons of AUC were conducted by the DeLong test. RESULTS The proposed DL signature yielded an AUC of 0.948-0.961 across all three cohorts, significantly superior to both clinical-semantic model and radiomics signature (all p < 0.05). The calibration curves show that DL signature predicted probabilities fit well the actual observed probabilities of LN metastasis. DL signature gained a higher net benefit than both clinical-semantic model and radiomics signature. The incorporation of radiomics signature or clinical-semantic risk predictors failed to reveal an incremental value over the DL signature. CONCLUSIONS The proposed DL signature based on Swin Transformer achieved a promising performance in predicting LN metastasis and could confer important information in noninvasive mediastinal LN staging and individualized therapeutic options. KEY POINTS • Accurate prediction for lymph node metastasis is crucial to formulate individualized therapeutic options for patients with lung adenocarcinoma. • The deep learning signature yielded an AUC of 0.948-0.961 across all three cohorts in predicting lymph node metastasis, superior to both radiomics signature and clinical-semantic model. • The incorporation of radiomics signature or clinical-semantic risk predictors into deep learning signature failed to reveal an incremental value over deep learning signature.
Collapse
Affiliation(s)
- Xiaoling Ma
- Medical Imaging Center, People's Hospital of Ningxia Hui Autonomous Region, Yinchuan, China
| | - Liming Xia
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Road, Qiaokou District, Wuhan, 430030, Hubei, China.
| | | | - Weijia Wan
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Road, Qiaokou District, Wuhan, 430030, Hubei, China
| | - Wen Zhou
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Road, Qiaokou District, Wuhan, 430030, Hubei, China
| |
Collapse
|
24
|
Hu D, Li S, Wu N, Lu X. A Multi-Modal Heterogeneous Graph Forest to Predict Lymph Node Metastasis of Non-Small Cell Lung Cancer. IEEE J Biomed Health Inform 2023; 27:1216-1224. [PMID: 37018304 DOI: 10.1109/jbhi.2022.3233387] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Lymph node metastasis (LNM) is critical for treatment decision-making for cancer patients, but it is difficult to diagnose accurately before surgery. Machine learning can learn nontrivial knowledge from multi-modal data to support accurate diagnosis. In this paper, we proposed a Multi-modal Heterogeneous Graph Forest (MHGF) approach to extract the deep representations of LNM from multi-modal data. Specifically, we first extracted the deep image features from CT images to represent the pathological anatomic extent of the primary tumor (pathological T stage) using a ResNet-Trans network. And then, a heterogeneous graph with six vertices and seven bi-directional relations was defined by medical experts to describe the possible relations between the clinical and image features. After that, we proposed a graph forest approach to construct the sub-graphs by removing each vertex in the complete graph iteratively. Finally, we used graph neural networks to learn the representations of each sub-graph in the forest to predict LNM and averaged all the prediction results as final results. We conducted experiments on 681 patients' multi-modal data. The proposed MHGF achieves the best performances with a 0.806 AUC value and 0.513 AP value compared with state-of-art machine learning and deep learning methods. The results indicate that the graph method can explore the relations between different types of features to learn effective deep representations for LNM prediction. Moreover, we found that the deep image features about the pathological anatomic extent of the primary tumor are useful for LNM prediction. And the graph forest approach can further improve the generalization ability and stability of the LNM prediction model.
Collapse
|
25
|
Chen QL, Li MM, Xue T, Peng H, Shi J, Li YY, Duan SF, Feng F. Radiomics nomogram integrating intratumoural and peritumoural features to predict lymph node metastasis and prognosis in clinical stage IA non-small cell lung cancer: a two-centre study. Clin Radiol 2023; 78:e359-e367. [PMID: 36858926 DOI: 10.1016/j.crad.2023.02.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 01/24/2023] [Accepted: 02/03/2023] [Indexed: 02/18/2023]
Abstract
AIM To investigate the value of a radiomics nomogram integrating intratumoural and peritumoural features in predicting lymph node metastasis and overall survival (OS) in patients with clinical stage IA non-small-cell lung cancer (NSCLC). MATERIALS AND METHODS This study retrospectively enrolled 199 patients (training cohort: 71 patients from Affiliated Tumour Hospital of Nantong University; internal validation cohort: 46 patients from Affiliated Tumour Hospital of Nantong University; external validation cohort: 82 patients from the public database). CT radiomics models were constructed based on four volumes of interest: gross tumour volume (GTV), gross and 3 mm peritumoural volume (GPTV3), gross and 6 mm peritumoural volume (GPTV6), and gross and 9 mm peritumoural volume (GPTV9). The optimal radiomics signature was further combined with independent clinical predictors to develop a nomogram. Univariable and multivariable Cox regression analysis were applied to determine the relationship between factors and OS. RESULTS GPTV6 radiomics yielded better performance than GTV, GPTV3, and, GPTV9 radiomics in the training (area under the curve [AUC], 0.81), internal validation (AUC, 0.79), and external validation cohorts (AUC, 0.71), respectively. The nomogram integrating GPTV6 radiomics and spiculation improved predictive ability, with AUCs of 0.85, 0.80, and 0.74 in three cohorts, respectively. Pathological lymph node metastasis, nomogram-predicted lymph node metastasis, and pleural indentation were independent risk predictors of OS (p<0.05). CONCLUSIONS The nomogram integrating GPTV6 radiomics features and independent clinical predictors performed well in predicting lymph node metastasis and prognosis in patients with clinical stage IA NSCLC.
Collapse
Affiliation(s)
- Q-L Chen
- Department of Radiology, Affiliated Tumour Hospital of Nantong University, Nantong, Jiangsu 226001, PR China
| | - M-M Li
- Department of Radiology, Affiliated Tumour Hospital of Nantong University, Nantong, Jiangsu 226001, PR China
| | - T Xue
- Department of Radiology, Affiliated Tumour Hospital of Nantong University, Nantong, Jiangsu 226001, PR China
| | - H Peng
- Department of Radiology, Affiliated Tumour Hospital of Nantong University, Nantong, Jiangsu 226001, PR China
| | - J Shi
- Department of Radiology, Affiliated Tumour Hospital of Nantong University, Nantong, Jiangsu 226001, PR China
| | - Y-Y Li
- Department of Radiology, Affiliated Tumour Hospital of Nantong University, Nantong, Jiangsu 226001, PR China
| | - S-F Duan
- GE Healthcare China, Shanghai City 210000, China
| | - F Feng
- Department of Radiology, Affiliated Tumour Hospital of Nantong University, Nantong, Jiangsu 226001, PR China.
| |
Collapse
|
26
|
Shimada Y, Kudo Y, Maehara S, Fukuta K, Masuno R, Park J, Ikeda N. Artificial intelligence-based radiomics for the prediction of nodal metastasis in early-stage lung cancer. Sci Rep 2023; 13:1028. [PMID: 36658301 PMCID: PMC9852472 DOI: 10.1038/s41598-023-28242-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2022] [Accepted: 01/16/2023] [Indexed: 01/20/2023] Open
Abstract
We aimed to investigate the value of computed tomography (CT)-based radiomics with artificial intelligence (AI) in predicting pathological lymph node metastasis (pN) in patients with clinical stage 0-IA non-small cell lung cancer (c-stage 0-IA NSCLC). This study enrolled 720 patients who underwent complete surgical resection for c-stage 0-IA NSCLC, and were assigned to the derivation and validation cohorts. Using the AI software Beta Version (Fujifilm Corporation, Japan), 39 AI imaging factors, including 17 factors from the AI ground-glass nodule analysis and 22 radiomics features from nodule characterization analysis, were extracted to identify factors associated with pN. Multivariate analysis showed that clinical stage IA3 (p = 0.028), solid-part size (p < 0.001), and average solid CT value (p = 0.033) were independently associated with pN. The receiver operating characteristic analysis showed that the area under the curve and optimal cut-off values of the average solid CT value relevant to pN were 0.761 and -103 Hounsfield units, and the threshold provided sensitivity, specificity, and negative predictive values of 69%, 65%, and 94% in the entire cohort, respectively. Measuring the average solid-CT value of tumors for pN may have broad applications such as guiding individualized surgical approaches and postoperative treatment.
Collapse
Affiliation(s)
- Yoshihisa Shimada
- Department of Thoracic Surgery, Tokyo Medical University, 6-7-1 Nishishinjuku, Shinjuku-Ku, Tokyo, 160-0023, Japan.
| | - Yujin Kudo
- Department of Thoracic Surgery, Tokyo Medical University, Tokyo, Japan
| | - Sachio Maehara
- Department of Thoracic Surgery, Tokyo Medical University, Tokyo, Japan
| | - Kentaro Fukuta
- Department of Thoracic Surgery, Tokyo Medical University, Tokyo, Japan
| | - Ryuhei Masuno
- Department of Radiology, Tokyo Medical University, Tokyo, Japan
| | - Jinho Park
- Department of Radiology, Tokyo Medical University, Tokyo, Japan
| | - Norihiko Ikeda
- Department of Thoracic Surgery, Tokyo Medical University, Tokyo, Japan
| |
Collapse
|
27
|
Ge G, Zhang J. Feature selection methods and predictive models in CT lung cancer radiomics. J Appl Clin Med Phys 2023; 24:e13869. [PMID: 36527376 PMCID: PMC9860004 DOI: 10.1002/acm2.13869] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Revised: 08/31/2022] [Accepted: 11/28/2022] [Indexed: 12/23/2022] Open
Abstract
Radiomics is a technique that extracts quantitative features from medical images using data-characterization algorithms. Radiomic features can be used to identify tissue characteristics and radiologic phenotyping that is not observable by clinicians. A typical workflow for a radiomics study includes cohort selection, radiomic feature extraction, feature and predictive model selection, and model training and validation. While there has been increasing attention given to radiomic feature extraction, standardization, and reproducibility, currently, there is a lack of rigorous evaluation of feature selection methods and predictive models. Herein, we review the published radiomics investigations in CT lung cancer and provide an overview of the commonly used radiomic feature selection methods and predictive models. We also compare limitations of various methods in clinical applications and present sources of uncertainty associated with those methods. This review is expected to help raise awareness of the impact of radiomic feature and model selection methods on the integrity of radiomics studies.
