1
|
Ferro A, Bottosso M, Dieci MV, Scagliori E, Miglietta F, Aldegheri V, Bonanno L, Caumo F, Guarneri V, Griguolo G, Pasello G. Clinical applications of radiomics and deep learning in breast and lung cancer: A narrative literature review on current evidence and future perspectives. Crit Rev Oncol Hematol 2024; 203:104479. [PMID: 39151838 DOI: 10.1016/j.critrevonc.2024.104479] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Revised: 07/22/2024] [Accepted: 08/10/2024] [Indexed: 08/19/2024] Open
Abstract
Radiomics, analysing quantitative features from medical imaging, has rapidly become an emerging field in translational oncology. Radiomics has been investigated in several neoplastic malignancies as it might allow for a non-invasive tumour characterization and for the identification of predictive and prognostic biomarkers. Over the last few years, evidence has been accumulating regarding potential clinical applications of machine learning in many crucial moments of cancer patients' history. However, the incorporation of radiomics in clinical decision-making process is still limited by low data reproducibility and study variability. Moreover, the need for prospective validations and standardizations is emerging. In this narrative review, we summarize current evidence regarding radiomic applications in high-incidence cancers (breast and lung) for screening, diagnosis, staging, treatment choice, response, and clinical outcome evaluation. We also discuss pro and cons of the radiomic approach, suggesting possible solutions to critical issues which might invalidate radiomics studies and propose future perspectives.
Collapse
Affiliation(s)
- Alessandra Ferro
- Division of Medical Oncology 2, Veneto Institute of Oncology IOV - IRCCS, via Gattamelata 64, Padua 35128, Italy
| | - Michele Bottosso
- Division of Medical Oncology 2, Veneto Institute of Oncology IOV - IRCCS, via Gattamelata 64, Padua 35128, Italy; Department of Surgery, Oncology and Gastroenterology, University of Padova, via Giustiniani 2, Padova 35128, Italy
| | - Maria Vittoria Dieci
- Division of Medical Oncology 2, Veneto Institute of Oncology IOV - IRCCS, via Gattamelata 64, Padua 35128, Italy; Department of Surgery, Oncology and Gastroenterology, University of Padova, via Giustiniani 2, Padova 35128, Italy.
| | - Elena Scagliori
- Radiology Unit, Veneto Institute of Oncology IOV - IRCCS, via Gattamelata 64, Padua 35128, Italy
| | - Federica Miglietta
- Division of Medical Oncology 2, Veneto Institute of Oncology IOV - IRCCS, via Gattamelata 64, Padua 35128, Italy; Department of Surgery, Oncology and Gastroenterology, University of Padova, via Giustiniani 2, Padova 35128, Italy
| | - Vittoria Aldegheri
- Radiology Unit, Veneto Institute of Oncology IOV - IRCCS, via Gattamelata 64, Padua 35128, Italy
| | - Laura Bonanno
- Division of Medical Oncology 2, Veneto Institute of Oncology IOV - IRCCS, via Gattamelata 64, Padua 35128, Italy
| | - Francesca Caumo
- Unit of Breast Radiology, Veneto Institute of Oncology IOV - IRCCS, via Gattamelata 64, Padua 35128, Italy
| | - Valentina Guarneri
- Division of Medical Oncology 2, Veneto Institute of Oncology IOV - IRCCS, via Gattamelata 64, Padua 35128, Italy; Department of Surgery, Oncology and Gastroenterology, University of Padova, via Giustiniani 2, Padova 35128, Italy
| | - Gaia Griguolo
- Division of Medical Oncology 2, Veneto Institute of Oncology IOV - IRCCS, via Gattamelata 64, Padua 35128, Italy; Department of Surgery, Oncology and Gastroenterology, University of Padova, via Giustiniani 2, Padova 35128, Italy
| | - Giulia Pasello
- Division of Medical Oncology 2, Veneto Institute of Oncology IOV - IRCCS, via Gattamelata 64, Padua 35128, Italy; Department of Surgery, Oncology and Gastroenterology, University of Padova, via Giustiniani 2, Padova 35128, Italy
| |
Collapse
|
2
|
Hu Y, Geng Y, Wang H, Chen H, Wang Z, Fu L, Huang B, Jiang W. Improved Prediction of Epidermal Growth Factor Receptor Status by Combined Radiomics of Primary Nonsmall-Cell Lung Cancer and Distant Metastasis. J Comput Assist Tomogr 2024; 48:780-788. [PMID: 38498926 DOI: 10.1097/rct.0000000000001591] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/20/2024]
Abstract
OBJECTIVES This study aimed to investigate radiomics based on primary nonsmall-cell lung cancer (NSCLC) and distant metastases to predict epidermal growth factor receptor (EGFR) mutation status. METHODS A total of 290 patients (mean age, 58.21 ± 9.28) diagnosed with brain (BM, n = 150) or spinal bone metastasis (SM, n = 140) from primary NSCLC were enrolled as a primary cohort. An external validation cohort, consisting of 69 patients (mean age, 59.87 ± 7.23; BM, n = 36; SM, n = 33), was enrolled from another center. Thoracic computed tomography-based features were extracted from the primary tumor and peritumoral area and selected using the least absolute shrinkage and selection operator regression to build a radiomic signature (RS-primary). Contrast-enhanced magnetic resonance imaging-based features were calculated and selected from the BM and SM to build RS-BM and RS-SM, respectively. The RS-BM-Com and RS-SM-Com were developed by integrating the most important features from the primary tumor, BM, and SM. RESULTS Six computed tomography-based features showed high association with EGFR mutation status: 3 from intratumoral and 3 from peritumoral areas. By combination of features from primary tumor and metastases, the developed RS-BM-Com and RS-SM-Com performed well with areas under curve in the training (RS-BM-Com vs RS-BM, 0.936 vs 0.885, P = 0.177; RS-SM-Com vs RS-SM, 0.929 vs 0.843, P = 0.003), internal validation (RS-BM-Com vs RS-BM, 0.920 vs 0.858, P = 0.492; RS-SM-Com vs RS-SM, 0.896 vs 0.859, P = 0.379), and external validation (RS-BM-Com vs RS-BM, 0.882 vs 0.805, P = 0.263; RS-SM-Com vs RS-SM, 0.865 vs 0.816, P = 0.312) cohorts. CONCLUSIONS This study indicates that the accuracy of detecting EGFR mutations significantly enhanced in the presence of metastases in primary NSCLC. The established radiomic signatures from this approach may be useful as new predictors for patients with distant metastases.
Collapse
Affiliation(s)
- Yue Hu
- From the Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing
| | - Yikang Geng
- School of Intelligent Medicine, China Medical University, Liaoning
| | - Huan Wang
- Radiation Oncology Department of Thoracic Cancer, Cancer Hospital of Dalian University of Technology (Liaoning Cancer Hospital & Institute), Liaoning
| | - Huanhuan Chen
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang
| | - Zekun Wang
- Department of Medical Iconography, Liaoning Cancer Hospital & Institute, Liaoning
| | - Langyuan Fu
- School of Intelligent Medicine, China Medical University, Liaoning
| | - Bo Huang
- Department of Pathology, Liaoning Cancer Hospital and Institute, Liaoning
| | - Wenyan Jiang
- Department of Scientific Research and Academic, Liaoning Cancer Hospital and Institute, Liaoning, People's Republic of China
| |
Collapse
|
3
|
Lan H, Wei C, Xu F, Yang E, Lu D, Feng Q, Li T. 2.5D peritumoural radiomics predicts postoperative recurrence in stage I lung adenocarcinoma. Front Oncol 2024; 14:1382815. [PMID: 39267836 PMCID: PMC11390697 DOI: 10.3389/fonc.2024.1382815] [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: 02/06/2024] [Accepted: 08/06/2024] [Indexed: 09/15/2024] Open
Abstract
Objective Radiomics can non-invasively predict the prognosis of a tumour by applying advanced imaging feature algorithms.The aim of this study was to predict the chance of postoperative recurrence by modelling tumour radiomics and peritumour radiomics and clinical features in patients with stage I lung adenocarcinoma (LUAD). Materials and methods Retrospective analysis of 190 patients with postoperative pathologically confirmed stage I LUAD from centre 1, who were divided into training cohort and internal validation cohort, with centre 2 added as external validation cohort. To develop a combined radiation-clinical omics model nomogram incorporating clinical features based on images from low-dose lung cancer screening CT plain for predicting postoperative recurrence and to evaluate the performance of the nomogram in the training cohort, internal validation cohort and external validation cohort. Results A total of 190 patients were included in the model in centre 1 and randomised into a training cohort of 133 and an internal validation cohort of 57 in a ratio of 7:3, and 39 were included in centre 2 as an external validation cohort. In the training cohort (AUC=0.865, 95% CI 0.824-0.906), internal validation cohort (AUC=0.902, 95% CI 0.851-0.953) and external validation cohort (AUC=0.830,95% CI 0.751-0.908), the combined radiation-clinical omics model had a good predictive ability. The combined model performed significantly better than the conventional single-modality models (clinical model, radiomic model), and the calibration curve and decision curve analysis (DCA) showed high accuracy and clinical utility of the nomogram. Conclusion The combined preoperative radiation-clinical omics model provides good predictive value for postoperative recurrence in stage ILUAD and combines the model's superiority in both internal and external validation cohorts, demonstrating its potential to aid in postoperative treatment strategies.
Collapse
Affiliation(s)
- Haimei Lan
- Department of Radiology, Liuzhou Workers Hospital, Liuzhou, Guangxi, China
| | - Chaosheng Wei
- Department of Radiology, Liuzhou Workers Hospital, Liuzhou, Guangxi, China
| | - Fengming Xu
- Department of Radiology, Liuzhou Workers Hospital, Liuzhou, Guangxi, China
| | - Eqing Yang
- Department of Radiology, Liuzhou Workers Hospital, Liuzhou, Guangxi, China
| | - Dayu Lu
- Department of Radiology, Longtan Hospital, Liuzhou, Guangxi, China
| | - Qing Feng
- Department of Radiology, Liuzhou Workers Hospital, Liuzhou, Guangxi, China
| | - Tao Li
- Department of Radiology, Liuzhou Workers Hospital, Liuzhou, Guangxi, China
| |
Collapse
|
4
|
Zhang R, Zhu H, Chen M, Sang W, Lu K, Li Z, Wang C, Zhang L, Yin FF, Yang Z. A dual-radiomics model for overall survival prediction in early-stage NSCLC patient using pre-treatment CT images. Front Oncol 2024; 14:1419621. [PMID: 39206157 PMCID: PMC11349529 DOI: 10.3389/fonc.2024.1419621] [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: 04/18/2024] [Accepted: 07/26/2024] [Indexed: 09/04/2024] Open
Abstract
Introduction Radiation therapy (RT) is one of the primary treatment options for early-stage non-small cell lung cancer (ES-NSCLC). Therefore, accurately predicting the overall survival (OS) rate following radiotherapy is crucial for implementing personalized treatment strategies. This work aims to develop a dual-radiomics (DR) model to (1) predict 3-year OS in ES-NSCLC patients receiving RT using pre-treatment CT images, and (2) provide explanations between feature importanceand model prediction performance. Methods The publicly available TCIA Lung1 dataset with 132 ES-NSCLC patients received RT were studied: 89/43 patients in the under/over 3-year OS group. For each patient, two types of radiomic features were examined: 56 handcrafted radiomic features (HRFs) extracted within gross tumor volume, and 512 image deep features (IDFs) extracted using a pre-trained U-Net encoder. They were combined as inputs to an explainable boosting machine (EBM) model for OS prediction. The EBM's mean absolute scores for HRFs and IDFs were used as feature importance explanations. To evaluate identified feature importance, the DR model was compared with EBM using either (1) key or (2) non-key feature type only. Comparison studies with other models, including supporting vector machine (SVM) and random forest (RF), were also included. The performance was evaluated by the area under the receiver operating characteristic curve (AUCROC), accuracy, sensitivity, and specificity with a 100-fold Monte Carlo cross-validation. Results The DR model showed highestperformance in predicting 3-year OS (AUCROC=0.81 ± 0.04), and EBM scores suggested that IDFs showed significantly greater importance (normalized mean score=0.0019) than HRFs (score=0.0008). The comparison studies showed that EBM with key feature type (IDFs-only demonstrated comparable AUCROC results (0.81 ± 0.04), while EBM with non-key feature type (HRFs-only) showed limited AUCROC (0.64 ± 0.10). The results suggested that feature importance score identified by EBM is highly correlated with OS prediction performance. Both SVM and RF models were unable to explain key feature type while showing limited overall AUCROC=0.66 ± 0.07 and 0.77 ± 0.06, respectively. Accuracy, sensitivity, and specificity showed a similar trend. Discussion In conclusion, a DR model was successfully developed to predict ES-NSCLC OS based on pre-treatment CT images. The results suggested that the feature importance from DR model is highly correlated to the model prediction power.
Collapse
Affiliation(s)
- Rihui Zhang
- Medical Physics Graduate Program, Duke Kunshan University, Kunshan, Jiangsu, China
| | - Haiming Zhu
- Medical Physics Graduate Program, Duke Kunshan University, Kunshan, Jiangsu, China
| | - Minbin Chen
- Department of Radiotherapy & Oncology, The First People’s Hospital of Kunshan, Kunshan, Jiangsu, China
| | - Weiwei Sang
- Medical Physics Graduate Program, Duke Kunshan University, Kunshan, Jiangsu, China
| | - Ke Lu
- Deparment of Radiation Oncology, Duke University, Durham, NC, United States
| | - Zhen Li
- Radiation Oncology Department, Shanghai Sixth People’s Hospital, Shanghai, China
| | - Chunhao Wang
- Deparment of Radiation Oncology, Duke University, Durham, NC, United States
| | - Lei Zhang
- Medical Physics Graduate Program, Duke Kunshan University, Kunshan, Jiangsu, China
| | - Fang-Fang Yin
- Medical Physics Graduate Program, Duke Kunshan University, Kunshan, Jiangsu, China
| | - Zhenyu Yang
- Medical Physics Graduate Program, Duke Kunshan University, Kunshan, Jiangsu, China
| |
Collapse
|
5
|
Duan C, Liu Q, Wang J, Tong Q, Bai F, Han J, Wang S, Hippe DS, Zeng J, Bowen SR. GWO+RuleFit: rule-based explainable machine-learning combined with heuristics to predict mid-treatment FDG PET response to chemoradiation for locally advanced non-small cell lung cancer. Phys Med Biol 2024; 69:10.1088/1361-6560/ad6118. [PMID: 38981590 PMCID: PMC11338282 DOI: 10.1088/1361-6560/ad6118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Accepted: 07/09/2024] [Indexed: 07/11/2024]
Abstract
Objective.Vital rules learned from fluorodeoxyglucose positron emission tomography (FDG-PET) radiomics of tumor subregional response can provide clinical decision support for precise treatment adaptation. We combined a rule-based machine learning (ML) model (RuleFit) with a heuristic algorithm (gray wolf optimizer, GWO) for mid-chemoradiation FDG-PET response prediction in patients with locally advanced non-small cell lung cancer.Approach.Tumors subregions were identified using K-means clustering. GWO+RuleFit consists of three main parts: (i) a random forest is constructed based on conventional features or radiomic features extracted from tumor regions or subregions in FDG-PET images, from which the initial rules are generated; (ii) GWO is used for iterative rule selection; (iii) the selected rules are fit to a linear model to make predictions about the target variable. Two target variables were considered: a binary response measure (ΔSUVmean ⩾ 20% decline) for classification and a continuous response measure (ΔSUVmean) for regression. GWO+RuleFit was benchmarked against common ML algorithms and RuleFit, with leave-one-out cross-validated performance evaluated by the area under the receiver operating characteristic curve (AUC) in classification and root-mean-square error (RMSE) in regression.Main results.GWO+RuleFit selected 15 rules from the radiomic feature dataset of 23 patients. For treatment response classification, GWO+RuleFit attained numerically better cross-validated performance than RuleFit across tumor regions and sets of features (AUC: 0.58-0.86 vs. 0.52-0.78,p= 0.170-0.925). GWO+Rulefit also had the best or second-best performance numerically compared to all other algorithms for all conditions. For treatment response regression prediction, GWO+RuleFit (RMSE: 0.162-0.192) performed better numerically for low-dimensional models (p= 0.097-0.614) and significantly better for high-dimensional models across all tumor regions except one (RMSE: 0.189-0.219,p< 0.004).Significance. The GWO+RuleFit selected rules were interpretable, highlighting distinct radiomic phenotypes that modulated treatment response. GWO+Rulefit achieved parsimonious models while maintaining utility for treatment response prediction, which can aid clinical decisions for patient risk stratification, treatment selection, and biologically driven adaptation. Clinical trial: NCT02773238.
Collapse
Affiliation(s)
- Chunyan Duan
- Department of Mechanical Engineering, School of Mechanical Engineering, Tongji University, 4800 Cao’an Highway, Shanghai 201804, P. R. China
| | - Qiantuo Liu
- Department of Mechanical Engineering, School of Mechanical Engineering, Tongji University, 4800 Cao’an Highway, Shanghai 201804, P. R. China
| | - Jiajie Wang
- Department of Mechanical Engineering, School of Mechanical Engineering, Tongji University, 4800 Cao’an Highway, Shanghai 201804, P. R. China
| | - Qianqian Tong
- Maseeh Department of Civil, Architectural and Environmental Engineering, Cockrell School of Engineering, The University of Texas at Austin, 301 East Dean Keeton Street, Austin, TX 78712, USA
| | - Fangyun Bai
- Shanghai Yangzhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center), Tongji University, 2209 Guangxing Road, Shanghai 201613, P. R. China
| | - Jie Han
- Department of Industrial, Manufacturing, and Systems Engineering, College of Engineering, The University of Texas at Arlington, 500 West First Street, Arlington, TX 76019, USA
| | - Shouyi Wang
- Department of Industrial, Manufacturing, and Systems Engineering, College of Engineering, The University of Texas at Arlington, 500 West First Street, Arlington, TX 76019, USA
| | - Daniel S. Hippe
- Clinical Research Division, Fred Hutchinson Cancer Center, 1100 Fairview Avenue North, Seattle, WA 98109, USA
| | - Jing Zeng
- Department of Radiation Oncology, School of Medicine, University of Washington, 1959 North East Pacific Street, Seattle, WA 98195, USA
| | - Stephen R. Bowen
- Clinical Research Division, Fred Hutchinson Cancer Center, 1100 Fairview Avenue North, Seattle, WA 98109, USA
- Department of Radiation Oncology, School of Medicine, University of Washington, 1959 North East Pacific Street, Seattle, WA 98195, USA
- Department of Radiology, School of Medicine, University of Washington, 1959 North East Pacific Street, Seattle, WA 98195, USA
| |
Collapse
|
6
|
Xu N, Wang J, Dai G, Lu T, Li S, Deng K, Song J. EfficientNet-Based System for Detecting EGFR-Mutant Status and Predicting Prognosis of Tyrosine Kinase Inhibitors in Patients with NSCLC. JOURNAL OF IMAGING INFORMATICS IN MEDICINE 2024; 37:1086-1099. [PMID: 38361006 PMCID: PMC11169294 DOI: 10.1007/s10278-024-01022-z] [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: 09/29/2023] [Revised: 12/29/2023] [Accepted: 01/09/2024] [Indexed: 02/17/2024]
Abstract
We aimed to develop and validate a deep learning-based system using pre-therapy computed tomography (CT) images to detect epidermal growth factor receptor (EGFR)-mutant status in patients with non-small cell lung cancer (NSCLC) and predict the prognosis of advanced-stage patients with EGFR mutations treated with EGFR tyrosine kinase inhibitors (TKI). This retrospective, multicenter study included 485 patients with NSCLC from four hospitals. Of them, 339 patients from three centers were included in the training dataset to develop an EfficientNetV2-L-based model (EME) for predicting EGFR-mutant status, and the remaining patients were assigned to an independent test dataset. EME semantic features were extracted to construct an EME-prognostic model to stratify the prognosis of EGFR-mutant NSCLC patients receiving EGFR-TKI. A comparison of EME and radiomics was conducted. Additionally, we included patients from The Cancer Genome Atlas lung adenocarcinoma dataset with both CT images and RNA sequencing data to explore the biological associations between EME score and EGFR-related biological processes. EME obtained an area under the curve (AUC) of 0.907 (95% CI 0.840-0.926) on the test dataset, superior to the radiomics model (P = 0.007). The EME and radiomics fusion model showed better (AUC, 0.941) but not significantly increased performance (P = 0.895) compared with EME. In prognostic stratification, the EME-prognostic model achieved the best performance (C-index, 0.711). Moreover, the EME-prognostic score showed strong associations with biological pathways related to EGFR expression and EGFR-TKI efficacy. EME demonstrated a non-invasive and biologically interpretable approach to predict EGFR status, stratify survival prognosis, and correlate biological pathways in patients with NSCLC.
Collapse
Affiliation(s)
- Nan Xu
- School of Health Management, China Medical University, Shenyang, Liaoning, 110122, China
| | - Jiajun Wang
- Department of Thoracic Surgery, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning, 110001, China
| | - Gang Dai
- Department of Radiology, The First Affiliated Hospital of University of Science and Technology of China (USTC), Division of Life Sciences and Medicine, USTC, Hefei, Anhui, 230036, China
| | - Tao Lu
- Department of Radiology, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning, 110001, China
| | - Shu Li
- School of Health Management, China Medical University, Shenyang, Liaoning, 110122, China
| | - Kexue Deng
- Department of Radiology, The First Affiliated Hospital of University of Science and Technology of China (USTC), Division of Life Sciences and Medicine, USTC, Hefei, Anhui, 230036, China
| | - Jiangdian Song
- School of Health Management, China Medical University, Shenyang, Liaoning, 110122, China.
| |
Collapse
|
7
|
Yu Y, Guo H, Zhang M, Hou F, Yang S, Huang C, Duan L, Wang H. Multi-institutional validation of a radiomics signature for identification of postoperative progression of soft tissue sarcoma. Cancer Imaging 2024; 24:59. [PMID: 38720384 PMCID: PMC11077743 DOI: 10.1186/s40644-024-00705-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2023] [Accepted: 04/27/2024] [Indexed: 05/12/2024] Open
Abstract
BACKGROUND To develop a magnetic resonance imaging (MRI)-based radiomics signature for evaluating the risk of soft tissue sarcoma (STS) disease progression. METHODS We retrospectively enrolled 335 patients with STS (training, validation, and The Cancer Imaging Archive sets, n = 168, n = 123, and n = 44, respectively) who underwent surgical resection. Regions of interest were manually delineated using two MRI sequences. Among 12 machine learning-predicted signatures, the best signature was selected, and its prediction score was inputted into Cox regression analysis to build the radiomics signature. A nomogram was created by combining the radiomics signature with a clinical model constructed using MRI and clinical features. Progression-free survival was analyzed in all patients. We assessed performance and clinical utility of the models with reference to the time-dependent receiver operating characteristic curve, area under the curve, concordance index, integrated Brier score, decision curve analysis. RESULTS For the combined features subset, the minimum redundancy maximum relevance-least absolute shrinkage and selection operator regression algorithm + decision tree classifier had the best prediction performance. The radiomics signature based on the optimal machine learning-predicted signature, and built using Cox regression analysis, had greater prognostic capability and lower error than the nomogram and clinical model (concordance index, 0.758 and 0.812; area under the curve, 0.724 and 0.757; integrated Brier score, 0.080 and 0.143, in the validation and The Cancer Imaging Archive sets, respectively). The optimal cutoff was - 0.03 and cumulative risk rates were calculated. DATA CONCLUSION To assess the risk of STS progression, the radiomics signature may have better prognostic power than a nomogram/clinical model.
Collapse
Affiliation(s)
- Yuan Yu
- Department of Radiology, The Affiliated Hospital of Qingdao University, 16 Jiangsu Road, Qingdao, Shandong, China
| | - Hongwei Guo
- Department of Operation Center, Women and Children's Hospital, Qingdao University, Shandong, China
| | - Meng Zhang
- Department of Radiology, The Affiliated Hospital of Qingdao University, 16 Jiangsu Road, Qingdao, Shandong, China
| | - Feng Hou
- Department of Pathology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Shifeng Yang
- Department of Radiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Chencui Huang
- Department of Research Collaboration, Research and Development (R&D) center, Beijing Deepwise & League of Philosophy Doctor (PHD) Technology Co., Ltd, Beijing, China
| | - Lisha Duan
- Department of Radiology, The Third Hospital of Hebei Medical University, Hebei, China.
| | - Hexiang Wang
- Department of Radiology, The Affiliated Hospital of Qingdao University, 16 Jiangsu Road, Qingdao, Shandong, China.
| |
Collapse
|
8
|
Wang S, Wang X, Yin X, Lv X, Cai J. Differentiating HCC from ICC and prediction of ICC grade based on MRI deep-radiomics: Using lesions and their extended regions. Phys Med 2024; 120:103322. [PMID: 38452430 DOI: 10.1016/j.ejmp.2024.103322] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2023] [Revised: 01/29/2024] [Accepted: 03/03/2024] [Indexed: 03/09/2024] Open
Abstract
PURPOSE This study aimed to evaluate the ability of MRI-based intratumoral and peritumoral radiomics features of liver tumors to differentiate between hepatocellular carcinoma (HCC) and intrahepatic cholangiocarcinoma (ICC) and to predict ICC differentiation. METHODS This study retrospectively collected 87 HCC patients and 75 ICC patients who were confirmed pathologically. The standard region of interest (ROI) of the lesion drawn by the radiologist manually shrank inward and expanded outward to form multiple ROI extended regions. A three-step feature selection method was used to select important radiomics features and convolution features from extended regions. The predictive performance of several machine learning classifiers on dominant feature sets was compared. The extended region performance was assessed by area under the curve (AUC), specificity, sensitivity, F1-score and accuracy. RESULTS The performance of the model is further improved by incorporating convolution features. Compared with the standard ROI, the extended region obtained better prediction performance, among which 6 mm extended region had the best prediction ability (Classification: AUC = 0.96, F1-score = 0.94, Accuracy: 0.94; Grading: AUC = 0.94, F1-score = 0.93, Accuracy = 0.89). CONCLUSION Larger extended region and fusion features can improve tumor predictive performance and have potential value in tumor radiology.
Collapse
Affiliation(s)
- Shuping Wang
- College of Automation Engineering, Nanjing University of Aeronautics and Astronautics, Nanjing 211106, China; College of Electronic and Information Engineering, Hebei University, Baoding 071002, China
| | - Xuehu Wang
- College of Electronic and Information Engineering, Hebei University, Baoding 071002, China; Research Center of Machine Vision Engineering & Technology of Hebei Province, Baoding 071002, China; Key Laboratory of Digital Medical Engineering of Hebei Province, Baoding 071002, China.
| | - Xiaoping Yin
- Affiliated Hospital of Hebei University, Baoding 071000, China
| | - Xiaoyan Lv
- Fifth Medical Center of Chinese PLA General Hospital, Beijing 100039, China
| | - Jianming Cai
- Fifth Medical Center of Chinese PLA General Hospital, Beijing 100039, China.
| |
Collapse
|
9
|
Li Z, Wang F, Zhang H, Xie S, Peng L, Xu H, Wang Y. A radiomics strategy based on CT intra-tumoral and peritumoral regions for preoperative prediction of neoadjuvant chemoradiotherapy for esophageal cancer. EUROPEAN JOURNAL OF SURGICAL ONCOLOGY 2024; 50:108052. [PMID: 38447320 DOI: 10.1016/j.ejso.2024.108052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2023] [Revised: 01/24/2024] [Accepted: 02/21/2024] [Indexed: 03/08/2024]
Abstract
OBJECTIVE Develop a method for selecting esophageal cancer patients achieving pathological complete response with pre-neoadjuvant therapy chest-enhanced CT scans. METHODS Two hundred and one patients from center 1 were enrolled, split into training and testing sets (7:3 ratio), with an external validation set of 30 patients from center 2. Radiomics features from intra-tumoral and peritumoral images were extracted and dimensionally reduced using Student's t-test and least absolute shrinkage and selection operator. Four machine learning classifiers were employed to build models, with the best-performing models selected based on accuracy and stability. ROC curves were utilized to determine the top prediction model, and its generalizability was evaluated on the external validation set. RESULTS Among 16 models, the integrated-XGBoost and integrated-random forest models performed the best, with average ROC AUCs of 0.906 and 0.918, respectively, and RSDs of 6.26 and 6.89 in the training set. In the testing set, AUCs were 0.845 and 0.871, showing no significant difference in ROC curves. External validation set AUCs for integrated-XGBoost and integrated-random forest models were 0.650 and 0.749. CONCLUSION Incorporating peritumoral radiomics features into the analysis enhances predictive performance for esophageal cancer patients undergoing neoadjuvant chemoradiotherapy, paving the way for improved treatment outcomes.