Collapse
Affiliation(s)
- Gary Ge
- Department of Radiology, University of Kentucky, Lexington, Kentucky, USA
| | - Jie Zhang
- Department of Radiology, University of Kentucky, Lexington, Kentucky, USA
| |
Collapse
|
28
|
Radiomics Signature Using Manual Versus Automated Segmentation for Lymph Node Staging of Bladder Cancer. Eur Urol Focus 2023; 9:145-153. [PMID: 36115774 DOI: 10.1016/j.euf.2022.08.015] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Revised: 07/26/2022] [Accepted: 08/30/2022] [Indexed: 11/23/2022]
Abstract
BACKGROUND Bladder cancer (BC) treatment algorithms depend on accurate tumor staging. To date, computed tomography (CT) is recommended for assessment of lymph node (LN) metastatic spread in muscle-invasive and high-risk BC. However, the diagnostic efficacy of radiologist-evaluated CT imaging studies is limited. OBJECTIVE To evaluate the performance of quantitative radiomics signatures for detection of LN metastases in BC. DESIGN, SETTING, AND PARTICIPANTS Of 1354 patients with BC who underwent radical cystectomy (RC) with lymphadenectomy who were screened, 391 with pathological nodal staging (pN0: n = 297; pN+: n = 94) were included and randomized into training (n = 274) and test (n = 117) cohorts. Pelvic LNs were segmented manually and automatically. A total of 1004 radiomics features were extracted from each LN and a machine learning model was trained to assess pN status using histopathology labels as the ground truth. OUTCOME MEASUREMENTS AND STATISTICAL ANALYSIS Radiologist assessment was compared to radiomics-based analysis using manual and automated LN segmentations for detection of LN metastases in BC. Statistical analysis was performed using the receiver operating characteristics curve method and evaluated in terms of sensitivity, specificity, and area under the curve (AUC). RESULTS AND LIMITATIONS In total, 1845 LNs were manually segmented. Automated segmentation correctly located 361/557 LNs in the test cohort. Manual and automatic masks achieved an AUC of 0.80 (95% confidence interval [CI] 0.69-0.91; p = 0.64) and 0.70 (95% CI: 0.58-0.82; p = 0.17), respectively, in the test cohort compared to radiologist assessment, with an AUC of 0.78 (95% CI 0.67-0.89). A combined model of a manually segmented radiomics signature and radiologist assessment reached an AUC of 0.81 (95% CI 0.71-0.92; p = 0.63). CONCLUSIONS A radiomics signature allowed discrimination of nodal status with high diagnostic accuracy. The model based on manual LN segmentation outperformed the fully automated approach. PATIENT SUMMARY For patients with bladder cancer, evaluation of computed tomography (CT) scans before surgery using a computer-based method for image analysis, called radiomics, may help in standardizing and improving the accuracy of assessment of lymph nodes. This could be a valuable tool for optimizing treatment options.
Collapse
|
29
|
Ma ZP, Li XL, Gao K, Zhang TL, Wang HD, Zhao YX. Application of radiomics based on chest CT-enhanced dual-phase imaging in the immunotherapy of non-small cell lung cancer. JOURNAL OF X-RAY SCIENCE AND TECHNOLOGY 2023; 31:1333-1340. [PMID: 37840466 DOI: 10.3233/xst-230189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/17/2023]
Abstract
OBJECTIVE To explore the value of applying computed tomography (CT) radiomics based on different CT-enhanced phases to determine the immunotherapeutic efficacy of non-small cell lung cancer (NSCLC). METHODS 106 patients with NSCLC who underwent immunotherapy are randomly divided into training (74) and validation (32) groups. CT-enhanced arterial and venous phase images of patients before treatment are collected. Region-of-interest (ROI) is segmented on the CT-enhanced images, and the radiomic features are extracted. One-way analysis of variance and least absolute shrinkage and selection operator (LASSO) are used to screen the optimal radiomics features and analyze the association between radiomics features and immunotherapy efficacy. The area under receiver-operated characteristic curves (AUC) along with the sensitivity and specificity are computed to evaluate diagnostic effectiveness. RESULTS LASSO regression analysis screens and selects 6 and 8 optimal features in the arterial and venous phases images, respectively. Applying to the training group, AUCs based on CT-enhanced arterial and venous phase images are 0.867 (95% CI:0.82-0.94) and 0.880 (95% CI:0.86-0.91) with the sensitivities of 73.91% and 76.19%, and specificities of 66.67% and 72.19%, respectively, while in validation group, AUCs of the arterial and venous phase images are 0.732 (95% CI:0.71-0.78) and 0.832 (95% CI:0.78-0.91) with sensitivities of 75.00% and 76.00%, and specificities of 73.07% and 75.00%, respectively. There are no significant differences between AUC values computed from arterial phases and venous phases images in both training and validation groups (P < 0.05). CONCLUSION The optimally selected radiomics features computed from CT-enhanced different-phase images can provide new imaging marks to evaluate efficacy of the targeted therapy in NSCLC with a high diagnostic value.
Collapse
Affiliation(s)
- Ze-Peng Ma
- Department of Radiology, Affiliated Hospital of Hebei University; Clinical Medical college, Hebei University, Baoding, Hebei Province, China
| | - Xiao-Lei Li
- Hebei Provincial Center for Disease Control and Prevention, Shijiazhuang City, Hebei Province, China
| | - Kai Gao
- Department of Radiology, Affiliated Hospital of Hebei University; Clinical Medical college, Hebei University, Baoding, Hebei Province, China
| | - Tian-Le Zhang
- Department of Radiology, Affiliated Hospital of Hebei University; Clinical Medical college, Hebei University, Baoding, Hebei Province, China
| | - Heng-Di Wang
- Department of Radiology, Affiliated Hospital of Hebei University; Clinical Medical college, Hebei University, Baoding, Hebei Province, China
| | - Yong-Xia Zhao
- Department of Radiology, Affiliated Hospital of Hebei University; Clinical Medical college, Hebei University, Baoding, Hebei Province, China
| |
Collapse
|
30
|
Wang M, Liu L, Dai Q, Jin M, Huang G. Developing a primary tumor and lymph node 18F-FDG PET/CT-clinical (TLPC) model to predict lymph node metastasis of resectable T2-4 NSCLC. J Cancer Res Clin Oncol 2023; 149:247-261. [PMID: 36565319 PMCID: PMC9889531 DOI: 10.1007/s00432-022-04545-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Accepted: 12/16/2022] [Indexed: 12/25/2022]
Abstract
PURPOSE The goal of this study was to investigate whether the combined PET/CT radiomic features of the primary tumor and lymph node could predict lymph node metastasis (LNM) of resectable non-small cell lung cancer (NSCLC) in stage T2-4. METHODS This retrospective study included 192 NSCLC patients who underwent tumor and node dissection between August 2016 and December 2017 and underwent 18F-fluorodeoxyglucose (18F-FDG) PET/CT scanning 1-3 weeks before surgery. In total, 192 primary tumors (> 3 cm) and 462 lymph nodes (LN > 0.5 cm) were analyzed. The pretreatment clinical features of these patients were recorded, and the radiomic features of their primary tumor and lymph node were extracted from PET/CT imaging. The Spearman's relevance combined with the least absolute shrinkage and selection operator was used for radiomic feature selection. Five independent machine learning models (multi-layer perceptron, extreme Gradient Boosting, light gradient boosting machine, gradient boosting decision tree, and support vector machine) were tested as classifiers for model development. We developed the following three models to predict LNM: tumor PET/CT-clinical (TPC), lymph PET/CT-clinical (LPC), and tumor and lymph PET/CT-clinical (TLPC). The performance of the models and the clinical node (cN) staging was evaluated using the ROC curve and confusion matrix analysis. RESULTS The ROC analysis showed that among the three models, the TLPC model had better predictive clinical utility and efficiency in predicting LNM of NSCLC (AUC = 0.93, accuracy = 85%; sensitivity = 0.93; specificity = 0.75) than both the TPC model (AUC = 0.54, accuracy = 50%; specificity = 0.38; sensitivity = 0.59) and the LPC model (AUC = 0.82, accuracy = 70%; specificity = 0.41; sensitivity = 0.92). The TLPC model also exhibited great potential in predicting the N2 stage in NSCLC (AUC = 0.94, accuracy = 79%; specificity = 0.64; sensitivity = 0.91). CONCLUSION The combination of CT and PET radiomic features of the primary tumor and lymph node showed great potential for predicting LNM of resectable T2-4 NSCLC. The TLPC model can non-invasively predict lymph node metastasis in NSCLC, which may be helpful for clinicians to develop more rational therapeutic strategies.
Collapse
Affiliation(s)
- Meng Wang
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai, 200093 China ,Shanghai Key Laboratory of Molecular Imaging, Jiading District Central Hospital Affiliated Shanghai University of Medicine and Health Sciences, Shanghai, 201318 China
| | - Liu Liu
- Department of Nuclear Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, 200003 China
| | - Qian Dai
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai, 200093 China ,Shanghai Key Laboratory of Molecular Imaging, Jiading District Central Hospital Affiliated Shanghai University of Medicine and Health Sciences, Shanghai, 201318 China
| | - Mingming Jin
- Shanghai Key Laboratory of Molecular Imaging, Jiading District Central Hospital Affiliated Shanghai University of Medicine and Health Sciences, Shanghai, 201318 China
| | - Gang Huang
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai, 200093 China ,Shanghai Key Laboratory of Molecular Imaging, Jiading District Central Hospital Affiliated Shanghai University of Medicine and Health Sciences, Shanghai, 201318 China , Shanghai Key Laboratory of Molecular Imaging, Zhoupu Hospital, Shanghai University of Medicine and Health Sciences, Shanghai, 201318 China
| |
Collapse
|
31
|
Shimada Y, Kudo Y, Maehara S, Amemiya R, Masuno R, Park J, Ikeda N. Radiomics with Artificial Intelligence for the Prediction of Early Recurrence in Patients with Clinical Stage IA Lung Cancer. Ann Surg Oncol 2022; 29:8185-8193. [PMID: 36070112 DOI: 10.1245/s10434-022-12516-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Accepted: 08/09/2022] [Indexed: 01/10/2023]
Abstract
BACKGROUND We seek to explore the ability of computed tomography (CT)-based radiomics coupled with artificial intelligence (AI) to predict early recurrence (< 2 years after surgery) in patients with clinical stage 0-IA non-small cell lung cancer (c-stage 0-IA NSCLC). PATIENTS AND METHODS Data of 642 patients were collected for early recurrence and assigned to the derivation and validation cohorts at a ratio of 2:1. Using the AI software Beta Version (Fujifilm Corporation, Japan), 39 AI imaging factors, including 17 factors from the AI ground-glass nodule analysis and 22 radiomic features from nodule characterization analysis, were extracted. RESULTS Multivariate analysis showed that male sex (p = 0.016), solid part size (p < 0.001), CT value standard deviation (p = 0.038), solid part volume ratio (p = 0.016), and bronchus translucency (p = 0.007) were associated with recurrence-free survival (RFS). Receiver operating characteristics analysis showed that the area under the curve and optimal cutoff values relevant to recurrence were 0.707 and 1.49 cm for solid part size, and 0.710 and 22.9% for solid part volume ratio, respectively. The 5-year RFS rates for patients in the validation set with solid part size ≤ 1.49 cm and > 1.49 cm were 92.2% and 70.4% (p < 0.001), whereas those for patients with solid part volume ratios ≤ 22.9% and > 22.9% were 97.8% and 71.7% (p < 0.001), respectively. CONCLUSIONS CT-based radiomics coupled with AI contributes to the noninvasive prediction of early recurrence in patients with c-stage 0-IA NSCLC.