Collapse
Affiliation(s)
- Zhiyang Li
- Department of Thoracic Surgery, West China Hospital, Sichuan University, China; West China School of Medicine, West China Hospital, Sichuan University, China
| | - Fuqiang Wang
- Department of Thoracic Surgery, West China Hospital, Sichuan University, China; West China School of Medicine, West China Hospital, Sichuan University, China
| | - Hanlu Zhang
- Department of Thoracic Surgery, West China Hospital, Sichuan University, China
| | - Shenglong Xie
- Department of Thoracic Surgery, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Lei Peng
- Department of Thoracic Surgery, West China Hospital, Sichuan University, China
| | - Hui Xu
- Department of Radiology, West China Hospital, Sichuan University, China.
| | - Yun Wang
- Department of Thoracic Surgery, West China Hospital, Sichuan University, China.
| |
Collapse
|
10
|
Wang F, Cheng M, Du B, Li J, Li L, Huang W, Gao J. Predicting microvascular invasion in small (≤ 5 cm) hepatocellular carcinomas using radiomics-based peritumoral analysis. Insights Imaging 2024; 15:90. [PMID: 38530498 DOI: 10.1186/s13244-024-01649-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2023] [Accepted: 02/10/2024] [Indexed: 03/28/2024] Open
Abstract
OBJECTIVE We assessed the predictive capacity of computed tomography (CT)-enhanced radiomics models in determining microvascular invasion (MVI) for isolated hepatocellular carcinoma (HCC) ≤ 5 cm within peritumoral margins of 5 and 10 mm. METHODS Radiomics software was used for feature extraction. We used the least absolute shrinkage and selection operator (LASSO) algorithm to establish an effective model to predict patients' preoperative MVI status. RESULTS The area under the curve (AUC) values in the validation sets for the 5- and 10-mm radiomics models concerning arterial tumors were 0.759 and 0.637, respectively. In the portal vein phase, they were 0.626 and 0.693, respectively. Additionally, the combined radiomics model for arterial tumors and the peritumoral 5-mm margin had an AUC value of 0.820. The decision curve showed that the combined tumor and peritumoral radiomics model exhibited a somewhat superior benefit compared to the traditional model, while the fusion model demonstrated an even greater advantage, indicating its significant potential in clinical application. CONCLUSION The 5-mm peritumoral arterial model had superior accuracy and sensitivity in predicting MVI. Moreover, the combined tumor and peritumoral radiomics model outperformed both the individual tumor and peritumoral radiomics models. The most effective combination was the arterial phase tumor and peritumor 5-mm margin combination. Using a fusion model that integrates tumor and peritumoral radiomics and clinical data can aid in the preoperative diagnosis of the MVI of isolated HCC ≤ 5 cm, indicating considerable practical value. CRITICAL RELEVANCE STATEMENT The radiomics model including a 5-mm peritumoral expansion is a promising noninvasive biomarker for preoperatively predicting microvascular invasion in patients diagnosed with a solitary HCC ≤ 5 cm. KEY POINTS • Radiomics features extracted at a 5-mm distance from the tumor could better predict hepatocellular carcinoma microvascular invasion. • Peritumoral radiomics can be used to capture tumor heterogeneity and predict microvascular invasion. • This radiomics model stands as a promising noninvasive biomarker for preoperatively predicting MVI in individuals.
Collapse
Affiliation(s)
- Fang Wang
- Department of Radiology, The First Affiliated Hospital of Zhengzhou University, 1 Jianshe East Road, Erqi, Zhengzhou, Henan, 450052, People's Republic of China
| | - Ming Cheng
- Information Department, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, People's Republic of China
| | - Binbin Du
- Vasculocardiology Department, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, People's Republic of China
| | - Jing Li
- Department of Radiology, The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, People's Republic of China
| | - Liming Li
- Department of Radiology, The First Affiliated Hospital of Zhengzhou University, 1 Jianshe East Road, Erqi, Zhengzhou, Henan, 450052, People's Republic of China
| | - Wenpeng Huang
- Department of Radiology, The First Affiliated Hospital of Zhengzhou University, 1 Jianshe East Road, Erqi, Zhengzhou, Henan, 450052, People's Republic of China
| | - Jianbo Gao
- Department of Radiology, The First Affiliated Hospital of Zhengzhou University, 1 Jianshe East Road, Erqi, Zhengzhou, Henan, 450052, People's Republic of China.
| |
Collapse
|
11
|
Zhang C, Zhong H, Zhao F, Ma ZY, Dai ZJ, Pang GD. Preoperatively predicting vessels encapsulating tumor clusters in hepatocellular carcinoma: Machine learning model based on contrast-enhanced computed tomography. World J Gastrointest Oncol 2024; 16:857-874. [PMID: 38577448 PMCID: PMC10989357 DOI: 10.4251/wjgo.v16.i3.857] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 12/26/2023] [Accepted: 01/29/2024] [Indexed: 03/12/2024] Open
Abstract
BACKGROUND Recently, vessels encapsulating tumor clusters (VETC) was considered as a distinct pattern of tumor vascularization which can primarily facilitate the entry of the whole tumor cluster into the bloodstream in an invasion independent manner, and was regarded as an independent risk factor for poor prognosis in hepatocellular carcinoma (HCC). AIM To develop and validate a preoperative nomogram using contrast-enhanced computed tomography (CECT) to predict the presence of VETC+ in HCC. METHODS We retrospectively evaluated 190 patients with pathologically confirmed HCC who underwent CECT scanning and immunochemical staining for cluster of differentiation 34 at two medical centers. Radiomics analysis was conducted on intratumoral and peritumoral regions in the portal vein phase. Radiomics features, essential for identifying VETC+ HCC, were extracted and utilized to develop a radiomics model using machine learning algorithms in the training set. The model's performance was validated on two separate test sets. Receiver operating characteristic (ROC) analysis was employed to compare the identified performance of three models in predicting the VETC status of HCC on both training and test sets. The most predictive model was then used to constructed a radiomics nomogram that integrated the independent clinical-radiological features. ROC and decision curve analysis were used to assess the performance characteristics of the clinical-radiological features, the radiomics features and the radiomics nomogram. RESULTS The study included 190 individuals from two independent centers, with the majority being male (81%) and a median age of 57 years (interquartile range: 51-66). The area under the curve (AUC) for the combined radiomics features selected from the intratumoral and peritumoral areas were 0.825, 0.788, and 0.680 in the training set and the two test sets. A total of 13 features were selected to construct the Rad-score. The nomogram, combining clinical-radiological and combined radiomics features could accurately predict VETC+ in all three sets, with AUC values of 0.859, 0.848 and 0.757. Decision curve analysis revealed that the radiomics nomogram was more clinically useful than both the clinical-radiological feature and the combined radiomics models. CONCLUSION This study demonstrates the potential utility of a CECT-based radiomics nomogram, incorporating clinical-radiological features and combined radiomics features, in the identification of VETC+ HCC.
Collapse
Affiliation(s)
- Chao Zhang
- Department of Radiology, The Second Hospital of Shandong University, Jinan 250033, Shandong Province, China
| | - Hai Zhong
- Department of Radiology, The Second Hospital of Shandong University, Jinan 250033, Shandong Province, China
| | - Fang Zhao
- Department of Radiology, Qilu Hospital of Shandong University, Jinan 250014, Shandong Province, China
| | - Zhen-Yu Ma
- Department of Radiology, Linglong Yingcheng Hospital, Yantai 265499, Shandong Province, China
| | - Zheng-Jun Dai
- Department of Scientific Research, Huiying Medical Technology Co., Ltd, Beijing 100192, China
| | - Guo-Dong Pang
- Department of Radiology, The Second Hospital of Shandong University, Jinan 250033, Shandong Province, China
| |
Collapse
|
12
|
Cheung BMF. Radiomics in stereotactic body radiotherapy for non-small cell lung cancer: a systematic review and radiomic quality score study. Radiat Oncol J 2024; 42:4-16. [PMID: 38549380 PMCID: PMC10982060 DOI: 10.3857/roj.2023.00612] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Revised: 09/22/2023] [Accepted: 10/10/2023] [Indexed: 04/04/2024] Open
Abstract
PURPOSE Stereotactic body radiotherapy (SBRT) has been widely utilized for curative treatment of early-stage non-small cell lung cancer (NSCLC). It has achieved good local control rate comparable to surgery. Currently, no standard risk model exists for SBRT outcome or complication prediction. Radiomics has the potential to improve clinical outcome prognostication. Here, we reviewed the current literature on the radiomic analyses of thoracic SBRT through the use of radiomic quality score (RQS). MATERIALS AND METHODS Literature search was conducted on PubMed and Embase to retrieve radiomics studies on SBRT for early NSCLC. The literature search included studies up to June 2021. Only full papers published in peer reviewed journals were included. Studies that included metastatic lung cancers or non-lung cancers were excluded. Two independent investigators evaluated each study using the RQS and resolved discrepancies through discussion. RESULTS A total number of 25 studies were analysed. The mean RQS was 7.76 of a maximum score of 36. This corresponds to 21.56% of the maximum score. Lack of feature reduction strategies, external validation and open data sharing were identified as key limitations of the reviewed studies. Meanwhile, various common radiomic signatures across different studies such as gray level co-occurrence matrix Homogeneity and energy have been identified. Multiple robust radiomic models have also been reviewed that may improve outcome or complication prediction. CONCLUSION Radiomics in thoracic SBRT has a very promising future as a prognostication tool. However, larger multicenter prospective studies are required to confirm radiomic signatures. Improvement in future study methodologies can also facilitate its wider application.
Collapse
|
13
|
Su Q, Wang N, Wang B, Wang Y, Dai Z, Zhao X, Li X, Li Q, Yang G, Nie P. Ct-based intratumoral and peritumoral radiomics for predicting prognosis in osteosarcoma: A multicenter study. Eur J Radiol 2024; 172:111350. [PMID: 38309216 DOI: 10.1016/j.ejrad.2024.111350] [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: 08/13/2023] [Revised: 01/09/2024] [Accepted: 01/25/2024] [Indexed: 02/05/2024]
Abstract
PURPOSE To evaluate the performance of CT-based intratumoral, peritumoral and combined radiomics signatures in predicting prognosis in patients with osteosarcoma. METHODS The data of 202 patients (training cohort:102, testing cohort:100) with osteosarcoma admitted to the two hospitals from August 2008 to February 2022 were retrospectively analyzed. Progression free survival (PFS) and overall survival (OS) were used as the end points. The radiomics features were extracted from CT images, three radiomics signatures(RSintratumoral, RSperitumoral, RScombined)were constructed based on intratumoral, peritumoral and combined radiomics features, respectively, and the radiomics score (Rad-score) were calculated. Kaplan-Meier survival analysis was used to evaluate the relationship between the Rad-score with PFS and OS, the Harrell's concordance index (C-index) was used to evaluate the predictive performance of the radiomics signatures. RESULTS Finally, 8, 6, and 21 features were selected for the establishment of RSintratumoral, RSperitumoral, and RScombined, respectively. Kaplan-Meier survival analysis confirmed that the Rad-scores of the three RSs were significantly correlated with the PFS and OS of patients with osteosarcoma. Among the three radiomics signatures, RScombined had better predictive performance, the C-index of PSF prediction was 0.833 in the training cohort and 0.814 in the testing cohort, the C-index of OS prediction was 0.796 in the training cohort and 0.764 in the testing cohort. CONCLUSIONS CT-based intratumoral, peritumoral and combined radiomics signatures can predict the prognosis of patients with osteosarcoma, which may assist in individualized treatment and improving the prognosis of osteosarcoma patients.
Collapse
Affiliation(s)
- Qiushi Su
- Department of Radiology, the Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Ning Wang
- Department of Radiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Bingyan Wang
- Department of Ultrasound, the Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | | | - Zhengjun Dai
- Scientific Research Department, Huiying Medical Technology Co., Ltd, Beijing, China
| | - Xia Zhao
- Department of Radiology, the Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Xiaoli Li
- Department of Radiology, the Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Qiyuan Li
- Department of Radiology, the Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Guangjie Yang
- Department of Nuclear Medicine, the Affiliated Hospital of Qingdao University, Qingdao, Shandong, China.
| | - Pei Nie
- Department of Radiology, the Affiliated Hospital of Qingdao University, Qingdao, Shandong, China.
| |
Collapse
|
14
|
Deboever N, Bayley EM, Eisenberg MA, Hofstetter WL, Mehran RJ, Rice DC, Rajaram R, Roth JA, Sepesi B, Swisher SG, Vaporciyan AA, Walsh GL, Bednarski BK, Morris VK, Antonoff MB. Lung surveillance following colorectal cancer pulmonary metastasectomy: Utilization of clinicopathologic risk factors to guide strategy. J Thorac Cardiovasc Surg 2024; 167:814-819.e2. [PMID: 37495170 DOI: 10.1016/j.jtcvs.2023.07.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 07/03/2023] [Accepted: 07/08/2023] [Indexed: 07/28/2023]
Abstract
BACKGROUND Appropriately selected patients clearly benefit from resection of colorectal cancer (CRC) pulmonary metastases (PMs). However, there remains equipoise surrounding optimal chest surveillance strategies following pulmonary metastasectomy. We aimed to identify risk factors that may inform chest surveillance in this population. METHODS Patients who underwent CRC pulmonary metastasectomy were identified from a single institution's prospectively maintained surgical database. Clinicopathologic and genomic characteristics were collected. Patients were stratified by diagnosis of subsequent PM within 6 months of the index lung resection. Multivariate modeling was used to evaluate risk factors. RESULTS A total of 197 patients met the study's inclusion criteria, of whom 52.3% (n = 103) developed subsequent PM, at a median of 9.51 months following the index metastasectomy. Patients with KRAS alterations (odds ratio [OR], 3.073; 95% confidence interval [CI], 1.363-6.926; P = .007), TP53 alterations (OR, 3.109; 95% CI, 1.318-7.341; P = .010) were found to be at risk of PM diagnosis within 6 months of the index metastasectomy, while those with an APC alteration (OR, .218; 95% CI, 0.080-0.598; P = .003) were protected. Moreover, patients who received systemic therapy within 3 months of the initial PM diagnosis also were more likely to develop early lung recurrence (OR, 2.105; 95% CI, 0.971-4.563; P = .059). CONCLUSIONS Patients with KRAS alterations, TP53 alterations, and no APC alterations developed early recurrence in the lung following pulmonary metastasectomy, as did those who received chemotherapy after their initial PM diagnosis. As such, these groups benefit from early lung imaging after metastasectomy, as chest surveillance protocols should be based on patient-centered clinicopathologic and genomic risk factors.
Collapse
Affiliation(s)
- Nathaniel Deboever
- Department of Thoracic and Cardiovascular Surgery, University of Texas MD Anderson Cancer Center, Houston, Tex
| | - Erin M Bayley
- Department of General Surgery, Baylor University, Houston, Tex
| | - Michael A Eisenberg
- Department of Thoracic and Cardiovascular Surgery, University of Texas MD Anderson Cancer Center, Houston, Tex
| | - Wayne L Hofstetter
- Department of Thoracic and Cardiovascular Surgery, University of Texas MD Anderson Cancer Center, Houston, Tex
| | - Reza J Mehran
- Department of Thoracic and Cardiovascular Surgery, University of Texas MD Anderson Cancer Center, Houston, Tex
| | - David C Rice
- Department of Thoracic and Cardiovascular Surgery, University of Texas MD Anderson Cancer Center, Houston, Tex
| | - Ravi Rajaram
- Department of Thoracic and Cardiovascular Surgery, University of Texas MD Anderson Cancer Center, Houston, Tex
| | - Jack A Roth
- Department of Thoracic and Cardiovascular Surgery, University of Texas MD Anderson Cancer Center, Houston, Tex
| | - Boris Sepesi
- Department of Thoracic and Cardiovascular Surgery, University of Texas MD Anderson Cancer Center, Houston, Tex
| | - Stephen G Swisher
- Department of Thoracic and Cardiovascular Surgery, University of Texas MD Anderson Cancer Center, Houston, Tex
| | - Ara A Vaporciyan
- Department of Thoracic and Cardiovascular Surgery, University of Texas MD Anderson Cancer Center, Houston, Tex
| | - Garrett L Walsh
- Department of Thoracic and Cardiovascular Surgery, University of Texas MD Anderson Cancer Center, Houston, Tex
| | - Brian K Bednarski
- Department of Colon and Rectal Surgery, University of Texas MD Anderson Cancer Center, Houston, Tex
| | - Van K Morris
- Department of Gastrointestinal Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, Tex
| | - Mara B Antonoff
- Department of Thoracic and Cardiovascular Surgery, University of Texas MD Anderson Cancer Center, Houston, Tex.
| |
Collapse
|
15
|
Mottola M, Golfieri R, Bevilacqua A. The Effectiveness of an Adaptive Method to Analyse the Transition between Tumour and Peritumour for Answering Two Clinical Questions in Cancer Imaging. SENSORS (BASEL, SWITZERLAND) 2024; 24:1156. [PMID: 38400314 PMCID: PMC10893370 DOI: 10.3390/s24041156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Revised: 01/29/2024] [Accepted: 02/06/2024] [Indexed: 02/25/2024]
Abstract
Based on the well-known role of peritumour characterization in cancer imaging to improve the early diagnosis and timeliness of clinical decisions, this study innovated a state-of-the-art approach for peritumour analysis, mainly relying on extending tumour segmentation by a predefined fixed size. We present a novel, adaptive method to investigate the zone of transition, bestriding tumour and peritumour, thought of as an annular-like shaped area, and detected by analysing gradient variations along tumour edges. For method validation, we applied it on two datasets (hepatocellular carcinoma and locally advanced rectal cancer) imaged by different modalities and exploited the zone of transition regions as well as the peritumour ones derived by adopting the literature approach for building predictive models. To measure the zone of transition's benefits, we compared the predictivity of models relying on both "standard" and novel peritumour regions. The main comparison metrics were informedness, specificity and sensitivity. As regards hepatocellular carcinoma, having circular and regular shape, all models showed similar performance (informedness = 0.69, sensitivity = 84%, specificity = 85%). As regards locally advanced rectal cancer, with jagged contours, the zone of transition led to the best informedness of 0.68 (sensitivity = 89%, specificity = 79%). The zone of transition advantages include detecting the peritumour adaptively, even when not visually noticeable, and minimizing the risk (higher in the literature approach) of including adjacent diverse structures, which was clearly highlighted during image gradient analysis.
Collapse
Affiliation(s)
- Margherita Mottola
- Alma Mater Research Institute on Global Challenges and Climate Change (Alma Climate), University of Bologna, 40126 Bologna, Italy;
| | - Rita Golfieri
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, 40138 Bologna, Italy;
| | - Alessandro Bevilacqua
- Department of Computer Science and Engineering (DISI), University of Bologna, 40126 Bologna, Italy
- Advanced Research Center on Electronic Systems (ARCES), University of Bologna, 40125 Bologna, Italy
| |
Collapse
|
16
|
Li S, Su X, Peng J, Chen N, Liu Y, Zhang S, Shao H, Tan Q, Yang X, Liu Y, Gong Q, Yue Q. Development and External Validation of an MRI-based Radiomics Nomogram to Distinguish Circumscribed Astrocytic Gliomas and Diffuse Gliomas: A Multicenter Study. Acad Radiol 2024; 31:639-647. [PMID: 37507329 DOI: 10.1016/j.acra.2023.06.033] [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: 06/02/2023] [Revised: 06/27/2023] [Accepted: 06/29/2023] [Indexed: 07/30/2023]
Abstract
RATIONALE AND OBJECTIVES The 5th edition of the World Health Organization classification of tumors of the Central Nervous System (WHO CNS) has introduced the term "diffuse" and its counterpart "circumscribed" to the category of gliomas. This study aimed to develop and validate models for distinguishing circumscribed astrocytic gliomas (CAGs) from diffuse gliomas (DGs). MATERIALS AND METHODS We retrospectively analyzed magnetic resonance imaging (MRI) data from patients with CAGs and DGs across three institutions. After tumor segmentation, three volume of interest (VOI) types were obtained: VOItumor and peritumor, VOIwhole, and VOIinterface. Clinical and combined models (incorporating radiomics and clinical features) were also established. To address imbalances in training dataset, Synthetic Minority Oversampling Technique was employed. RESULTS A total of 475 patients (DGs: n = 338, CAGs: n = 137) were analyzed. The VOIinterface model demonstrated the best performance for differentiating CAGs from DGs, achieving an area under the curve (AUC) of 0.806 and area under the precision-recall curve (PRAUC)of 0.894 in the cross-validation set. Using analysis of variance (ANOVA) feature selector and Support Vector Machine (SVM) classifier, seven features were selected. The model achieved an AUC and AUPRC of 0.912 and 0.972 in the internal validation dataset, and 0.897 and 0.930 in the external validation dataset. The combined model, incorporating interface radiomics and clinical features, showed improved performance in the external validation set, with an AUC of 0.94 and PRAUC of 0.959. CONCLUSION Radiomics models incorporating the peritumoral area demonstrate greater potential for distinguishing CAGs from DGs compared to intratumoral models. These findings may hold promise for evaluating tumor nature before surgery and improving clinical management of glioma patients.
Collapse
Affiliation(s)
- Shuang Li
- Huaxi MR Research Center (HMRRC), Department of Radiology, West China Hospital of Sichuan University, Chengdu, Sichuan, China (S.L., X.S., S.Z., H.S., Q.T., Q.G.); Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, Chengdu, Sichuan, China (S.L.); Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital of Sichuan University, Chengdu, Sichuan, China (S.L.)
| | - Xiaorui Su
- Huaxi MR Research Center (HMRRC), Department of Radiology, West China Hospital of Sichuan University, Chengdu, Sichuan, China (S.L., X.S., S.Z., H.S., Q.T., Q.G.)
| | - Juan Peng
- Department of Radiology, The First Affiliated Hospital, Chongqing Medical University, Chongqing, China (J.P.)
| | - Ni Chen
- Department of Pathology, West China Hospital of Sichuan University, Chengdu, Sichuan, China (N.C.)
| | - Yanhui Liu
- Department of Neurosurgery, West China Hospital of Sichuan University, Chengdu, Sichuan, China (Y.L.)
| | - Simin Zhang
- Huaxi MR Research Center (HMRRC), Department of Radiology, West China Hospital of Sichuan University, Chengdu, Sichuan, China (S.L., X.S., S.Z., H.S., Q.T., Q.G.)
| | - Hanbing Shao
- Huaxi MR Research Center (HMRRC), Department of Radiology, West China Hospital of Sichuan University, Chengdu, Sichuan, China (S.L., X.S., S.Z., H.S., Q.T., Q.G.)
| | - Qiaoyue Tan
- Huaxi MR Research Center (HMRRC), Department of Radiology, West China Hospital of Sichuan University, Chengdu, Sichuan, China (S.L., X.S., S.Z., H.S., Q.T., Q.G.); Division of Radiation Physics, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital of Sichuan University, Chengdu, Sichuan, China (Q.T.)
| | - Xibiao Yang
- Department of Radiology, West China Hospital of Sichuan University, Chengdu, Sichuan, China (X.Y., Q.Y.)
| | - Yaou Liu
- Department of Radiology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China (Y.L.)
| | - Qiyong Gong
- Huaxi MR Research Center (HMRRC), Department of Radiology, West China Hospital of Sichuan University, Chengdu, Sichuan, China (S.L., X.S., S.Z., H.S., Q.T., Q.G.); Department of Radiology, West China Xiamen Hospital of Sichuan University, Xiamen, Fujian, China (Q.G.)
| | - Qiang Yue
- Department of Radiology, West China Hospital of Sichuan University, Chengdu, Sichuan, China (X.Y., Q.Y.).
| |
Collapse
|
17
|
Lu T, Ma J, Zou J, Jiang C, Li Y, Han J. CT-based intratumoral and peritumoral deep transfer learning features prediction of lymph node metastasis in non-small cell lung cancer. JOURNAL OF X-RAY SCIENCE AND TECHNOLOGY 2024; 32:597-609. [PMID: 38578874 DOI: 10.3233/xst-230326] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/07/2024]
Abstract
BACKGROUND The main metastatic route for lung cancer is lymph node metastasis, and studies have shown that non-small cell lung cancer (NSCLC) has a high risk of lymph node infiltration. OBJECTIVE This study aimed to compare the performance of handcrafted radiomics (HR) features and deep transfer learning (DTL) features in Computed Tomography (CT) of intratumoral and peritumoral regions in predicting the metastatic status of NSCLC lymph nodes in different machine learning classifier models. METHODS We retrospectively collected data of 199 patients with pathologically confirmed NSCLC. All patients were divided into training (n = 159) and validation (n = 40) cohorts, respectively. The best HR and DTL features in the intratumoral and peritumoral regions were extracted and selected, respectively. Support Vector Machine (SVM), k-Nearest Neighbors (KNN), Light Gradient Boosting Machine (Light GBM), Multilayer Perceptron (MLP), and Logistic Regression (LR) models were constructed, and the performance of the models was evaluated. RESULTS Among the five models in the training and validation cohorts, the LR classifier model performed best in terms of HR and DTL features. The AUCs of the training cohort were 0.841 (95% CI: 0.776-0.907) and 0.955 (95% CI: 0.926-0.983), and the AUCs of the validation cohort were 0.812 (95% CI: 0.677-0.948) and 0.893 (95% CI: 0.795-0.991), respectively. The DTL signature was superior to the handcrafted radiomics signature. CONCLUSIONS Compared with the radiomics signature, the DTL signature constructed based on intratumoral and peritumoral areas in CT can better predict NSCLC lymph node metastasis.
Collapse
Affiliation(s)
- Tianyu Lu
- Department of Radiology, The First Hospital of Jiaxing or The Affiliated Hospital of Jiaxing University, Jiaxing, China
| | - Jianbing Ma
- Department of Radiology, The First Hospital of Jiaxing or The Affiliated Hospital of Jiaxing University, Jiaxing, China
| | - Jiajun Zou
- Department of Radiology, The First Hospital of Jiaxing or The Affiliated Hospital of Jiaxing University, Jiaxing, China
| | - Chenxu Jiang
- Department of Radiology, The First Hospital of Jiaxing or The Affiliated Hospital of Jiaxing University, Jiaxing, China
| | - Yangyang Li
- Department of Radiology, The First Hospital of Jiaxing or The Affiliated Hospital of Jiaxing University, Jiaxing, China
| | - Jun Han
- Department of Radiology, The First Hospital of Jiaxing or The Affiliated Hospital of Jiaxing University, Jiaxing, China
| |
Collapse
|
18
|
Li Q, Huang Y, Xia Y, Li M, Tang W, Zhang M, Zhao Z. Radiogenomics for predicting microsatellite instability status and PD-L1 expression with machine learning in endometrial cancers: A multicenter study. Heliyon 2023; 9:e23166. [PMID: 38149198 PMCID: PMC10750045 DOI: 10.1016/j.heliyon.2023.e23166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Revised: 11/22/2023] [Accepted: 11/28/2023] [Indexed: 12/28/2023] Open
Abstract
Purpose To evaluate the effectiveness of machine learning model based on magnetic resonance imaging (MRI) in identifying microsatellite instability (MSI) status and PD-L1 expression in endometrial cancer (EC). Methods This retrospective study included 82 EC patients from 2 independent centers. Radiomics features from the intratumoral and peritumoral regions, obtained from four conventional MRI sequences (T2-weighted images; contrast-enhanced T1-weighted images; diffusion-weighted images; apparent diffusion coefficient), were combined with clinicopathologic characteristics to develop machine learning model for predicting MSI status and PD-L1 expression. 60 patients from center 1 were used as the training set for model construction, while 22 patients from center 2 were used as an external validation set for model evaluation. Results For predicting MSI status, the clinicopathologic model, radscore model, and combination model achieved area under the curves (AUCs) of 0.728, 0.833, and 0.889 in the training set, respectively, and 0.595, 0.790, and 0.848 in the validation set, respectively. For predicting PD-L1 expression, the clinicopathologic model, radscore model, and combination model achieved AUCs of 0.648, 0.814, and 0.834 in the training set, respectively, and 0.660, 0.708, and 0.764 in the validation set, respectively. Calibration curve analysis and decision curve analysis demonstrated good calibration and clinical utility of the combination model. Conclusion The machine learning model incorporating MRI-based radiomics features and clinicopathologic characteristics could be a potential tool for predicting MSI status and PD-L1 expression in EC. This approach may contribute to precision medicine for EC patients.