Collapse
Affiliation(s)
- Yoshihisa Shimada
- Department of Thoracic Surgery, Tokyo Medical University, Tokyo, Japan.
| | - Yujin Kudo
- Department of Thoracic Surgery, Tokyo Medical University, Tokyo, Japan
| | - Sachio Maehara
- Department of Thoracic Surgery, Tokyo Medical University, Tokyo, Japan
| | - Ryosuke Amemiya
- Department of Thoracic Surgery, Tokyo Medical University, Tokyo, Japan
| | - Ryuhei Masuno
- Department of Radiology, Tokyo Medical University, Tokyo, Japan
| | - Jinho Park
- Department of Radiology, Tokyo Medical University, Tokyo, Japan
| | - Norihiko Ikeda
- Department of Thoracic Surgery, Tokyo Medical University, Tokyo, Japan
| |
Collapse
|
32
|
Zhao K, Jiang B, Zhang S, Zhang L, Zhang L, Feng Y, Li J, Zhang Y, Xie X. Measurement Accuracy and Repeatability of RECIST-Defined Pulmonary Lesions and Lymph Nodes in Ultra-Low-Dose CT Based on Deep Learning Image Reconstruction. Cancers (Basel) 2022; 14:5016. [PMID: 36291800 PMCID: PMC9599467 DOI: 10.3390/cancers14205016] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Accepted: 10/11/2022] [Indexed: 08/16/2023] Open
Abstract
BACKGROUND Deep learning image reconstruction (DLIR) improves image quality. We aimed to compare the measured diameter of pulmonary lesions and lymph nodes between DLIR-based ultra-low-dose CT (ULDCT) and contrast-enhanced CT. METHODS The consecutive adult patients with noncontrast chest ULDCT (0.07-0.14 mSv) and contrast-enhanced CT (2.38 mSv) were prospectively enrolled. Patients with poor image quality and body mass index ≥ 30 kg/m2 were excluded. The diameter of pulmonary target lesions and lymph nodes defined by Response Evaluation Criteria in Solid Tumors (RECIST) was measured. The measurement variability between ULDCT and enhanced CT was evaluated by Bland-Altman analysis. RESULTS The 141 enrolled patients (62 ± 12 years) had 89 RECIST-defined measurable pulmonary target lesions (including 30 malignant lesions, mainly adenocarcinomas) and 45 measurable mediastinal lymph nodes (12 malignant). The measurement variation of pulmonary lesions between high-strength DLIR (DLIR-H) images of ULDCT and contrast-enhanced CT was 2.2% (95% CI: 1.7% to 2.6%) and the variation of lymph nodes was 1.4% (1.0% to 1.9%). CONCLUSIONS The measured diameters of pulmonary lesions and lymph nodes in DLIR-H images of ULDCT are highly close to those of contrast-enhanced CT. DLIR-based ULDCT may facilitate evaluating target lesions with greatly reduced radiation exposure in tumor evaluation and lung cancer screening.
Collapse
Affiliation(s)
- Keke Zhao
- Radiology Department, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Haining Rd.100, Shanghai 200080, China
| | - Beibei Jiang
- Radiology Department, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Haining Rd.100, Shanghai 200080, China
| | - Shuai Zhang
- CT Imaging Research Center, GE Healthcare China, Shanghai 201203, China
| | - Lu Zhang
- Radiology Department, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Haining Rd.100, Shanghai 200080, China
| | - Lin Zhang
- Radiology Department, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Haining Rd.100, Shanghai 200080, China
| | - Yan Feng
- Radiology Department, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Haining Rd.100, Shanghai 200080, China
| | - Jianying Li
- CT Imaging Research Center, GE Healthcare China, Shanghai 201203, China
| | - Yaping Zhang
- Radiology Department, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Haining Rd.100, Shanghai 200080, China
| | - Xueqian Xie
- Radiology Department, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Haining Rd.100, Shanghai 200080, China
| |
Collapse
|
33
|
Xie D, Xu F, Zhu W, Pu C, Huang S, Lou K, Wu Y, Huang D, He C, Hu H. Delta radiomics model for the prediction of progression-free survival time in advanced non-small-cell lung cancer patients after immunotherapy. Front Oncol 2022; 12:990608. [PMID: 36276082 PMCID: PMC9583844 DOI: 10.3389/fonc.2022.990608] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2022] [Accepted: 09/22/2022] [Indexed: 11/22/2022] Open
Abstract
Objective To assess the validity of pre- and posttreatment computed tomography (CT)-based radiomics signatures and delta radiomics signatures for predicting progression-free survival (PFS) in stage III-IV non-small-cell lung cancer (NSCLC) patients after immune checkpoint inhibitor (ICI) therapy. Methods Quantitative image features of the largest primary lung tumours were extracted on CT-enhanced imaging at baseline (time point 0, TP0) and after the 2nd-3rd immunotherapy cycles (time point 1, TP1). The critical features were selected to construct TP0, TP1 and delta radiomics signatures for the risk stratification of patient survival after ICI treatment. In addition, a prediction model integrating the clinicopathologic risk characteristics and phenotypic signature was developed for the prediction of PFS. Results The C-index of TP0, TP1 and delta radiomics models in the training and validation cohort were 0.64, 0.75, 0.80, and 0.61, 0.68, 0.78, respectively. The delta radiomics score exhibited good accuracy for distinguishing patients with slow and rapid progression to ICI treatment. The predictive accuracy of the combined prediction model was higher than that of the clinical prediction model in both training and validation sets (P<0.05), with a C-index of 0.83 and 0.70, respectively. Additionally, the delta radiomics model (C-index of 0.86) had a higher predictive accuracy compared to PD-L1 expression (C-index of 0.50) (P<0.0001). Conclusions The combined prediction model including clinicopathologic characteristics (tumour anatomical classification and brain metastasis) and the delta radiomics signature could achieve the individualized prediction of PFS in ICIs-treated NSCLC patients.
Collapse
Affiliation(s)
- Dong Xie
- Department of Radiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Department of Radiology, Shaoxing Second Hospital, Shaoxing, China
| | - Fangyi Xu
- Department of Radiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Wenchao Zhu
- Department of Radiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Cailing Pu
- Department of Radiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Shaoyu Huang
- Department of Radiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Department of Radiology, Ningbo Medical Center LiHuili Hospital, Ningbo, China
| | - Kaihua Lou
- Department of Radiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yan Wu
- Department of Radiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Dingpin Huang
- Department of Radiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Cong He
- Department of Radiology, Shaoxing Second Hospital, Shaoxing, China
| | - Hongjie Hu
- Department of Radiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- *Correspondence: Hongjie Hu,
| |
Collapse
|
34
|
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
|
35
|
Chen L, Yu L, Li X, Tian Z, Lin X. Value of CT Radiomics and Clinical Features in Predicting Bone Metastases in Patients with NSCLC. CONTRAST MEDIA & MOLECULAR IMAGING 2022; 2022:7642511. [PMID: 36051936 PMCID: PMC9424036 DOI: 10.1155/2022/7642511] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 06/17/2022] [Accepted: 07/20/2022] [Indexed: 11/23/2022]
Abstract
Objective To explore the CT radiomic features and clinical imaging features of the primary tumor in patients with nonsmall cell lung cancer (NSCLC) before treatment and their predictive value for the occurrence of bone metastases. Methods From June 2017 to June 2021, 195 patients with NSCLC who were pathologically diagnosed without any treatment in the Cancer Hospital Affiliated to Hainan Medical College were retrospectively analyzed, and they were divided into a bone metastasis group and a nonbone metastasis group. The relationship between clinical imaging features and bone metastasis in patients was analyzed by the t-test, rank sum test, and χ 2 test. At the same time, ITK software was used to extract the radiomic characteristics of the primary tumor of the patients, and the patients were randomly divided into a training group and a validation group in a ratio of 7 : 3. The training model was validated in the validation group, and the performance of the model for predicting bone metastases in NSCLC patients was verified by the ROC curve, and a multivariate logistic regression prediction model was established based on the omics parameters extracted from the best prediction model combined with clinical image features. Results Seven features were screened from the primary tumor by LASSO to establish a model for predicting metastasis. The area under the curve was 0.82 and 0.73 in the training and validation sets. The best omics signature and univariate analysis suggested clinical imaging factors (P < 0.05) associated with bone metastases were included in multivariate binary logistic analysis to obtain clinical characteristics of the primary tumor such as gender (OR = 0.141, 95% CI: 0.022-0.919, P = 0.04), increased Cyfra21-1 (OR = 0.12, 95% CI: 0.018-0.782, P = 0.027), Fe content in blood (OR = 0.774, 95% CI: 0.626-0.958, P = 0.018), CT signs such as lesion homogeneity (OR = 0.052, 95% CI: 0.006-0.419, P = 0.006), pleural indentation sign (OR = 0.007, 95% CI: 0.001-0.696, P = 0.034), and omics characteristics glszm_Small Area High Gray Level Emphasis (OR = 0.016, 95% CI: 0.001-0.286, P = 0.005) were independent risk factors for bone metastasis in patients. Conclusion The prediction model established based on radiomics and clinical imaging features has high predictive performance for the occurrence of bone metastasis in NSCLC patients.