Collapse
Affiliation(s)
- Qianling Li
- Department of Radiology, Shaoxing People's Hospital (Shaoxing Hospital, Zhejiang University School of Medicine), Zhejiang University School of Medicine, Shaoxing, 312000, China
| | - Ya'nan Huang
- Department of Radiology, Shaoxing People's Hospital (Shaoxing Hospital, Zhejiang University School of Medicine), Shaoxing, 312000, China
| | - Yang Xia
- Department of Radiology, Shaoxing Maternity and Child Health Care Hospital, Shaoxing, 312000, China
| | - Meiping Li
- Department of Pathology, Shaoxing Maternity and Child Health Care Hospital, Shaoxing, Zhejiang, 312000, China
| | - Wei Tang
- Department of Radiology, Shaoxing People's Hospital (Shaoxing Hospital, Zhejiang University School of Medicine), Shaoxing, 312000, China
| | - Minming Zhang
- Department of Radiology, The Second Affiliated Hospital of Zhejiang University, Hangzhou, 310000, China
| | - Zhenhua Zhao
- Department of Radiology, Shaoxing People's Hospital (Shaoxing Hospital, Zhejiang University School of Medicine), Shaoxing, 312000, China
| |
Collapse
|
19
|
Roisman LC, Kian W, Anoze A, Fuchs V, Spector M, Steiner R, Kassel L, Rechnitzer G, Fried I, Peled N, Bogot NR. Radiological artificial intelligence - predicting personalized immunotherapy outcomes in lung cancer. NPJ Precis Oncol 2023; 7:125. [PMID: 37990050 PMCID: PMC10663598 DOI: 10.1038/s41698-023-00473-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Accepted: 10/24/2023] [Indexed: 11/23/2023] Open
Abstract
Personalized medicine has revolutionized approaches to treatment in the field of lung cancer by enabling therapies to be specific to each patient. However, physicians encounter an immense number of challenges in providing the optimal treatment regimen for the individual given the sheer complexity of clinical aspects such as tumor molecular profile, tumor microenvironment, expected adverse events, acquired or inherent resistance mechanisms, the development of brain metastases, the limited availability of biomarkers and the choice of combination therapy. The integration of innovative next-generation technologies such as deep learning-a subset of machine learning-and radiomics has the potential to transform the field by supporting clinical decision making in cancer treatment and the delivery of precision therapies while integrating numerous clinical considerations. In this review, we present a brief explanation of the available technologies, the benefits of using these technologies in predicting immunotherapy response in lung cancer, and the expected future challenges in the context of precision medicine.
Collapse
Affiliation(s)
- Laila C Roisman
- The Hebrew University, Helmsley Cancer Center, Shaare Zedek Medical Center, Jerusalem, Israel.
- Ben-Gurion University of the Negev, Be'er Sheva, Israel.
| | - Waleed Kian
- The Hebrew University, Helmsley Cancer Center, Shaare Zedek Medical Center, Jerusalem, Israel
- The Institute of Oncology, Assuta Ashdod, Ashdod, Israel
| | - Alaa Anoze
- The Hebrew University, Helmsley Cancer Center, Shaare Zedek Medical Center, Jerusalem, Israel
| | - Vered Fuchs
- Ben-Gurion University of the Negev, Be'er Sheva, Israel
| | - Maria Spector
- The Department of Radiology, Shaare Zedek Medical Center, Jerusalem, Israel
| | - Roee Steiner
- The Institute for Nuclear Medicine, Shaare Zedek Medical Center, Jerusalem, Israel
| | - Levi Kassel
- The Hebrew University, Helmsley Cancer Center, Shaare Zedek Medical Center, Jerusalem, Israel
| | - Gilad Rechnitzer
- The Hebrew University, Helmsley Cancer Center, Shaare Zedek Medical Center, Jerusalem, Israel
| | - Iris Fried
- The Hebrew University, Helmsley Cancer Center, Shaare Zedek Medical Center, Jerusalem, Israel
| | - Nir Peled
- The Hebrew University, Helmsley Cancer Center, Shaare Zedek Medical Center, Jerusalem, Israel.
| | - Naama R Bogot
- The Department of Radiology, Shaare Zedek Medical Center, Jerusalem, Israel
| |
Collapse
|
20
|
Zhang L, Yang Y, Wang T, Chen X, Tang M, Deng J, Cai Z, Cui W. Intratumoral and peritumoral MRI-based radiomics prediction of histopathological grade in soft tissue sarcomas: a two-center study. Cancer Imaging 2023; 23:103. [PMID: 37885031 PMCID: PMC10601231 DOI: 10.1186/s40644-023-00622-2] [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: 06/20/2023] [Accepted: 10/16/2023] [Indexed: 10/28/2023] Open
Abstract
OBJECTIVES This study aims to develop a model based on intratumoral and peritumoral radiomics from fat-suppressed T2-weighted(FS-T2WI) images to predict the histopathological grade of soft tissue sarcoma (STS). METHODS This retrospective study included 160 patients with STS from two centers, of which 82 were low-grade and 78were high-grade. Radiomics features were extracted and selected from the region of tumor mass volume (TMV) and peritumoral tumor volume (PTV) respectively. The TMV, PTV, and combined(TM-PTV) radiomics models were established in the training cohort (n = 111)for the prediction of histopathological grade. Finally, a radiomics nomogram was constructed by combining the TM-PTV radiomics signature (Rad-score) and the selected clinical-MRI predictor. The ROC and calibration curves were used to determine the performance of the TMV, PTV, and TM-PTV models in the training and validation cohort (n = 49). The decision curve analysis (DCA) and calibration curves were used to investigate the clinical usefulness and calibration of the nomogram, respectively. RESULTS The TMV model, PTV model, and TM-PTV model had AUCs of 0.835, 0.879, and 0.917 in the training cohort and 0.811, 0.756, 0.896 in the validation cohort. The nomogram, including the TM-PTV signatures and peritumoral hyperintensity, achieved good calibration and discrimination with a C-index of 0.948 (95% CI, 0.906 to 0.990) in the training cohort and 0.921 (95% CI, 0.840 to 0.995) in the validation cohort. Decision curve analysis demonstrated the clinical usefulness of the nomogram. CONCLUSION The proposed model based on intratumoral and peritumoral radiomics showed good performance in distinguishing low-grade from high-grade STSs.
Collapse
Affiliation(s)
- Liyuan Zhang
- Department of Plastic Surgery, Sichuan Provincial People's Hospital, School of Medicine,University of Electronic Science and Technology of China, Chengdu, 610000, People's Republic of China
| | - Yang Yang
- Department of Radiology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610000, People's Republic of China
| | - Ting Wang
- Department of Plastic Surgery, The First People's Hospital of Yibin, Yibin, 644000, People's Republic of China
| | - Xi Chen
- Sichuan College of Traditional Chinese Medicine, Mianyang, 621000, People's Republic of China
| | - Mingyue Tang
- Department of Radiology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610000, People's Republic of China
| | - Junnan Deng
- Department of Plastic Surgery, Sichuan Provincial People's Hospital, School of Medicine,University of Electronic Science and Technology of China, Chengdu, 610000, People's Republic of China
| | - Zhen Cai
- Department of Plastic Surgery, Sichuan Provincial People's Hospital, School of Medicine,University of Electronic Science and Technology of China, Chengdu, 610000, People's Republic of China.
| | - Wei Cui
- Department of Plastic Surgery, Sichuan Provincial People's Hospital, School of Medicine,University of Electronic Science and Technology of China, Chengdu, 610000, People's Republic of China.
| |
Collapse
|
21
|
Crispin-Ortuzar M, Woitek R, Reinius MAV, Moore E, Beer L, Bura V, Rundo L, McCague C, Ursprung S, Escudero Sanchez L, Martin-Gonzalez P, Mouliere F, Chandrananda D, Morris J, Goranova T, Piskorz AM, Singh N, Sahdev A, Pintican R, Zerunian M, Rosenfeld N, Addley H, Jimenez-Linan M, Markowetz F, Sala E, Brenton JD. Integrated radiogenomics models predict response to neoadjuvant chemotherapy in high grade serous ovarian cancer. Nat Commun 2023; 14:6756. [PMID: 37875466 PMCID: PMC10598212 DOI: 10.1038/s41467-023-41820-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Accepted: 09/20/2023] [Indexed: 10/26/2023] Open
Abstract
High grade serous ovarian carcinoma (HGSOC) is a highly heterogeneous disease that typically presents at an advanced, metastatic state. The multi-scale complexity of HGSOC is a major obstacle to predicting response to neoadjuvant chemotherapy (NACT) and understanding critical determinants of response. Here we present a framework to predict the response of HGSOC patients to NACT integrating baseline clinical, blood-based, and radiomic biomarkers extracted from all primary and metastatic lesions. We use an ensemble machine learning model trained to predict the change in total disease volume using data obtained at diagnosis (n = 72). The model is validated in an internal hold-out cohort (n = 20) and an independent external patient cohort (n = 42). In the external cohort the integrated radiomics model reduces the prediction error by 8% with respect to the clinical model, achieving an AUC of 0.78 for RECIST 1.1 classification compared to 0.47 for the clinical model. Our results emphasize the value of including radiomics data in integrative models of treatment response and provide methods for developing new biomarker-based clinical trials of NACT in HGSOC.
Collapse
Affiliation(s)
- Mireia Crispin-Ortuzar
- Department of Oncology, University of Cambridge, Cambridge, UK.
- Cancer Research UK Cambridge Centre, University of Cambridge, Cambridge, UK.
| | - Ramona Woitek
- Cancer Research UK Cambridge Centre, University of Cambridge, Cambridge, UK
- Department of Radiology, University of Cambridge, Cambridge, UK
- Centre for Medical Image Analysis and AI (MIAAI), Danube Private University, Krems, Austria
| | - Marika A V Reinius
- Cancer Research UK Cambridge Centre, University of Cambridge, Cambridge, UK
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK
- Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Elizabeth Moore
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK
| | - Lucian Beer
- Cancer Research UK Cambridge Centre, University of Cambridge, Cambridge, UK
- Department of Radiology, University of Cambridge, Cambridge, UK
- Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, Vienna, Austria
| | - Vlad Bura
- Cancer Research UK Cambridge Centre, University of Cambridge, Cambridge, UK
- Department of Radiology, University of Cambridge, Cambridge, UK
| | - Leonardo Rundo
- Cancer Research UK Cambridge Centre, University of Cambridge, Cambridge, UK
- Department of Radiology, University of Cambridge, Cambridge, UK
- Department of Information and Electrical Engineering and Applied Mathematics, University of Salerno, Fisciano, SA, Italy
| | - Cathal McCague
- Cancer Research UK Cambridge Centre, University of Cambridge, Cambridge, UK
- Department of Radiology, University of Cambridge, Cambridge, UK
- Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Stephan Ursprung
- Cancer Research UK Cambridge Centre, University of Cambridge, Cambridge, UK
- Department of Radiology, University of Cambridge, Cambridge, UK
| | - Lorena Escudero Sanchez
- Cancer Research UK Cambridge Centre, University of Cambridge, Cambridge, UK
- Department of Radiology, University of Cambridge, Cambridge, UK
| | - Paula Martin-Gonzalez
- Cancer Research UK Cambridge Centre, University of Cambridge, Cambridge, UK
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK
| | - Florent Mouliere
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK
- Department of Pathology, Amsterdam UMC location Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | | | - James Morris
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK
| | - Teodora Goranova
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK
| | - Anna M Piskorz
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK
| | - Naveena Singh
- Department of Cellular Pathology, Barts Health NHS Trust, London, UK
| | - Anju Sahdev
- Department of Radiology, Barts Health NHS Trust, London, UK
| | - Roxana Pintican
- "Iuliu Hatieganu" University of Medicine and Pharmacy, Cluj-Napoca, Romania
- Department of Radiology, County Clinical Emergency Hospital, Cluj-Napoca, Romania
| | - Marta Zerunian
- Department of Surgical and Medical Sciences and Translational Medicine, Sapienza University of Rome-Sant'Andrea University Hospital, Rome, Italy
| | - Nitzan Rosenfeld
- Cancer Research UK Cambridge Centre, University of Cambridge, Cambridge, UK
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK
| | - Helen Addley
- Cancer Research UK Cambridge Centre, University of Cambridge, Cambridge, UK
- Department of Radiology, University of Cambridge, Cambridge, UK
- Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Mercedes Jimenez-Linan
- Cancer Research UK Cambridge Centre, University of Cambridge, Cambridge, UK
- Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Florian Markowetz
- Cancer Research UK Cambridge Centre, University of Cambridge, Cambridge, UK
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK
| | - Evis Sala
- Cancer Research UK Cambridge Centre, University of Cambridge, Cambridge, UK
- Department of Radiology, University of Cambridge, Cambridge, UK
- Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
- Dipartimento di Scienze Radiologiche ed Ematologiche, Universita Cattolica del Sacro Cuore, Rome, Italy
- Dipartimento Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
- Western Balkans University, Tirana, Albania
| | - James D Brenton
- Cancer Research UK Cambridge Centre, University of Cambridge, Cambridge, UK
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK
- Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| |
Collapse
|
22
|
Sun J, Li F, Yang J, Lin C, Zhou X, Liu N, Zhang B, Song G, Wang W, Huang C, Song Z, Shi L. Pretherapy investigations using highly robust visualized biomarkers from CT imaging by multiple machine-learning techniques toward its prognosis prediction for ALK-inhibitor therapy in NSCLC: a feasibility study. J Cancer Res Clin Oncol 2023; 149:7341-7353. [PMID: 36928998 DOI: 10.1007/s00432-023-04615-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2022] [Accepted: 01/27/2023] [Indexed: 03/18/2023]
Abstract
PURPOSE Molecularly targeted therapy has revolutionized the therapeutic landscape and is emerging as the first-line treatment option for ALK-rearranged non-small-cell lung cancer (NSCLC). In this study, the highly informative and robust biomarkers based on pre-treatment CT images and clinicopathologic features will be developed and validated to predict the prognosis for ALK-inhibitor therapy in NSCLC patients. METHODS A total of 161 ALK-positive NSCLC patients treated with ALK inhibitors were retrospectively collected as training, validation and test sets from multi-center institutions. Cox proportional hazard regression (CPH) penalized by LASSO and random survival forest (RSF) coupled with recursive feature elimination (RFE) were used for radiomics and clinical features identification and model construction. An overlapping post-processing method was extra added to training process to investigate the stronger biomarker on the whole set. RESULTS 123 of the collected cases progressed after a median follow-up of 15.5 months (IQR, 8.3-25.3). The T and M staging, pericardial effusion, age and ALK inhibitor-alectinib were determined as significant predictors in the survival analysis. Furthermore, we visualized the finally retained 4 radiomics feature. The RSF models built from overlapping-processed clinical and radiomics features respectively reached the maximum C-index of 0.68 and 0.75,but the combination of them,radioclinical signature, improved the score to 0.78. The model on the validation and external test datasets yielded the C-index of 0.73 and 0.79, with the iAUC of 0.76 and 0.83, the IBS of 0.119 and 0.112. CONCLUSION With respect to a simple selection strategy of overlapping optimal radiomics and clinical features from different survival models may promote better progression-free survival(PFS) prediction than conventional survival analysis, which provides a potential method for guiding personalized pre-treatment options of NSCLC.
Collapse
Affiliation(s)
- Jingjing Sun
- Department of Radiology, Zhejiang Cancer Hospital, Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, 310022, Zhejiang, China
| | - Feng Li
- Department of Research Collaboration, R&D Center, Beijing Deepwise & League of PHD Technology Co, Ltd, Beijing, 100080, China
| | - Jiantao Yang
- Department of Radiology, Zhejiang Cancer Hospital, Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, 310022, Zhejiang, China
| | - Chen Lin
- Department of Radiology, Zhejiang Cancer Hospital, Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, 310022, Zhejiang, China
| | - Xianglan Zhou
- Department of Radiology, Zhejiang Cancer Hospital, Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, 310022, Zhejiang, China
| | - Na Liu
- Department of Radiology, Zhejiang Cancer Hospital, Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, 310022, Zhejiang, China
| | - Bingqian Zhang
- Department of Radiology, Zhejiang Cancer Hospital, Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, 310022, Zhejiang, China
| | - Ge Song
- Department of Radiology, Zhejiang Cancer Hospital, Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, 310022, Zhejiang, China
| | - Wenxian Wang
- Department of Medical Oncology, Zhejiang Cancer Hospital, Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, 310022, Zhejiang, China
| | - Chencui Huang
- Department of Research Collaboration, R&D Center, Beijing Deepwise & League of PHD Technology Co, Ltd, Beijing, 100080, China
| | - Zhengbo Song
- Department of Clinical Trial, Zhejiang Cancer Hospital, Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, 310022, Zhejiang, China.
| | - Lei Shi
- Department of Radiology, Zhejiang Cancer Hospital, Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, 310022, Zhejiang, China.
| |
Collapse
|
23
|
Wilk AM, Kozłowska E, Borys D, D’Amico A, Fujarewicz K, Gorczewska I, Debosz-Suwinska I, Suwinski R, Smieja J, Swierniak A. Radiomic signature accurately predicts the risk of metastatic dissemination in late-stage non-small cell lung cancer. Transl Lung Cancer Res 2023; 12:1372-1383. [PMID: 37577306 PMCID: PMC10413035 DOI: 10.21037/tlcr-23-60] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Accepted: 06/13/2023] [Indexed: 08/15/2023]
Abstract
Background Non-small cell lung cancer (NSCLC) is the most common type of lung cancer, and the median overall survival (OS) is approximately 2-3 years among patients with stage III disease. Furthermore, it is one of the deadliest types of cancer globally due to non-specific symptoms and the lack of a biomarker for early detection. The most important decision that clinicians need to make after a lung cancer diagnosis is the selection of a treatment schedule. This decision is based on, among others factors, the risk of developing metastasis. Methods A cohort of 115 NSCLC patients treated using chemotherapy and radiotherapy (RT) with curative intent was retrospectively collated and included patients for whom positron emission tomography/computed tomography (PET/CT) images, acquired before RT, were available. The PET/CT images were used to compute radiomic features extracted from a region of interest (ROI), the primary tumor. Radiomic and clinical features were then classified to stratify the patients into short and long time to metastasis, and regression analysis was used to predict the risk of metastasis. Results Classification based on binarized metastasis-free survival (MFS) was applied with moderate success. Indeed, an accuracy of 0.73 was obtained for the selection of features based on the Wilcoxon test and logistic regression model. However, the Cox regression model for metastasis risk prediction performed very well, with a concordance index (C-index) score equal to 0.84. Conclusions It is possible to accurately predict the risk of metastasis in NSCLC patients based on radiomic features. The results demonstrate the potential use of features extracted from cancer imaging in predicting the risk of metastasis.
Collapse
Affiliation(s)
- Agata Małgorzata Wilk
- Department of Systems Biology and Engineering, Silesian University of Technology, Gliwice, Poland
- Department of Biostatistics and Bioinformatics, Maria Sklodowska-Curie National Research Institute of Oncology, Gliwice Branch, Gliwice, Poland
| | - Emilia Kozłowska
- Department of Systems Biology and Engineering, Silesian University of Technology, Gliwice, Poland
| | - Damian Borys
- Department of Systems Biology and Engineering, Silesian University of Technology, Gliwice, Poland
- Department of Nuclear Medicine and Endocrine Oncology, PET Diagnostics Unit, Maria Sklodowska-Curie National Research Institute of Oncology, Gliwice Branch, Gliwice, Poland
| | - Andrea D’Amico
- Department of Nuclear Medicine and Endocrine Oncology, PET Diagnostics Unit, Maria Sklodowska-Curie National Research Institute of Oncology, Gliwice Branch, Gliwice, Poland
| | - Krzysztof Fujarewicz
- Department of Systems Biology and Engineering, Silesian University of Technology, Gliwice, Poland
| | - Izabela Gorczewska
- Department of Nuclear Medicine and Endocrine Oncology, PET Diagnostics Unit, Maria Sklodowska-Curie National Research Institute of Oncology, Gliwice Branch, Gliwice, Poland
| | - Iwona Debosz-Suwinska
- Department of Radiotherapy, Maria Sklodowska-Curie National Research Institute of Oncology, Gliwice Branch, Gliwice, Poland
| | - Rafał Suwinski
- II Radiotherapy and Chemotherapy Clinic, Maria Sklodowska-Curie National Research Institute of Oncology, Gliwice Branch, Gliwice, Poland
| | - Jarosław Smieja
- Department of Systems Biology and Engineering, Silesian University of Technology, Gliwice, Poland
| | - Andrzej Swierniak
- Department of Systems Biology and Engineering, Silesian University of Technology, Gliwice, Poland
| |
Collapse
|
24
|
Cilla S, Pistilli D, Romano C, Macchia G, Pierro A, Arcelli A, Buwenge M, Morganti AG, Deodato F. CT-based radiomics prediction of complete response after stereotactic body radiation therapy for patients with lung metastases. Strahlenther Onkol 2023:10.1007/s00066-023-02086-6. [PMID: 37256303 DOI: 10.1007/s00066-023-02086-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Accepted: 04/11/2023] [Indexed: 06/01/2023]
Abstract
PURPOSE Stereotactic body radiotherapy (SBRT) is a key treatment modality for lung cancer patients. This study aims to develop a machine learning-based prediction model of complete response for lung oligometastatic cancer patients undergoing SBRT. MATERIALS AND METHODS CT images of 80 pulmonary oligometastases from 56 patients treated with SBRT were analyzed. The gross tumor volumes (GTV) were contoured on CT images. Patients that achieved complete response (CR) at 4 months were defined as responders. For each GTV, 107 radiomic features were extracted using the Pyradiomics software. The concordance correlation coefficients (CCC) between the region of interest (ROI)-based radiomics features obtained by the two segmentations were calculated. Pairwise feature interdependencies were evaluated using the Spearman rank correlation coefficient. The association of clinical variables and radiomics features with CR was evaluated with univariate logistic regression. Two supervised machine learning models, the logistic regression (LR) and the classification and regression tree analysis (CART), were trained to predict CR. The models were cross-validated using a five-fold cross-validation. The performance of models was assessed by receiver operating characteristic curve (ROC) and class-specific accuracy, precision, recall, and F1-measure evaluation metrics. RESULTS Complete response was associated with four radiomics features, namely the surface to volume ratio (SVR; p = 0.003), the skewness (Skew; p = 0.027), the correlation (Corr; p = 0.024), and the grey normalized level uniformity (GNLU; p = 0.015). No significant relationship between clinical parameters and CR was found. In the validation set, the developed LR and CART machine learning models had an accuracy, precision, and recall of 0.644 and 0.750, 0.644 and 0.651, and 0.635 and 0.754, respectively. The area under the curve for CR prediction was 0.707 and 0.753 for the LR and CART models, respectively. CONCLUSION This analysis demonstrates that radiomics features obtained from pretreatment CT could predict complete response of lung oligometastases following SBRT.
Collapse
Affiliation(s)
- Savino Cilla
- Gemelli Molise Hospital, Medical Physics Unit, Largo Gemelli 1, 86100, Campobasso, Italy.
| | - Domenico Pistilli
- Gemelli Molise Hospital, Medical Physics Unit, Largo Gemelli 1, 86100, Campobasso, Italy
| | - Carmela Romano
- Gemelli Molise Hospital, Medical Physics Unit, Largo Gemelli 1, 86100, Campobasso, Italy
| | | | - Antonio Pierro
- Radiology Unit, Gemelli Molise Hospital, Campobasso, Italy
| | - Alessandra Arcelli
- Radiation Oncology Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Milly Buwenge
- Radiation Oncology Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Alessio Giuseppe Morganti
- Radiation Oncology Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
- Department of Experimental, Diagnostic, and Specialty Medicine-DIMES, Alma Mater Studiorum, Università di Bologna, Diagnostic, Italy
| | - Francesco Deodato
- Radiation Oncology Unit, Gemelli Molise Hospital, Campobasso, Italy
- Istituto di Radiologia, Università Cattolica del Sacro Cuore, Roma, Italy
| |
Collapse
|
25
|
Li S, Su X, Ning Y, Zhang S, Shao H, Wan X, Tan Q, Yang X, Peng J, Gong Q, Yue Q. CT based intratumor and peritumoral radiomics for differentiating complete from incomplete capsular characteristics of parotid pleomorphic adenoma: a two-center study. Discov Oncol 2023; 14:76. [PMID: 37217656 DOI: 10.1007/s12672-023-00665-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Accepted: 04/20/2023] [Indexed: 05/24/2023] Open
Abstract
OBJECTIVE Capsular characteristics of pleomorphic adenoma (PA) has various forms. Patients without complete capsule has a higher risk of recurrence than patients with complete capsule. We aimed to develop and validate CT-based intratumoral and peritumoral radiomics models to make a differential diagnosis between parotid PA with and without complete capsule. METHODS Data of 260 patients (166 patients with PA from institution 1 (training set) and 94 patients (test set) from institution 2) were retrospectively analyzed. Three Volume of interest (VOIs) were defined in the CT images of each patient: tumor volume of interest (VOItumor), VOIperitumor, and VOIintra-plus peritumor. Radiomics features were extracted from each VOI and used to train nine different machine learning algorithms. Model performance was evaluated using receiver operating characteristic (ROC) curves and the area under the curve (AUC). RESULTS The results showed that the radiomics models based on features from VOIintra-plus peritumor achieved higher AUCs compared to models based on features from VOItumor. The best performing model was Linear discriminant analysis, which achieved an AUC of 0.86 in the tenfold cross-validation and 0.869 in the test set. The model was based on 15 features, including shape-based features and texture features. CONCLUSIONS We demonstrated the feasibility of combining artificial intelligence with CT-based peritumoral radiomics features can be used to accurately predict capsular characteristics of parotid PA. This may assist in clinical decision-making by preoperative identification of capsular characteristics of parotid PA.
Collapse
Affiliation(s)
- Shuang Li
- Huaxi MR Research Center (HMRRC), Department of Radiology, West China Hospital of Sichuan University, Chengdu, 610041, China
- Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, Chengdu, Sichuan, China
- Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Xiaorui Su
- Huaxi MR Research Center (HMRRC), Department of Radiology, West China Hospital of Sichuan University, Chengdu, 610041, China
| | - Youquan Ning
- Department of Radiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Simin Zhang
- Huaxi MR Research Center (HMRRC), Department of Radiology, West China Hospital of Sichuan University, Chengdu, 610041, China
| | - Hanbing Shao
- Huaxi MR Research Center (HMRRC), Department of Radiology, West China Hospital of Sichuan University, Chengdu, 610041, China
| | - Xinyue Wan
- Huaxi MR Research Center (HMRRC), Department of Radiology, West China Hospital of Sichuan University, Chengdu, 610041, China
| | - Qiaoyue Tan
- Huaxi MR Research Center (HMRRC), Department of Radiology, West China Hospital of Sichuan University, Chengdu, 610041, China
- Division of Radiation Physics, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital of Sichuan University, Chengdu, China
| | - Xibiao Yang
- Department of Radiology, West China Hospital of Sichuan University, #37 GuoXue Xiang, Chengdu, 610041, Sichuan, China
| | - Juan Peng
- Department of Radiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Qiyong Gong
- Huaxi MR Research Center (HMRRC), Department of Radiology, West China Hospital of Sichuan University, Chengdu, 610041, China.