Collapse
Affiliation(s)
- Lu Chen
- Hainan Cancer Hospital (Affiliated Cancer Hospital of Hainan Medical College), Nuclear Medicine Department, Haikou, China
| | - Lijuan Yu
- Hainan Cancer Hospital (Affiliated Cancer Hospital of Hainan Medical College), Nuclear Medicine Department, Haikou, China
| | - Xueyan Li
- Hainan Cancer Hospital (Affiliated Cancer Hospital of Hainan Medical College), Nuclear Medicine Department, Haikou, China
| | - Zhanyu Tian
- College of Bioinformatics, Hainan Medical University, Haikou, China
| | - Xiuyan Lin
- Hainan Cancer Hospital (Affiliated Cancer Hospital of Hainan Medical College), Nuclear Medicine Department, Haikou, China
| |
Collapse
|
36
|
Prediction of Two-Year Recurrence-Free Survival in Operable NSCLC Patients Using Radiomic Features from Intra- and Size-Variant Peri-Tumoral Regions on Chest CT Images. Diagnostics (Basel) 2022; 12:diagnostics12061313. [PMID: 35741123 PMCID: PMC9221791 DOI: 10.3390/diagnostics12061313] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 05/13/2022] [Accepted: 05/20/2022] [Indexed: 02/04/2023] Open
Abstract
To predict the two-year recurrence-free survival of patients with non-small cell lung cancer (NSCLC), we propose a prediction model using radiomic features of the inner and outer regions of the tumor. The intratumoral region and the peritumoral regions from the boundary to 3 cm were used to extract the radiomic features based on the intensity, texture, and shape features. Feature selection was performed to identify significant radiomic features to predict two-year recurrence-free survival, and patient classification was performed into recurrence and non-recurrence groups using SVM and random forest classifiers. The probability of two-year recurrence-free survival was estimated with the Kaplan–Meier curve. In the experiment, CT images of 217 non-small-cell lung cancer patients at stages I-IIIA who underwent surgical resection at the Veterans Health Service Medical Center (VHSMC) were used. Regarding the classification performance on whole tumors, the combined radiomic features for intratumoral and peritumoral regions of 6 mm and 9 mm showed improved performance (AUC 0.66, 0.66) compared to T stage and N stage (AUC 0.60), intratumoral (AUC 0.64) and peritumoral 6 mm and 9 mm classifiers (AUC 0.59, 0.62). In the assessment of the classification performance according to the tumor size, combined regions of 21 mm and 3 mm were significant when predicting outcomes compared to other regions of tumors under 3 cm (AUC 0.70) and 3 cm~5 cm (AUC 0.75), respectively. For tumors larger than 5 cm, the combined 3 mm region was significant in predictions compared to the other features (AUC 0.71). Through this experiment, it was confirmed that peritumoral and combined regions showed higher performance than the intratumoral region for tumors less than 5 cm in size and that intratumoral and combined regions showed more stable performance than the peritumoral region in tumors larger than 5 cm.
Collapse
|
37
|
Liang G, Yu W, Liu SQ, Xie MG, Liu M. The value of radiomics based on dual-energy CT for differentiating benign from malignant solitary pulmonary nodules. BMC Med Imaging 2022; 22:95. [PMID: 35597900 PMCID: PMC9123722 DOI: 10.1186/s12880-022-00824-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Accepted: 05/12/2022] [Indexed: 12/24/2022] Open
Abstract
OBJECTIVE To investigate the value of monochromatic dual-energy CT (DECT) images based on radiomics in differentiating benign from malignant solitary pulmonary nodules. MATERIALS AND METHODS This retrospective study was approved by the institutional review board, and informed consent was waived. Pathologically confirmed lung nodules smaller than 3 cm with integrated arterial phase and venous phase (AP and VP) gemstone spectral imaging were retrospectively identified. After extracting the radiomic features of each case, principal component analysis (PCA) was used for feature selection, and after training with the logistic regression method, three classification models (ModelAP, ModelVP and ModelCombination) were constructed. The performance was assessed by the area under the receiver operating curve (AUC), and the efficacy of the models was validated using an independent cohort. RESULTS A total of 153 patients were included and divided into a training cohort (n = 107) and a validation cohort (n = 46). A total of 1130 radiomic features were extracted from each case. The PCA method selected 22, 25 and 35 principal components to construct the three models. The diagnostic accuracy of ModelAP, ModelVP and ModelCombination was 0.8043, 0.6739, and 0.7826 in the validation set, with AUCs of 0.8148 (95% CI 0.682-0.948), 0.7485 (95% CI 0.602-0.895), and 0.8772 (95% CI 0.780-0.974), respectively. The DeLong test showed that there were significant differences in the AUCs between ModelAP and ModelCombination (P = 0.0396) and between ModelVP and ModelCombination (P = 0.0465). However, the difference in AUCs between ModelAP and ModelVP was not significant (P = 0.5061). These results demonstrate that ModelCombination shows a better performance than the other models. Decision curve analysis proved the clinical utility of this model. CONCLUSIONS We developed a radiomics model based on monochromatic DECT images to identify solitary pulmonary nodules. This model could serve as an effective tool for discriminating benign from malignant pulmonary nodules in patients. The combination of arterial phase and venous phase imaging could significantly improve the model performance.
Collapse
Affiliation(s)
- Gao Liang
- Department of Radiology, Hospital of ChengDu University of Traditional Chinese Medicine, Chengdu, 610075, China
| | - Wei Yu
- Department of Radiology, Hospital of ChengDu University of Traditional Chinese Medicine, Chengdu, 610075, China
| | - Shu-Qin Liu
- Department of Radiology, Hospital of ChengDu University of Traditional Chinese Medicine, Chengdu, 610075, China
| | - Ming-Guo Xie
- Department of Radiology, Hospital of ChengDu University of Traditional Chinese Medicine, Chengdu, 610075, China.
| | - Min Liu
- Toxicology Department, WestChina-Frontier PharmaTech Co., Ltd. (WCFP), Chengdu, 610075, China
| |
Collapse
|
38
|
Li W, Zhang H, Ren L, Zou Y, Tian F, Ji X, Li Q, Wang W, Ma G, Xia S. Radiomics of dual-energy computed tomography for predicting progression-free survival in patients with early glottic cancer. Future Oncol 2022; 18:1873-1884. [PMID: 35293227 DOI: 10.2217/fon-2021-1125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2021] [Accepted: 02/02/2022] [Indexed: 11/21/2022] Open
Abstract
Aim: This study aimed to predict progression-free survival (PFS) in patients with early glottic cancer using radiomic features on dual-energy computed tomography iodine maps. Methods: Radiomic features were extracted from arterial and venous phase iodine maps, and radiomic risk scores were determined by univariate Cox proportional hazards regression analysis and least absolute shrinkage and selection operator regression with tenfold cross-validation. The Kaplan-Meier method was used to evaluate the association between radiomic risk scores and PFS. Results: Patients were stratified into low-risk and high-risk groups using radiomics, the PFS corresponding rates with statistical significance between the two groups. The high-risk group showed better survival, benefiting from laryngectomy. Conclusion: Radiomics could provide a promising biomarker for predicting the PFS of early glottic cancer patients.
Collapse
Affiliation(s)
- Wenfei Li
- Department of Radiology, The First Central Clinical School, Tianjin Medical University, No. 24 Fukang Road, Nankai District, Tianjin, 300192, China
- Department of Radiology, The First Hospital of Qinhuangdao, No. 258 Wenhua Road, Haigang District, Qinhuangdao, 066000, China
| | - Huanlei Zhang
- Department of Radiology, The First Central Clinical School, Tianjin Medical University, No. 24 Fukang Road, Nankai District, Tianjin, 300192, China
| | - Lei Ren
- Department of Radiology, The First Central Clinical School, Tianjin Medical University, No. 24 Fukang Road, Nankai District, Tianjin, 300192, China
- Department of Medical Imaging, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, No. 88 Changling Road, Xiqing District, Tianjin, 300381, China
| | - Ying Zou
- Department of Medical Imaging, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, No. 88 Changling Road, Xiqing District, Tianjin, 300381, China
| | - Fengyue Tian
- Department of Radiology, Affiliated Hospital of Nankai University (Tianjin No. 4 Hospital), No. 4 Weishan Road, Hexi District, Tianjin, 300222, China
| | - Xiaodong Ji
- Department of Radiology, Tianjin First Central Hospital, School of Medicine, Nankai University, No. 24 Fukang Road, Nankai District, Tianjin, 300192, China
| | - Qing Li
- Department of Radiology, Tianjin First Central Hospital, School of Medicine, Nankai University, No. 24 Fukang Road, Nankai District, Tianjin, 300192, China
| | - Wei Wang
- Department of Otolaryngology, Tianjin First Central Hospital, School of Medicine, Nankai University, No. 24 Fukang Road, Nankai District, Tianjin, 300192, China
| | - Guolin Ma
- Department of Radiology, China-Japan Friendship Hospital, No. 2 East Yinghua Road, Chaoyang District, Beijing, 100029, China
| | - Shuang Xia
- Department of Radiology, Tianjin First Central Hospital, School of Medicine, Nankai University, No. 24 Fukang Road, Nankai District, Tianjin, 300192, China
| |
Collapse
|
39
|
Hu D, Li S, Zhang H, Wu N, Lu X. Using Natural Language Processing and Machine Learning to Preoperatively Predict Lymph Node Metastasis for Non-Small Cell Lung Cancer With Electronic Medical Records: Development and Validation Study. JMIR Med Inform 2022; 10:e35475. [PMID: 35468085 PMCID: PMC9086872 DOI: 10.2196/35475] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 03/31/2022] [Accepted: 04/11/2022] [Indexed: 11/21/2022] Open
Abstract
Background Lymph node metastasis (LNM) is critical for treatment decision making of patients with resectable non–small cell lung cancer, but it is difficult to precisely diagnose preoperatively. Electronic medical records (EMRs) contain a large volume of valuable information about LNM, but some key information is recorded in free text, which hinders its secondary use. Objective This study aims to develop LNM prediction models based on EMRs using natural language processing (NLP) and machine learning algorithms. Methods We developed a multiturn question answering NLP model to extract features about the primary tumor and lymph nodes from computed tomography (CT) reports. We then combined these features with other structured clinical characteristics to develop LNM prediction models using machine learning algorithms. We conducted extensive experiments to explore the effectiveness of the predictive models and compared them with size criteria based on CT image findings (the maximum short axis diameter of lymph node >10 mm was regarded as a metastatic node) and clinician’s evaluation. Since the NLP model may extract features with mistakes, we also calculated the concordance correlation between the predicted probabilities of models using NLP-extracted features and gold standard features to explore the influence of NLP-driven automatic extraction. Results Experimental results show that the random forest models achieved the best performances with 0.792 area under the receiver operating characteristic curve (AUC) value and 0.456 average precision (AP) value for pN2 LNM prediction and 0.768 AUC value and 0.524 AP value for pN1&N2 LNM prediction. And all machine learning models outperformed the size criteria and clinician’s evaluation. The concordance correlation between the random forest models using NLP-extracted features and gold standard features is 0.950 and improved to 0.984 when the top 5 important NLP-extracted features were replaced with gold standard features. Conclusions The LNM models developed can achieve competitive performance using only limited EMR data such as CT reports and tumor markers in comparison with the clinician’s evaluation. The multiturn question answering NLP model can extract features effectively to support the development of LNM prediction models, which may facilitate the clinical application of predictive models.