- Department of Radiology, West China Xiamen Hospital of Sichuan University, Xiamen, Fujian, China.
| | - Qiang Yue
- Department of Radiology, West China Hospital of Sichuan University, #37 GuoXue Xiang, Chengdu, 610041, Sichuan, China.
| |
Collapse
|
26
|
Kim G, Park YM, Yoon HJ, Choi JH. A multi-kernel and multi-scale learning based deep ensemble model for predicting recurrence of non-small cell lung cancer. PeerJ Comput Sci 2023; 9:e1311. [PMID: 37346527 PMCID: PMC10280639 DOI: 10.7717/peerj-cs.1311] [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: 11/15/2022] [Accepted: 03/06/2023] [Indexed: 06/23/2023]
Abstract
Predicting recurrence in patients with non-small cell lung cancer (NSCLC) before treatment is vital for guiding personalized medicine. Deep learning techniques have revolutionized the application of cancer informatics, including lung cancer time-to-event prediction. Most existing convolutional neural network (CNN) models are based on a single two-dimensional (2D) computational tomography (CT) image or three-dimensional (3D) CT volume. However, studies have shown that using multi-scale input and fusing multiple networks provide promising performance. This study proposes a deep learning-based ensemble network for recurrence prediction using a dataset of 530 patients with NSCLC. This network assembles 2D CNN models of various input slices, scales, and convolutional kernels, using a deep learning-based feature fusion model as an ensemble strategy. The proposed framework is uniquely designed to benefit from (i) multiple 2D in-plane slices to provide more information than a single central slice, (ii) multi-scale networks and multi-kernel networks to capture the local and peritumoral features, (iii) ensemble design to integrate features from various inputs and model architectures for final prediction. The ensemble of five 2D-CNN models, three slices, and two multi-kernel networks, using 5 × 5 and 6 × 6 convolutional kernels, achieved the best performance with an accuracy of 69.62%, area under the curve (AUC) of 72.5%, F1 score of 70.12%, and recall of 70.81%. Furthermore, the proposed method achieved competitive results compared with the 2D and 3D-CNN models for cancer outcome prediction in the benchmark studies. Our model is also a potential adjuvant treatment tool for identifying NSCLC patients with a high risk of recurrence.
Collapse
Affiliation(s)
- Gihyeon Kim
- Department of Computational Medicine, Graduate Program in System Health Science and Engineering, Ewha Womans University, Seoul, South Korea
| | - Young Mi Park
- Department of Molecular Medicine, College of Medicine, Ewha Womans University, Seoul, South Korea
| | - Hyun Jung Yoon
- Department of Radiology, Veterans Health Service Medical Center, Seoul, South Korea
| | - Jang-Hwan Choi
- Division of Mechanical and Biomedical Engineering, Graduate Program in System Health Science and Engineering, Ewha Womans University, Seoul, South Korea
- Department of Artificial Intelligence, Ewha Womans University, Seoul, South Korea
| |
Collapse
|
27
|
Yan W, Quan C, Waleed M, Yuan J, Shi Z, Yang J, Lu Q, Zhang J. Application of radiomics in lung immuno‐oncology. PRECISION RADIATION ONCOLOGY 2023. [DOI: 10.1002/pro6.1191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/08/2023] Open
Affiliation(s)
- Weisi Yan
- Baptist Health System Lexington Kentucky USA
| | - Chen Quan
- City of Hope Comprehensive Cancer Center Duarte California USA
| | - Mourad Waleed
- Department of Radiation Medicine University of Kentucky Lexington Kentucky USA
| | - Jianda Yuan
- Translational Oncology at Merck & Co Kenilworth New Jersey USA
| | | | - Jun Yang
- Foshan Chancheng Hospital Foshan Guangdong China
| | - Qiuxia Lu
- Foshan Chancheng Hospital Foshan Guangdong China
| | - Jie Zhang
- Department of Radiology University of Kentucky Lexington Kentucky USA
| |
Collapse
|
28
|
Intratumoral and peritumoral radiomics nomograms for the preoperative prediction of lymphovascular invasion and overall survival in non-small cell lung cancer. Eur Radiol 2023; 33:947-958. [PMID: 36064979 DOI: 10.1007/s00330-022-09109-3] [Citation(s) in RCA: 21] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Revised: 06/03/2022] [Accepted: 07/24/2022] [Indexed: 02/03/2023]
Abstract
OBJECTIVES To evaluate the predictive value of intratumoral and peritumoral radiomics and radiomics nomogram for preoperative lymphovascular invasion (LVI) status and overall survival (OS) in patients with non-small cell lung cancer (NSCLC). METHODS In total, 240 NSCLC patients from our institution were randomly divided into the training cohort (n = 145) and internal validation cohort (n = 95) with a ratio of 6:4, and 65 patients from the Cancer Imaging Archive were enrolled as the external validation cohort. We extracted 1217 CT-based radiomics features from the gross tumor volume (GTV) and gross tumor volume incorporating peritumoral 3, 6, and 9 mm regions (GPTV3, GPTV6, GPTV9). A radiomics nomogram based on clinical independent predictors and radiomics score (Radscore) of the best radiomics model was constructed. The correlation between factors and OS was evaluated with the Kaplan-Meier survival analysis and Cox proportional hazards regression analysis. RESULTS Compared with GTV, GPTV3, and GPTV6 radiomics models, GPTV9 radiomics model exhibited better prediction performance with the AUCs of 0.82, 0.75, and 0.67 in the training, internal validation, and external validation cohorts, respectively. In the clinical model, smoking and clinical stage were independent predictors. The nomogram incorporating independent predictors and GPTV9-Radscore was clinically useful, with the AUCs of 0.89, 0.83, and 0.66 in three cohorts. Pathological LVI, GPTV9-Radscore-predicted, and Nomoscore-predicted LVI were associated with poor OS (p < 0.05). CONCLUSIONS CT-based radiomics nomogram can predict LVI and OS in patients with NSCLC and may help in making personalized treatment strategies before surgery. KEY POINTS • Compared with GTV, GPTV3, and GPTV6 radiomics models, GPTV9 radiomics model showed better prediction performance for LVI status in NSCLC. • The radiomics nomogram based on GPTV9 radiomics features and clinical independent predictors could effectively predict LVI status and OS in NSCLC and outperformed the clinical model. • The radiomics nomogram had a wider scope of clinical application.
Collapse
|
29
|
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: 10] [Impact Index Per Article: 10.0] [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
|
30
|
Ouyang G, Chen Z, Dou M, Luo X, Wen H, Deng X, Meng W, Yu Y, Wu B, Jiang D, Wang Z, Yao Y, Wang X. Predicting Rectal Cancer Response to Total Neoadjuvant Treatment Using an Artificial Intelligence Model Based on Magnetic Resonance Imaging and Clinical Data. Technol Cancer Res Treat 2023; 22:15330338231186467. [PMID: 37431270 PMCID: PMC10338728 DOI: 10.1177/15330338231186467] [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: 02/08/2023] [Revised: 05/15/2023] [Accepted: 05/24/2023] [Indexed: 07/12/2023] Open
Abstract
PURPOSE To develop a model for predicting response to total neoadjuvant treatment (TNT) for patients with locally advanced rectal cancer (LARC) based on baseline magnetic resonance imaging (MRI) and clinical data using artificial intelligence methods. METHODS Baseline MRI and clinical data were curated from patients with LARC and analyzed using logistic regression (LR) and deep learning (DL) methods to predict TNT response retrospectively. We defined two groups of response to TNT as pathological complete response (pCR) versus non-pCR (Group 1), and high sensitivity [tumor regression grade (TRG) 0 and TRG 1] versus moderate sensitivity (TRG 2 or patients with TRG 3 and a reduction in tumor volume of at least 20% compared to baseline) versus low sensitivity (TRG 3 and a reduction in tumor volume <20% compared to baseline) (Group 2). We extracted and selected clinical and radiomic features on baseline T2WI. Then we built LR models and DL models. Receiver operating characteristic (ROC) curves analysis was performed to assess predictive performance of models. RESULTS Eighty-nine patients were assigned to the training cohort, and 29 patients were assigned to the testing cohort. The area under receiver operating characteristics curve (AUC) of LR models, which were predictive of high sensitivity and pCR, were 0.853 and 0.866, respectively. Whereas the AUCs of DL models were 0.829 and 0.838, respectively. After 10 rounds of cross validation, the accuracy of the models in Group 1 is higher than in Group 2. CONCLUSION There was no significant difference between LR model and DL model. Artificial Intelligence-based radiomics biomarkers may have potential clinical implications for adaptive and personalized therapy.
Collapse
Affiliation(s)
- Ganlu Ouyang
- Department of Radiation Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
- Lung Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Zhebin Chen
- Chengdu Institute of Computer Application, Chinese Academy of Sciences, Chengdu, Sichuan, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Meng Dou
- Chengdu Institute of Computer Application, Chinese Academy of Sciences, Chengdu, Sichuan, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Xu Luo
- Chengdu Institute of Computer Application, Chinese Academy of Sciences, Chengdu, Sichuan, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Han Wen
- Chengdu Institute of Computer Application, Chinese Academy of Sciences, Chengdu, Sichuan, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Xiangbing Deng
- Department of Gastrointestinal Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Wenjian Meng
- Department of Gastrointestinal Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Yongyang Yu
- Department of Gastrointestinal Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Bing Wu
- Department of Radiology, West China Hospital, Sichuan University, Chengdu, China
| | - Dan Jiang
- Department of Pathology, West China Hospital, Sichuan University, Chengdu, China
| | - Ziqiang Wang
- Department of Gastrointestinal Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Yu Yao
- Chengdu Institute of Computer Application, Chinese Academy of Sciences, Chengdu, Sichuan, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Xin Wang
- Department of Radiation Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
- Department of Abdominal Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
31
|
Can quantitative peritumoral CT radiomics features predict the prognosis of patients with non-small cell lung cancer? A systematic review. Eur Radiol 2023; 33:2105-2117. [PMID: 36307554 PMCID: PMC9935659 DOI: 10.1007/s00330-022-09174-8] [Citation(s) in RCA: 21] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Revised: 08/20/2022] [Accepted: 09/16/2022] [Indexed: 11/04/2022]
Abstract
OBJECTIVES To provide an overarching evaluation of the value of peritumoral CT radiomics features for predicting the prognosis of non-small cell lung cancer and to assess the quality of the available studies. METHODS The PubMed, Embase, Web of Science, and Cochrane Library databases were searched for studies predicting the prognosis in patients with non-small cell lung cancer (NSCLC) using CT-based peritumoral radiomics features. Information about the patient, CT-scanner, and radiomics analyses were all extracted for the included studies. Study quality was assessed using the Radiomics Quality Score (RQS) and the Prediction Model Risk of Bias Assessment Tool (PROBAST). RESULTS Thirteen studies were included with 2942 patients from 2017 to 2022. Only one study was prospective, and the others were all retrospectively designed. Manual segmentation and multicenter studies were performed by 69% and 46% of the included studies, respectively. 3D-Slicer and MATLAB software were most commonly used for the segmentation of lesions and extraction of features. The peritumoral region was most frequently defined as dilated from the tumor boundary of 15 mm, 20 mm, or 30 mm. The median RQS of the studies was 13 (range 4-19), while all of included studies were assessed as having a high risk of bias (ROB) overall. CONCLUSIONS Peritumoral radiomics features based on CT images showed promise in predicting the prognosis of NSCLC, although well-designed studies and further biological validation are still needed. KEY POINTS • Peritumoral radiomics features based on CT images are promising and encouraging for predicting the prognosis of non-small cell lung cancer. • The peritumoral region was often dilated from the tumor boundary of 15 mm or 20 mm because these were considered safe margins. • The median Radiomics Quality Score of the included studies was 13 (range 4-19), and all of studies were considered to have a high risk of bias overall.
Collapse
|
32
|
Zhang X, Lu B, Yang X, Lan D, Lin S, Zhou Z, Li K, Deng D, Peng P, Zeng Z, Long L. Prognostic analysis and risk stratification of lung adenocarcinoma undergoing EGFR-TKI therapy with time-serial CT-based radiomics signature. Eur Radiol 2023; 33:825-835. [PMID: 36166088 PMCID: PMC9889474 DOI: 10.1007/s00330-022-09123-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 08/05/2022] [Accepted: 08/19/2022] [Indexed: 02/04/2023]
Abstract
OBJECTIVES To evaluate the value of time-serial CT radiomics features in predicting progression-free survival (PFS) for lung adenocarcinoma (LUAD) patients after epidermal growth factor receptor-tyrosine kinase inhibitors (EGFR-TKIs) therapy. MATERIALS AND METHODS LUAD patients treated with EGFR-TKIs were retrospectively included from three independent institutes and divided into training and validation cohorts. Intratumoral and peritumoral features were extracted from time-serial non-contrast chest CT (including pre-therapy and first follow-up images); moreover, the percentage variation per unit time (day) was introduced to adjust for the different follow-up periods of each patient. Test-retest was performed to exclude irreproducible features, while the Boruta algorithm was used to select critical radiomics features. Radiomics signatures were constructed with random forest survival models in the training cohort and compared against baseline clinical characteristics through Cox regression and nonparametric testing of concordance indices (C-indices). RESULTS The training cohort included 131 patients (74 women, 56.5%) from one institute and the validation cohort encompassed 41 patients (24 women, 58.5%) from two other institutes. The optimal signature contained 10 features and 7 were unit time feature variations. The comprehensive radiomics model outperformed the pre-therapy clinical characteristics in predicting PFS (training: 0.78, 95% CI: [0.72, 0.84] versus 0.55, 95% CI: [0.49, 0.62], p < 0.001; validation: 0.72, 95% CI: [0.60, 0.84] versus 0.54, 95% CI: [0.42, 0.66], p < 0.001). CONCLUSION Radiomics signature derived from time-serial CT images demonstrated optimal prognostic performance of disease progression. This dynamic imaging biomarker holds the promise of monitoring treatment response and achieving personalized management. KEY POINTS • The intrinsic tumor heterogeneity can be highly dynamic under the therapeutic effect of EGFR-TKI treatment, and the inevitable development of drug resistance may disrupt the duration of clinical benefit. Decision-making remained challenging in practice to detect the emergence of acquired resistance during the early response phase. • Time-serial CT-based radiomics signature integrating intra- and peritumoral features offered the potential to predict progression-free survival for LUAD patients treated with EGFR-TKIs. • The dynamic imaging signature allowed for prognostic risk stratification.