Collapse
Affiliation(s)
- Danqing Hu
- College of Biomedical Engineering and Instrumental Science, Zhejiang University, Hangzhou, China
| | - Shaolei Li
- Department of Thoracic Surgery II, Peking University Cancer Hospital and Institute, Beijing, China
| | - Huanyao Zhang
- College of Biomedical Engineering and Instrumental Science, Zhejiang University, Hangzhou, China
| | - Nan Wu
- Department of Thoracic Surgery II, Peking University Cancer Hospital and Institute, Beijing, China
| | - Xudong Lu
- College of Biomedical Engineering and Instrumental Science, Zhejiang University, Hangzhou, China
| |
Collapse
|
40
|
Chang C, Ruan M, Lei B, Yu H, Zhao W, Ge Y, Duan S, Teng W, Wu Q, Qian X, Wang L, Yan H, Liu C, Liu L, Feng J, Xie W. Development of a PET/CT molecular radiomics-clinical model to predict thoracic lymph node metastasis of invasive lung adenocarcinoma ≤ 3 cm in diameter. EJNMMI Res 2022; 12:23. [PMID: 35445899 PMCID: PMC9023644 DOI: 10.1186/s13550-022-00895-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2021] [Accepted: 04/05/2022] [Indexed: 12/25/2022] Open
Abstract
Background To investigate the value of 18F-FDG PET/CT molecular radiomics combined with a clinical model in predicting thoracic lymph node metastasis (LNM) in invasive lung adenocarcinoma (≤ 3 cm). Methods A total of 528 lung adenocarcinoma patients were enrolled in this retrospective study. Five models were developed for the prediction of thoracic LNM, including PET radiomics, CT radiomics, PET/CT radiomics, clinical and integrated PET/CT radiomics-clinical models. Ten PET/CT radiomics features and two clinical characteristics were selected for the construction of the integrated PET/CT radiomics-clinical model. The predictive performance of all models was examined by receiver operating characteristic (ROC) curve analysis, and clinical utility was validated by nomogram analysis and decision curve analysis (DCA). Results According to ROC curve analysis, the integrated PET/CT molecular radiomics-clinical model outperformed the clinical model and the three other radiomics models, and the area under the curve (AUC) values of the integrated model were 0.95 (95% CI: 0.93–0.97) in the training group and 0.94 (95% CI: 0.89–0.97) in the test group. The nomogram analysis and DCA confirmed the clinical application value of this integrated model in predicting thoracic LNM. Conclusions The integrated PET/CT molecular radiomics-clinical model proposed in this study can ensure a higher level of accuracy in predicting the thoracic LNM of clinical invasive lung adenocarcinoma (≤ 3 cm) compared with the radiomics model or clinical model alone. Supplementary Information The online version contains supplementary material available at 10.1186/s13550-022-00895-x.
Collapse
Affiliation(s)
- Cheng Chang
- Department of Nuclear Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University, No. 241 West Huaihai Road, Shanghai, 200030, China.,Clinical and Translational Center in Shanghai Chest Hospital, Shanghai Key Laboratory for Molecular Imaging, Shanghai University of Medicine and Health Sciences, Shanghai, China
| | - Maomei Ruan
- Department of Nuclear Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University, No. 241 West Huaihai Road, Shanghai, 200030, China.,Clinical and Translational Center in Shanghai Chest Hospital, Shanghai Key Laboratory for Molecular Imaging, Shanghai University of Medicine and Health Sciences, Shanghai, China
| | - Bei Lei
- Department of Nuclear Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University, No. 241 West Huaihai Road, Shanghai, 200030, China.,Clinical and Translational Center in Shanghai Chest Hospital, Shanghai Key Laboratory for Molecular Imaging, Shanghai University of Medicine and Health Sciences, Shanghai, China
| | - Hong Yu
- Department of Radiology, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Wenlu Zhao
- Department of Radiology, Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Yaqiong Ge
- GE Healthcare China, Pudong New Town, Shanghai, China
| | - Shaofeng Duan
- GE Healthcare China, Pudong New Town, Shanghai, China
| | - Wenjing Teng
- Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Qianfu Wu
- Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xiaohua Qian
- Institute for Medical Imaging Technology, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Lihua Wang
- Department of Nuclear Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University, No. 241 West Huaihai Road, Shanghai, 200030, China
| | - Hui Yan
- Department of Nuclear Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University, No. 241 West Huaihai Road, Shanghai, 200030, China
| | - Ciyi Liu
- Department of Nuclear Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University, No. 241 West Huaihai Road, Shanghai, 200030, China
| | - Liu Liu
- Department of Nuclear Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University, No. 241 West Huaihai Road, Shanghai, 200030, China.,Clinical and Translational Center in Shanghai Chest Hospital, Shanghai Key Laboratory for Molecular Imaging, Shanghai University of Medicine and Health Sciences, Shanghai, China
| | - Jian Feng
- Department of Thoracic Surgery, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Wenhui Xie
- Department of Nuclear Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University, No. 241 West Huaihai Road, Shanghai, 200030, China. .,Clinical and Translational Center in Shanghai Chest Hospital, Shanghai Key Laboratory for Molecular Imaging, Shanghai University of Medicine and Health Sciences, Shanghai, China.
| |
Collapse
|
41
|
Zan X, Li S, Wei S, Gao L, Zhao L, Yan X, Zhao Y, Shi J, Wang Y, Liu R, Zhang Y, Wan Y, Zhou Y. COL8A1 Promotes NSCLC Progression Through IFIT1/IFIT3-Mediated EGFR Activation. Front Oncol 2022; 12:707525. [PMID: 35280763 PMCID: PMC8907630 DOI: 10.3389/fonc.2022.707525] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Accepted: 01/27/2022] [Indexed: 12/04/2022] Open
Abstract
Activation of EGFR is a major risk factor for non-small cell lung cancer (NSCLC). Understanding the molecular events promoting EGFR activation can help us gain more insights into the progression of NSCLC. In this study, we demonstrate that collagen type VIII alpha 1 chain (COL8A1), an extracellular matrix component, was overexpressed in NSCLC. In NSCLC cells, knockdown of COL8A1 suppressed cell growth, cycle progression, and migration, and induced cell apoptosis. While COL8A1 overexpression promoted cell proliferation and inhibited cell apoptosis. In addition, we found that COL8A1 depletion reduced interferon response signaling and downregulated (IFIT1) and interferon-induced proteins with tetratricopeptide repeats 3 (IFIT3). Moreover, we indicated that COL8A1 could upregulate IFIT1 and IFIT3 mediated EGFR activation in vitro and in vivo. Lastly, there was a positive correlation among COL8A1, IFIT1, and IFIT3 expression, and EGFR activity in patients with NSCLC. Overall, our data demonstrate that COL8A1 contributes to NSCLC proliferation and invasion through EGFR activation, dependent on IFIT1 and IFIT3 expression.
Collapse
Affiliation(s)
- Xiangyi Zan
- Department of Pneumology, The Second Hospital of Lanzhou University, Lanzhou, China.,Department of Gastroenterology, The First Hospital of Lanzhou University, Lanzhou, China.,Key Laboratory for Gastrointestinal Diseases of Gansu Province, Lanzhou University, Lanzhou, China
| | - Shuyan Li
- Department of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, China
| | - Shixiong Wei
- Biological Science, University of California, Davis, Davis, CA, United States
| | - Liping Gao
- Department of Pneumology, The Second Hospital of Lanzhou University, Lanzhou, China
| | - Lanting Zhao
- Department of Pneumology, The Second Hospital of Lanzhou University, Lanzhou, China
| | - Xiaoxia Yan
- Department of Pneumology, The Second Hospital of Lanzhou University, Lanzhou, China
| | - Yan Zhao
- Department of Pneumology, The Second Hospital of Lanzhou University, Lanzhou, China
| | - Junnian Shi
- Department of Pneumology, The Second Hospital of Lanzhou University, Lanzhou, China
| | - Yuping Wang
- Department of Gastroenterology, The First Hospital of Lanzhou University, Lanzhou, China.,Key Laboratory for Gastrointestinal Diseases of Gansu Province, Lanzhou University, Lanzhou, China
| | - Rong Liu
- Department of Gastroenterology, The First Hospital of Lanzhou University, Lanzhou, China.,Key Laboratory for Gastrointestinal Diseases of Gansu Province, Lanzhou University, Lanzhou, China
| | - Yuanyi Zhang
- Clinical Medicine, University of South China, Hengyang, China
| | - Yixin Wan
- Department of Pneumology, The Second Hospital of Lanzhou University, Lanzhou, China
| | - Yongning Zhou
- Department of Gastroenterology, The First Hospital of Lanzhou University, Lanzhou, China.,Key Laboratory for Gastrointestinal Diseases of Gansu Province, Lanzhou University, Lanzhou, China
| |
Collapse
|
42
|
Chen W, Xu M, Sun Y, Ji C, Chen L, Liu S, Zhou K, Zhou Z. Integrative Predictive Models of Computed Tomography Texture Parameters and Hematological Parameters for Lymph Node Metastasis in Lung Adenocarcinomas. J Comput Assist Tomogr 2022; 46:315-324. [PMID: 35297587 PMCID: PMC8929299 DOI: 10.1097/rct.0000000000001264] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Accepted: 09/30/2021] [Indexed: 11/26/2022]
Abstract
OBJECTIVES The aims of the study were to integrate characteristics of computed tomography (CT), texture, and hematological parameters and to establish predictive models for lymph node (LN) metastasis in lung adenocarcinoma. METHODS A total of 207 lung adenocarcinoma cases with confirmed postoperative pathology and preoperative CT scans between February 2017 and April 2019 were included in this retrospective study. All patients were divided into training and 2 validation cohorts chronologically in the ratio of 3:1:1. The χ2 test or Fisher exact test were used for categorical variables. The Shapiro-Wilk test and Mann-Whitney U test were used for continuous variables. Logistic regression and machine learning algorithm models based on CT characteristics, texture, and hematological parameters were used to predict LN metastasis. The performance of the multivariate models was evaluated using a receiver operating characteristic curve; prediction performance was evaluated in the validation cohorts. Decision curve analysis confirmed its clinical utility. RESULTS Logistic regression analysis demonstrated that pleural thickening (P = 0.013), percentile 25th (P = 0.033), entropy gray-level co-occurrence matrix 10 (P = 0.019), red blood cell distribution width (P = 0.012), and lymphocyte-to-monocyte ratio (P = 0.049) were independent risk factors associated with LN metastasis. The area under the curve of the predictive model established using the previously mentioned 5 independent risk factors was 0.929 in the receiver operating characteristic analysis. The highest area under the curve was obtained in the training cohort (0.777 using Naive Bayes algorithm). CONCLUSIONS Integrative predictive models of CT characteristics, texture, and hematological parameters could predict LN metastasis in lung adenocarcinomas. These findings may provide a reference for clinical decision making.
Collapse
Affiliation(s)
- Wenping Chen
- From the Department of Radiology, Nanjing DrumTower Hospital, Clinical College of Nanjing Medical University
| | - Mengying Xu
- Department of Radiology, The Affiliated Hospital of Nanjing University Medical School
| | | | - Changfeng Ji
- From the Department of Radiology, Nanjing DrumTower Hospital, Clinical College of Nanjing Medical University
| | - Ling Chen
- Pathology, Nanjing Drum Tower Hospital, Clinical College of Nanjing Medical University, Nanjing, China
| | - Song Liu
- From the Department of Radiology, Nanjing DrumTower Hospital, Clinical College of Nanjing Medical University
| | - Kefeng Zhou
- From the Department of Radiology, Nanjing DrumTower Hospital, Clinical College of Nanjing Medical University
| | - Zhengyang Zhou
- From the Department of Radiology, Nanjing DrumTower Hospital, Clinical College of Nanjing Medical University
| |
Collapse
|
43
|
Development and Validation of a CT-Based Signature for the Prediction of Distant Metastasis Before Treatment of Non-Small Cell Lung Cancer. Acad Radiol 2022; 29 Suppl 2:S62-S72. [PMID: 33402298 DOI: 10.1016/j.acra.2020.12.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 12/11/2020] [Accepted: 12/11/2020] [Indexed: 01/06/2023]
Abstract
RATIONALE AND OBJECTIVES To develop and validate a radiomics model, a clinical-semantic model and a combined model by using standard methods for the pretreatment prediction of distant metastasis (DM) in patients with non-small-cell lung cancer (NSCLC) and to explore whether the combined model provides added value compared to the individual models. MATERIALS AND METHODS This retrospective study involved 356 patients with NSCLC. According to the image biomarker standardization initiative reference manual, we standardized the image processing and feature extraction using in-house software. Finally, 6692 radiomics features were extracted from each lesion based on contrast-enhanced chest CT images. The least absolute shrinkage selection operator and the recursive feature elimination algorithm were used to select features. The logistic regression classifier was used to build the model. Three models (radiomics model, clinical-semantic model and combined model) were constructed to predict DM in NSCLC. Area under the receiver operating characteristic curves were used to validate the ability of the three models to predict DM. A visual nomogram based on the combined model was developed for DM risk assessment in each patient. RESULTS The receiver operating characteristic curve showed predictive performance for DM of the radiomics model (area under the curve [AUC] values for training and validation were 0.76 [95% CI, 0.704 - 0.820] and 0.76 [95% CI, 0.653 - 0.858], respectively). The combined model had AUCs of 0.78 (95% CI, 0.723 - 0.835) and 0.77 (95% CI, 0.673 - 0.870) in the training and validation cohorts, respectively. Both the radiomics model and combined model performed better than the clinical-semantic model (0.70 [95% CI, 0.634 - 0.760] and 0.67 [95% CI, 0.554 - 0.787] in the training and validation cohorts, respectively). CONCLUSION The radiomics model and combined model may be useful for the prediction of DM in patients with NSCLC.
Collapse
|
44
|
Smyczynska U, Grabia S, Nowicka Z, Papis-Ubych A, Bibik R, Latusek T, Rutkowski T, Fijuth J, Fendler W, Tomasik B. Prediction of Radiation-Induced Hypothyroidism Using Radiomic Data Analysis Does Not Show Superiority over Standard Normal Tissue Complication Models. Cancers (Basel) 2021; 13:cancers13215584. [PMID: 34771747 PMCID: PMC8582656 DOI: 10.3390/cancers13215584] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Revised: 11/02/2021] [Accepted: 11/04/2021] [Indexed: 12/03/2022] Open
Abstract
Simple Summary Radiation-induced hypothyroidism (RIHT) commonly develops in cancer survivors that receive radiation therapy for cancers in the head and neck region. The state-of-art normal tissue complication probability (NTCP) models perform satisfactorily; however, they do not use the whole spectrum of information that can be obtained from imaging techniques. The radiomic approach offers the ability to efficiently mine features, which are imperceptible to the human eye, but may provide crucial data about the patient’s condition. We gathered CT images and clinical data from 98 patients undergoing radiotherapy for head and neck cancers, 27 of whom later developed RIHT. For them, we created machine-learning models to predict RIHT using automatically extracted radiomic features and appropriate clinical and dosimetric parameters. We also validated the well-established external state-of-art NTCP models on our datasets and observed that our radiomic-based models performed very similarly to them. This shows that automated tools may perform as well as the current standard but can be theoretically applied faster and be implemented into existing imaging software used when planning radiotherapy. Abstract State-of-art normal tissue complication probability (NTCP) models do not take into account more complex individual anatomical variations, which can be objectively quantitated and compared in radiomic analysis. The goal of this project was development of radiomic NTCP model for radiation-induced hypothyroidism (RIHT) using imaging biomarkers (radiomics). We gathered CT images and clinical data from 98 patients, who underwent intensity-modulated radiation therapy (IMRT) for head and neck cancers with a planned total dose of 70.0 Gy (33–35 fractions). During the 28-month (median) follow-up 27 patients (28%) developed RIHT. For each patient, we extracted 1316 radiomic features from original and transformed images using manually contoured thyroid masks. Creating models based on clinical, radiomic features or a combination thereof, we considered 3 variants of data preprocessing. Based on their performance metrics (sensitivity, specificity), we picked best models for each variant ((0.8, 0.96), (0.9, 0.93), (0.9, 0.89) variant-wise) and compared them with external NTCP models ((0.82, 0.88), (0.82, 0.88), (0.76, 0.91)). We showed that radiomic-based models did not outperform state-of-art NTCP models (p > 0.05). The potential benefit of radiomic-based approach is that it is dose-independent, and models can be used prior to treatment planning allowing faster selection of susceptible population.
Collapse
Affiliation(s)
- Urszula Smyczynska
- Department of Biostatistics and Translational Medicine, Medical University of Lodz, 92-215 Lodz, Poland; (U.S.); (S.G.); (Z.N.); (B.T.)
| | - Szymon Grabia
- Department of Biostatistics and Translational Medicine, Medical University of Lodz, 92-215 Lodz, Poland; (U.S.); (S.G.); (Z.N.); (B.T.)
| | - Zuzanna Nowicka
- Department of Biostatistics and Translational Medicine, Medical University of Lodz, 92-215 Lodz, Poland; (U.S.); (S.G.); (Z.N.); (B.T.)
| | - Anna Papis-Ubych
- Department of Radiotherapy, N. Copernicus Memorial Regional Specialist Hospital, 93-513 Lodz, Poland; (A.P.-U.); (J.F.)
| | - Robert Bibik
- Department of Radiation Oncology, Oncology Center of Radom, 26-600 Radom, Poland;
| | - Tomasz Latusek
- Radiotherapy Department, Maria Sklodowska-Curie National Research Institute of Oncology (MSCNRIO)—Branch in Gliwice, 44-101 Gliwice, Poland;
| | - Tomasz Rutkowski
- I Radiation and Clinical Oncology Department, Maria Sklodowska-Curie National Research Institute of Oncology (MSCNRIO)—Branch in Gliwice, 44-101 Gliwice, Poland;
| | - Jacek Fijuth
- Department of Radiotherapy, N. Copernicus Memorial Regional Specialist Hospital, 93-513 Lodz, Poland; (A.P.-U.); (J.F.)
- Department of Radiotherapy, Chair of Oncology, Medical University of Lodz, 93-509 Lodz, Poland
| | - Wojciech Fendler
- Department of Biostatistics and Translational Medicine, Medical University of Lodz, 92-215 Lodz, Poland; (U.S.); (S.G.); (Z.N.); (B.T.)
- Department of Radiation Oncology, Dana-Farber Cancer Institute, Boston, MA 02115, USA
- Correspondence:
| | - Bartlomiej Tomasik
- Department of Biostatistics and Translational Medicine, Medical University of Lodz, 92-215 Lodz, Poland; (U.S.); (S.G.); (Z.N.); (B.T.)
- Department of Radiation Oncology, Dana-Farber Cancer Institute, Boston, MA 02115, USA
| |
Collapse
|
45
|
One deep learning local-global model based on CT imaging to differentiate between nodular cryptococcosis and lung cancer which are hard to be diagnosed. Comput Med Imaging Graph 2021; 94:102009. [PMID: 34741847 DOI: 10.1016/j.compmedimag.2021.102009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2021] [Revised: 10/05/2021] [Accepted: 10/14/2021] [Indexed: 12/24/2022]
Abstract
OBJECTIVES We aim to evaluate a deep learning (DL) model and radiomic model for preoperative differentiation of nodular cryptococcosis from solitary lung cancer in patients with malignant features on CT images. MATERIALS AND METHODS We retrospectively recruited 319 patients with solitary pulmonary nodules and suspicious signs of malignancy from three hospitals. All lung nodules were resected, and one by one radiologic-pathologic correlation was performed. A three-dimensional DL model was used for tumor segmentation and extraction of three-dimensional radiomic features. We used the Max-Relevance and Min-Redundancy algorithm and the eXtreme Gradient Boosting algorithm to select the nodular radiomics features. We proposed a DL local-global model, a DL local model and radiomic model to preoperatively differentiate nodular cryptococcosis from solitary lung cancer. The DL local-global model includes information of both nodules and the whole lung, while the DL local model only includes information of solitary lung nodules. Five-fold cross-validation was used to select and validate these models. The prediction performance of the model was evaluated using receiver operating characteristic curve (ROC) and calibration curve. A new loss function was applied in our deep learning framework to optimize the area under the ROC curve (AUC) directly. RESULTS 295 patients were enrolled and they were non-symptomatic, with negative tumor markers and fungus markers in blood tests. These patients have not been diagnosed by the combination of CT imaging, laboratory results and clinical data. The lung volume was slightly larger in patients with lung cancers than that in patients with cryptococcosis (3552.8 ± 1184.6 ml vs 3491.9 ± 1017.8 ml). The DL local-global model achieved the best performance in differentiating between nodular cryptococcosis and lung cancer (area under the curve [AUC] = 0.88), which was higher than that of the DL local model (AUC = 0.84) and radiomic (AUC = 0.79) model. CONCLUSION The DL local-global model is a non-invasive diagnostic tool to differentiate between nodular cryptococcosis and lung cancer nodules which are hard to be diagnosed by the combination of CT imaging, laboratory results and clinical data, and overtreatment may be avoided.
Collapse
|
46
|
Zheng X, Shao J, Zhou L, Wang L, Ge Y, Wang G, Feng F. A Comprehensive Nomogram Combining CT Imaging with Clinical Features for Prediction of Lymph Node Metastasis in Stage I-IIIB Non-small Cell Lung Cancer. Ther Innov Regul Sci 2021; 56:155-167. [PMID: 34699046 DOI: 10.1007/s43441-021-00345-1] [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: 06/03/2021] [Accepted: 10/12/2021] [Indexed: 12/24/2022]
Abstract
OBJECTIVE The status of lymph node metastasis (LNM) is highly correlated with the recurrence and survival outcomes of patients with lung cancer. Thus, a tool that predicts LNM could benefit patient treatment and prognosis. The present study established a new radiomic model by combining computed tomography (CT) radiomic features and clinical parameters to predict the LNM status in patients with non-small cell lung cancer (NSCLC). METHODS Demographic parameters and clinical laboratory values were analyzed in 217 patients with stage I-IIIB NSCLC; 107 of the patients received CT scanning and radiomic characteristics were used for LNM assessment (76 in the training cohort and 31 in the validation cohort). The minimum redundancy maximum relevance (mRMR) and the least absolute shrinkage and selection operator (LASSO) regression model were used to select the most predictive features on the basis of the 76 patients in the training set. The value of the area under the receiver operator characteristic (ROC) curve (AUC) was adopted to determine the correlation between LN status and the radiomics signature in training cohorts and then validated in the 31 patients of validation set. The radiomics nomogram was analyzed using univariate and multivariate logistic regression. Decision curve analysis (DCA) was performed to evaluate the clinical utility of this model. RESULTS This was a retrospective study. Five radiomic characteristics were significantly correlated with LNM in the two cohorts (P < 0.05). The radiomic nomogram that incorporated the above radiomic characteristics, the RDW, and the CT-based LN status had satisfactory discrimination and calibration in the training (AUC, 0.79; 95% CI 0.69-0.89) and validation cohorts (AUC, 0.70; 95% CI 0.50-0.89).The DCA showed that the developed nomogram had promising clinical utility. CONCLUSIONS The developed nomogram, combined with preoperative radiomics evidence, the RDW, and the CT-based LN status, has the potential to preoperatively predict LNM with high accuracy and can facilitate the prediction of LN status for NSCLC patients.
Collapse
Affiliation(s)
- Xingxing Zheng
- Department of Radiology, Affiliated Tumor Hospital of Nantong University, No. 30 Tongyangbei Road, Tongzhou District, Nantong, 226361, China.,Department of Radiology, Baoji Central Hospital, Baoji, 721000, China
| | - Jingjing Shao
- Key Laboratory of Cancer Research Center Nantong, Affiliated Tumor Hospital of Nantong University, Nantong, 226361, China
| | - Linli Zhou
- Department of Radiology, Affiliated Tumor Hospital of Nantong University, No. 30 Tongyangbei Road, Tongzhou District, Nantong, 226361, China
| | - Li Wang
- Department of Radiology, Baoji Central Hospital, Baoji, 721000, China
| | - Yaqiong Ge
- GE Healthcare China, Shanghai, 210000, China
| | - Gaoren Wang
- Department of Radiotherapy, Affiliated Tumor Hospital of Nantong University, Nantong, 226361, China.
| | - Feng Feng
- Department of Radiology, Affiliated Tumor Hospital of Nantong University, No. 30 Tongyangbei Road, Tongzhou District, Nantong, 226361, China.
| |
Collapse
|
47
|
Caruso D, Polici M, Zerunian M, Pucciarelli F, Guido G, Polidori T, Landolfi F, Nicolai M, Lucertini E, Tarallo M, Bracci B, Nacci I, Rucci C, Eid M, Iannicelli E, Laghi A. Radiomics in Oncology, Part 2: Thoracic, Genito-Urinary, Breast, Neurological, Hematologic and Musculoskeletal Applications. Cancers (Basel) 2021; 13:cancers13112681. [PMID: 34072366 PMCID: PMC8197789 DOI: 10.3390/cancers13112681] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 05/26/2021] [Accepted: 05/27/2021] [Indexed: 01/08/2023] Open
Abstract
Simple Summary This Part II is an overview of the main applications of Radiomics in oncologic imaging with a focus on diagnosis, prognosis prediction and assessment of response to therapy in thoracic, genito-urinary, breast, neurologic, hematologic and musculoskeletal oncology. In this part II we describe the radiomic applications, limitations and future perspectives for each pre-eminent tumor. In the future, Radiomics could have a pivotal role in management of cancer patients as an imaging tool to support clinicians in decision making process. However, further investigations need to obtain some stable results and to standardize radiomic analysis (i.e., image acquisitions, segmentation and model building) in clinical routine. Abstract Radiomics has the potential to play a pivotal role in oncological translational imaging, particularly in cancer detection, prognosis prediction and response to therapy evaluation. To date, several studies established Radiomics as a useful tool in oncologic imaging, able to support clinicians in practicing evidence-based medicine, uniquely tailored to each patient and tumor. Mineable data, extracted from medical images could be combined with clinical and survival parameters to develop models useful for the clinicians in cancer patients’ assessment. As such, adding Radiomics to traditional subjective imaging may provide a quantitative and extensive cancer evaluation reflecting histologic architecture. In this Part II, we present an overview of radiomic applications in thoracic, genito-urinary, breast, neurological, hematologic and musculoskeletal oncologic applications.
Collapse
Affiliation(s)
- Damiano Caruso
- Radiology Unit, Department of Medical Surgical Sciences and Translational Medicine, Sapienza University of Rome-Sant’Andrea University Hospital, Via di Grottarossa, 1035-1039, 00189 Rome, Italy; (D.C.); (M.P.); (M.Z.); (F.P.); (G.G.); (T.P.); (F.L.); (M.N.); (E.L.); (B.B.); (I.N.); (C.R.); (E.I.)
| | - Michela Polici
- Radiology Unit, Department of Medical Surgical Sciences and Translational Medicine, Sapienza University of Rome-Sant’Andrea University Hospital, Via di Grottarossa, 1035-1039, 00189 Rome, Italy; (D.C.); (M.P.); (M.Z.); (F.P.); (G.G.); (T.P.); (F.L.); (M.N.); (E.L.); (B.B.); (I.N.); (C.R.); (E.I.)
| | - Marta Zerunian
- Radiology Unit, Department of Medical Surgical Sciences and Translational Medicine, Sapienza University of Rome-Sant’Andrea University Hospital, Via di Grottarossa, 1035-1039, 00189 Rome, Italy; (D.C.); (M.P.); (M.Z.); (F.P.); (G.G.); (T.P.); (F.L.); (M.N.); (E.L.); (B.B.); (I.N.); (C.R.); (E.I.)
| | - Francesco Pucciarelli
- Radiology Unit, Department of Medical Surgical Sciences and Translational Medicine, Sapienza University of Rome-Sant’Andrea University Hospital, Via di Grottarossa, 1035-1039, 00189 Rome, Italy; (D.C.); (M.P.); (M.Z.); (F.P.); (G.G.); (T.P.); (F.L.); (M.N.); (E.L.); (B.B.); (I.N.); (C.R.); (E.I.)
| | - Gisella Guido
- Radiology Unit, Department of Medical Surgical Sciences and Translational Medicine, Sapienza University of Rome-Sant’Andrea University Hospital, Via di Grottarossa, 1035-1039, 00189 Rome, Italy; (D.C.); (M.P.); (M.Z.); (F.P.); (G.G.); (T.P.); (F.L.); (M.N.); (E.L.); (B.B.); (I.N.); (C.R.); (E.I.)
| | - Tiziano Polidori
- Radiology Unit, Department of Medical Surgical Sciences and Translational Medicine, Sapienza University of Rome-Sant’Andrea University Hospital, Via di Grottarossa, 1035-1039, 00189 Rome, Italy; (D.C.); (M.P.); (M.Z.); (F.P.); (G.G.); (T.P.); (F.L.); (M.N.); (E.L.); (B.B.); (I.N.); (C.R.); (E.I.)
| | - Federica Landolfi
- Radiology Unit, Department of Medical Surgical Sciences and Translational Medicine, Sapienza University of Rome-Sant’Andrea University Hospital, Via di Grottarossa, 1035-1039, 00189 Rome, Italy; (D.C.); (M.P.); (M.Z.); (F.P.); (G.G.); (T.P.); (F.L.); (M.N.); (E.L.); (B.B.); (I.N.); (C.R.); (E.I.)
| | - Matteo Nicolai
- Radiology Unit, Department of Medical Surgical Sciences and Translational Medicine, Sapienza University of Rome-Sant’Andrea University Hospital, Via di Grottarossa, 1035-1039, 00189 Rome, Italy; (D.C.); (M.P.); (M.Z.); (F.P.); (G.G.); (T.P.); (F.L.); (M.N.); (E.L.); (B.B.); (I.N.); (C.R.); (E.I.)
| | - Elena Lucertini
- Radiology Unit, Department of Medical Surgical Sciences and Translational Medicine, Sapienza University of Rome-Sant’Andrea University Hospital, Via di Grottarossa, 1035-1039, 00189 Rome, Italy; (D.C.); (M.P.); (M.Z.); (F.P.); (G.G.); (T.P.); (F.L.); (M.N.); (E.L.); (B.B.); (I.N.); (C.R.); (E.I.)
| | - Mariarita Tarallo
- Department of Surgery “Pietro Valdoni”, Sapienza University of Rome, 00161 Rome, Italy;
| | - Benedetta Bracci
- Radiology Unit, Department of Medical Surgical Sciences and Translational Medicine, Sapienza University of Rome-Sant’Andrea University Hospital, Via di Grottarossa, 1035-1039, 00189 Rome, Italy; (D.C.); (M.P.); (M.Z.); (F.P.); (G.G.); (T.P.); (F.L.); (M.N.); (E.L.); (B.B.); (I.N.); (C.R.); (E.I.)
| | - Ilaria Nacci
- Radiology Unit, Department of Medical Surgical Sciences and Translational Medicine, Sapienza University of Rome-Sant’Andrea University Hospital, Via di Grottarossa, 1035-1039, 00189 Rome, Italy; (D.C.); (M.P.); (M.Z.); (F.P.); (G.G.); (T.P.); (F.L.); (M.N.); (E.L.); (B.B.); (I.N.); (C.R.); (E.I.)
| | - Carlotta Rucci
- Radiology Unit, Department of Medical Surgical Sciences and Translational Medicine, Sapienza University of Rome-Sant’Andrea University Hospital, Via di Grottarossa, 1035-1039, 00189 Rome, Italy; (D.C.); (M.P.); (M.Z.); (F.P.); (G.G.); (T.P.); (F.L.); (M.N.); (E.L.); (B.B.); (I.N.); (C.R.); (E.I.)
| | - Marwen Eid
- Internal Medicine, Northwell Health Staten Island University Hospital, Staten Island, New York, NY 10305, USA;
| | - Elsa Iannicelli
- Radiology Unit, Department of Medical Surgical Sciences and Translational Medicine, Sapienza University of Rome-Sant’Andrea University Hospital, Via di Grottarossa, 1035-1039, 00189 Rome, Italy; (D.C.); (M.P.); (M.Z.); (F.P.); (G.G.); (T.P.); (F.L.); (M.N.); (E.L.); (B.B.); (I.N.); (C.R.); (E.I.)
| | - Andrea Laghi
- Radiology Unit, Department of Medical Surgical Sciences and Translational Medicine, Sapienza University of Rome-Sant’Andrea University Hospital, Via di Grottarossa, 1035-1039, 00189 Rome, Italy; (D.C.); (M.P.); (M.Z.); (F.P.); (G.G.); (T.P.); (F.L.); (M.N.); (E.L.); (B.B.); (I.N.); (C.R.); (E.I.)
- Correspondence: ; Tel.: +39-0633775285
| |
Collapse
|
48
|
Li Q, He XQ, Fan X, Zhu CN, Lv JW, Luo TY. Development and Validation of a Combined Model for Preoperative Prediction of Lymph Node Metastasis in Peripheral Lung Adenocarcinoma. Front Oncol 2021; 11:675877. [PMID: 34109124 PMCID: PMC8180898 DOI: 10.3389/fonc.2021.675877] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Accepted: 04/23/2021] [Indexed: 12/25/2022] Open
Abstract
Background Based on the “seed and soil” theory proposed by previous studies, we aimed to develop and validate a combined model of machine learning for predicting lymph node metastasis (LNM) in patients with peripheral lung adenocarcinoma (PLADC). Methods Radiomics models were developed in a primary cohort of 390 patients (training cohort) with pathologically confirmed PLADC from January 2016 to August 2018. The patients were divided into the LNM (−) and LNM (+) groups. Thereafter, the patients were subdivided according to TNM stages N0, N1, N2, and N3. Radiomic features from unenhanced computed tomography (CT) were extracted. Radiomic signatures of the primary tumor (R1) and adjacent pleura (R2) were built as predictors of LNM. CT morphological features and clinical characteristics were compared between both groups. A combined model incorporating R1, R2, and CT morphological features, and clinical risk factors was developed by multivariate analysis. The combined model’s performance was assessed by receiver operating characteristic (ROC) curve. An internal validation cohort containing 166 consecutive patients from September 2018 to November 2019 was also assessed. Results Thirty-one radiomic features of R1 and R2 were significant predictors of LNM (all P < 0.05). Sex, smoking history, tumor size, density, air bronchogram, spiculation, lobulation, necrosis, pleural effusion, and pleural involvement also differed significantly between the groups (all P < 0.05). R1, R2, tumor size, and spiculation in the combined model were independent risk factors for predicting LNM in patients with PLADC, with area under the ROC curves (AUCs) of 0.897 and 0.883 in the training and validation cohorts, respectively. The combined model identified N0, N1, N2, and N3, with AUCs ranging from 0.691–0.927 in the training cohort and 0.700–0.951 in the validation cohort, respectively, thereby indicating good performance. Conclusion CT phenotypes of the primary tumor and adjacent pleura were significantly associated with LNM. A combined model incorporating radiomic signatures, CT morphological features, and clinical risk factors can assess LNM of patients with PLADC accurately and non-invasively.
Collapse
Affiliation(s)
- Qi Li
- Department of Radiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xiao-Qun He
- Department of Radiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xiao Fan
- Department of Radiology, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China
| | - Chao-Nan Zhu
- Hangzhou YITU Healthcare Technology, Hangzhou, China
| | - Jun-Wei Lv
- Hangzhou YITU Healthcare Technology, Hangzhou, China
| | - Tian-You Luo
- Department of Radiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
49
|
Artificial Intelligence and the Medical Physicist: Welcome to the Machine. APPLIED SCIENCES-BASEL 2021. [DOI: 10.3390/app11041691] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Artificial intelligence (AI) is a branch of computer science dedicated to giving machines or computers the ability to perform human-like cognitive functions, such as learning, problem-solving, and decision making. Since it is showing superior performance than well-trained human beings in many areas, such as image classification, object detection, speech recognition, and decision-making, AI is expected to change profoundly every area of science, including healthcare and the clinical application of physics to healthcare, referred to as medical physics. As a result, the Italian Association of Medical Physics (AIFM) has created the “AI for Medical Physics” (AI4MP) group with the aims of coordinating the efforts, facilitating the communication, and sharing of the knowledge on AI of the medical physicists (MPs) in Italy. The purpose of this review is to summarize the main applications of AI in medical physics, describe the skills of the MPs in research and clinical applications of AI, and define the major challenges of AI in healthcare.
Collapse
|
50
|
Dong M, Hou G, Li S, Li N, Zhang L, Xu K. Preoperatively Estimating the Malignant Potential of Mediastinal Lymph Nodes: A Pilot Study Toward Establishing a Robust Radiomics Model Based on Contrast-Enhanced CT Imaging. Front Oncol 2021; 10:558428. [PMID: 33489871 PMCID: PMC7821835 DOI: 10.3389/fonc.2020.558428] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2020] [Accepted: 11/18/2020] [Indexed: 11/23/2022] Open
Abstract
PURPOSE To establish and validate a radiomics model to estimate the malignancy of mediastinal lymph nodes (LNs) based on contrast-enhanced CT imaging. METHOD In total, 201 pathologically confirmed mediastinal LNs from 129 patients were enrolled and assigned to training and test sets. Radiomics features were extracted from the region of interest (ROI) delineated on venous-phase CT imaging of LN. Feature selection was performed with least absolute shrinkage and selection operator (LASSO) binary logistic regression. Multivariate logistic regression was performed with the backward stepwise elimination. A model was fitted to associate mediastinal LN malignancy with selected features. The performance of the model was assessed and compared to that of five other machine learning algorithms (support vector machine, naive Bayes, random forest, decision tree, K-nearest neighbor) using receiver operating characteristic (ROC) curves. Calibration curves and Hosmer-Lemeshow tests were used to assess the calibration degree. Decision curve analysis (DCA) was used to assess the clinical usefulness of the logistic regression model in both the training and test sets. Stratified analysis was performed for different scanners and slice thicknesses. RESULT Among the six machine learning methods, the logistic regression model with the eight strongest features showed a significant association with mediastinal LN status and the satisfactory diagnostic performance for distinguishing malignant LNs from benign LNs. The accuracy, sensitivity, specificity and area under the ROC curve (AUC) were 0.850/0.803, 0.821/0.806, 0.893/0.800, and 0.922/0.850 in the training/test sets, respectively. The Hosmer-Lemeshow test showed that the P value was > 0.05, indicating good calibration, and the calibration curves showed good agreement between the classifications and actual observations. DCA showed that the model would obtain more benefit when the threshold probability was between 30% and 90% in the test set. Stratified analysis showed that the performance was not affected by different scanners or slice thicknesses. There was no significant difference (DeLong test, P > 0.05) between any two subgroups, which showed the generalization of the radiomics score across different factors. CONCLUSION The model we built could help assist the preoperative estimation of mediastinal LN malignancy based on contrast-enhanced CT imaging, with stability for different scanners and slice thicknesses.
Collapse
Affiliation(s)
- Mengshi Dong
- Department of Radiology, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Gang Hou
- Institute of Respiratory Disease, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Shu Li
- Department of Radiology, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Nan Li
- Department of Radiology, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Lina Zhang
- Department of Radiology, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Ke Xu
- Department of Radiology, The First Affiliated Hospital of China Medical University, Shenyang, China
| |
Collapse
|