Collapse
Affiliation(s)
- Xiaobo Zhang
- Department of Radiology, The First Affiliated Hospital of Guangxi Medical University, No.6 Shuangyong Road, Qingxiu District, Nanning, 530021 Guangxi China
| | - Bingfeng Lu
- Department of Radiology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi China
| | - Xinguan Yang
- Department of Radiology, Guilin People’s Hospital, Guilin, Guangxi China
| | - Dong Lan
- Department of Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi China
| | | | - Zhipeng Zhou
- Department of Radiology, Affiliated Hospital of Guilin Medical University, Guilin, Guangxi China
| | - Kai Li
- Department of Radiology, The First Affiliated Hospital of Guangxi Medical University, No.6 Shuangyong Road, Qingxiu District, Nanning, 530021 Guangxi China
| | - Dong Deng
- Department of Radiology, The First Affiliated Hospital of Guangxi Medical University, No.6 Shuangyong Road, Qingxiu District, Nanning, 530021 Guangxi China
| | - Peng Peng
- Department of Radiology, The First Affiliated Hospital of Guangxi Medical University, No.6 Shuangyong Road, Qingxiu District, Nanning, 530021 Guangxi China
| | - Zisan Zeng
- Department of Radiology, The First Affiliated Hospital of Guangxi Medical University, No.6 Shuangyong Road, Qingxiu District, Nanning, 530021 Guangxi China
| | - Liling Long
- Department of Radiology, The First Affiliated Hospital of Guangxi Medical University, No.6 Shuangyong Road, Qingxiu District, Nanning, 530021 Guangxi China
| |
Collapse
|
33
|
Amyar A, Modzelewski R, Vera P, Morard V, Ruan S. Multi-task multi-scale learning for outcome prediction in 3D PET images. Comput Biol Med 2022; 151:106208. [PMID: 36306580 DOI: 10.1016/j.compbiomed.2022.106208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 09/18/2022] [Accepted: 10/09/2022] [Indexed: 12/27/2022]
Abstract
BACKGROUND AND OBJECTIVES Predicting patient response to treatment and survival in oncology is a prominent way towards precision medicine. To this end, radiomics has been proposed as a field of study where images are used instead of invasive methods. The first step in radiomic analysis in oncology is lesion segmentation. However, this task is time consuming and can be physician subjective. Automated tools based on supervised deep learning have made great progress in helping physicians. However, they are data hungry, and annotated data remains a major issue in the medical field where only a small subset of annotated images are available. METHODS In this work, we propose a multi-task, multi-scale learning framework to predict patient's survival and response. We show that the encoder can leverage multiple tasks to extract meaningful and powerful features that improve radiomic performance. We also show that subsidiary tasks serve as an inductive bias so that the model can better generalize. RESULTS Our model was tested and validated for treatment response and survival in esophageal and lung cancers, with an area under the ROC curve of 77% and 71% respectively, outperforming single-task learning methods. CONCLUSIONS Multi-task multi-scale learning enables higher performance of radiomic analysis by extracting rich information from intratumoral and peritumoral regions.
Collapse
Affiliation(s)
- Amine Amyar
- General Electric Healthcare, Buc, France; LITIS - EA4108 - Quantif, University of Rouen, Rouen, France.
| | - Romain Modzelewski
- LITIS - EA4108 - Quantif, University of Rouen, Rouen, France; Nuclear Medicine Department, Henri Becquerel Center, Rouen, France
| | - Pierre Vera
- LITIS - EA4108 - Quantif, University of Rouen, Rouen, France; Nuclear Medicine Department, Henri Becquerel Center, Rouen, France
| | | | - Su Ruan
- LITIS - EA4108 - Quantif, University of Rouen, Rouen, France
| |
Collapse
|
34
|
Soliman MA, Kelahan LC, Magnetta M, Savas H, Agrawal R, Avery RJ, Aouad P, Liu B, Xue Y, Chae YK, Salem R, Benson AB, Yaghmai V, Velichko YS. A Framework for Harmonization of Radiomics Data for Multicenter Studies and Clinical Trials. JCO Clin Cancer Inform 2022; 6:e2200023. [PMID: 36332157 PMCID: PMC9668564 DOI: 10.1200/cci.22.00023] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Revised: 07/01/2022] [Accepted: 09/21/2022] [Indexed: 11/06/2022] Open
Abstract
PURPOSE Variability in computed tomography images intrinsic to individual scanners limits the application of radiomics in clinical and research settings. The development of reproducible and generalizable radiomics-based models to assess lesions requires harmonization of data. The purpose of this study was to develop, test, and analyze the efficacy of a radiomics data harmonization model. MATERIALS AND METHODS Radiomic features from biopsy-proven untreated hepatic metastasis (N = 380) acquired from 167 unique patients with pancreatic, colon, and breast cancers were analyzed. Radiomic features from volume-match 551 samples of normal liver tissue and 188 hepatic cysts were included as references. A novel linear mixed effect model was used to identify effects associated with lesion size, tissue type, and scanner model. Six separate machine learning models were then used to test the effectiveness of radiomic feature harmonization using multivariate analysis. RESULTS Proposed model identifies and removes scanner-associated effects while preserving cancer-specific functional dependence of radiomic features on the tumor size. Data harmonization improves the performance of classification models by reducing the scanner-associated variability. For example, the multiclass logistic regression model, LogitBoost, demonstrated the improvement in sensitivity in the range from 15% to 40% for each type of liver metastasis, whereas the overall model accuracy and the kappa coefficient increased by 5% and 8% accordingly. CONCLUSION The model removed scanner-associated effects while preserving cancer-specific functional dependence of radiomic features.
Collapse
Affiliation(s)
- Moataz A.S. Soliman
- Department of Radiology, Feinberg School of Medicine, Northwestern University, Chicago, IL
| | - Linda C. Kelahan
- Department of Radiology, Feinberg School of Medicine, Northwestern University, Chicago, IL
| | - Michael Magnetta
- Department of Radiology, Feinberg School of Medicine, Northwestern University, Chicago, IL
| | - Hatice Savas
- Department of Radiology, Feinberg School of Medicine, Northwestern University, Chicago, IL
| | - Rishi Agrawal
- Department of Radiology, Feinberg School of Medicine, Northwestern University, Chicago, IL
| | - Ryan J. Avery
- Department of Radiology, Feinberg School of Medicine, Northwestern University, Chicago, IL
| | - Pascale Aouad
- Department of Radiology, Feinberg School of Medicine, Northwestern University, Chicago, IL
| | - Benjamin Liu
- Department of Radiology, Feinberg School of Medicine, Northwestern University, Chicago, IL
| | - Yue Xue
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, IL
| | - Young K. Chae
- Department of Medicine, Division of Hematology/Oncology, Feinberg School of Medicine, Northwestern University, Chicago, IL
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL
| | - Riad Salem
- Department of Radiology, Feinberg School of Medicine, Northwestern University, Chicago, IL
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL
| | - Al B. Benson
- Department of Medicine, Division of Hematology/Oncology, Feinberg School of Medicine, Northwestern University, Chicago, IL
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL
| | - Vahid Yaghmai
- Department of Radiological Sciences, University of California, Irvine UCI Health, University of California Irvine, Orange, CA
| | - Yuri S. Velichko
- Department of Radiology, Feinberg School of Medicine, Northwestern University, Chicago, IL
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL
| |
Collapse
|
35
|
Gross tumour volume radiomics for prognostication of recurrence & death following radical radiotherapy for NSCLC. NPJ Precis Oncol 2022; 6:77. [PMID: 36302938 PMCID: PMC9613990 DOI: 10.1038/s41698-022-00322-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Accepted: 10/14/2022] [Indexed: 11/26/2022] Open
Abstract
Recurrence occurs in up to 36% of patients treated with curative-intent radiotherapy for NSCLC. Identifying patients at higher risk of recurrence for more intensive surveillance may facilitate the earlier introduction of the next line of treatment. We aimed to use radiotherapy planning CT scans to develop radiomic classification models that predict overall survival (OS), recurrence-free survival (RFS) and recurrence two years post-treatment for risk-stratification. A retrospective multi-centre study of >900 patients receiving curative-intent radiotherapy for stage I-III NSCLC was undertaken. Models using radiomic and/or clinical features were developed, compared with 10-fold cross-validation and an external test set, and benchmarked against TNM-stage. Respective validation and test set AUCs (with 95% confidence intervals) for the radiomic-only models were: (1) OS: 0.712 (0.592–0.832) and 0.685 (0.585–0.784), (2) RFS: 0.825 (0.733–0.916) and 0.750 (0.665–0.835), (3) Recurrence: 0.678 (0.554–0.801) and 0.673 (0.577–0.77). For the combined models: (1) OS: 0.702 (0.583–0.822) and 0.683 (0.586–0.78), (2) RFS: 0.805 (0.707–0.903) and 0·755 (0.672–0.838), (3) Recurrence: 0·637 (0.51–0.·765) and 0·738 (0.649–0.826). Kaplan-Meier analyses demonstrate OS and RFS difference of >300 and >400 days respectively between low and high-risk groups. We have developed validated and externally tested radiomic-based prediction models. Such models could be integrated into the routine radiotherapy workflow, thus informing a personalised surveillance strategy at the point of treatment. Our work lays the foundations for future prospective clinical trials for quantitative personalised risk-stratification for surveillance following curative-intent radiotherapy for NSCLC.
Collapse
|
36
|
Braman N, Prasanna P, Bera K, Alilou M, Khorrami M, Leo P, Etesami M, Vulchi M, Turk P, Gupta A, Jain P, Fu P, Pennell N, Velcheti V, Abraham J, Plecha D, Madabhushi A. Novel Radiomic Measurements of Tumor-Associated Vasculature Morphology on Clinical Imaging as a Biomarker of Treatment Response in Multiple Cancers. Clin Cancer Res 2022; 28:4410-4424. [PMID: 35727603 PMCID: PMC9588630 DOI: 10.1158/1078-0432.ccr-21-4148] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2021] [Revised: 03/14/2022] [Accepted: 06/17/2022] [Indexed: 12/14/2022]
Abstract
PURPOSE The tumor-associated vasculature (TAV) differs from healthy blood vessels by its convolutedness, leakiness, and chaotic architecture, and these attributes facilitate the creation of a treatment-resistant tumor microenvironment. Measurable differences in these attributes might also help stratify patients by likely benefit of systemic therapy (e.g., chemotherapy). In this work, we present a new category of computational image-based biomarkers called quantitative tumor-associated vasculature (QuanTAV) features, and demonstrate their ability to predict response and survival across multiple cancer types, imaging modalities, and treatment regimens involving chemotherapy. EXPERIMENTAL DESIGN We isolated tumor vasculature and extracted mathematical measurements of twistedness and organization from routine pretreatment radiology (CT or contrast-enhanced MRI) of a total of 558 patients, who received one of four first-line chemotherapy-based therapeutic intervention strategies for breast (n = 371) or non-small cell lung cancer (NSCLC, n = 187). RESULTS Across four chemotherapy-based treatment strategies, classifiers of QuanTAV measurements significantly (P < 0.05) predicted response in held out testing cohorts alone (AUC = 0.63-0.71) and increased AUC by 0.06-0.12 when added to models of significant clinical variables alone. Similarly, we derived QuanTAV risk scores that were prognostic of recurrence-free survival in treatment cohorts who received surgery following chemotherapy for breast cancer [P = 0.0022; HR = 1.25; 95% confidence interval (CI), 1.08-1.44; concordance index (C-index) = 0.66] and chemoradiation for NSCLC (P = 0.039; HR = 1.28; 95% CI, 1.01-1.62; C-index = 0.66). From vessel-based risk scores, we further derived categorical QuanTAV high/low risk groups that were independently prognostic among all treatment groups, including patients with NSCLC who received chemotherapy only (P = 0.034; HR = 2.29; 95% CI, 1.07-4.94; C-index = 0.62). QuanTAV response and risk scores were independent of clinicopathologic risk factors and matched or exceeded models of clinical variables including posttreatment response. CONCLUSIONS Across these domains, we observed an association of vascular morphology on CT and MRI-as captured by metrics of vessel curvature, torsion, and organizational heterogeneity-and treatment outcome. Our findings suggest the potential of shape and structure of the TAV in developing prognostic and predictive biomarkers for multiple cancers and different treatment strategies.
Collapse
Affiliation(s)
- Nathaniel Braman
- Case Western Reserve University, Cleveland, OH
- Picture Health, Cleveland, OH
| | - Prateek Prasanna
- Case Western Reserve University, Cleveland, OH
- Stony Brook University, New York, NY
| | - Kaustav Bera
- Case Western Reserve University, Cleveland, OH
- University Hospitals Cleveland Medical Center, Cleveland, OH
| | | | | | - Patrick Leo
- Case Western Reserve University, Cleveland, OH
| | - Maryam Etesami
- Yale School of Medicine, Department of Radiology & Biomedical Imaging, New Haven, CT
| | - Manasa Vulchi
- The Cleveland Clinic Foundation (CCF), Cleveland, OH
| | - Paulette Turk
- The Cleveland Clinic Foundation (CCF), Cleveland, OH
| | - Amit Gupta
- University Hospitals Cleveland Medical Center, Cleveland, OH
| | - Prantesh Jain
- University Hospitals Cleveland Medical Center, Cleveland, OH
| | - Pingfu Fu
- Case Western Reserve University, Cleveland, OH
| | | | | | - Jame Abraham
- The Cleveland Clinic Foundation (CCF), Cleveland, OH
| | - Donna Plecha
- University Hospitals Cleveland Medical Center, Cleveland, OH
| | - Anant Madabhushi
- Case Western Reserve University, Cleveland, OH
- Louis Stokes Cleveland Veterans Medical Center, Cleveland, OH
| |
Collapse
|
37
|
Wang S, Lin C, Kolomaya A, Ostdiek-Wille GP, Wong J, Cheng X, Lei Y, Liu C. Compute Tomography Radiomics Analysis on Whole Pancreas Between Healthy Individual and Pancreatic Ductal Adenocarcinoma Patients: Uncertainty Analysis and Predictive Modeling. Technol Cancer Res Treat 2022; 21:15330338221126869. [PMID: 36184987 PMCID: PMC9530578 DOI: 10.1177/15330338221126869] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
Abstract
Radiomics is a rapidly growing field that quantitatively extracts image features
in a high-throughput manner from medical imaging. In this study, we analyzed the
radiomics features of the whole pancreas between healthy individuals and
pancreatic cancer patients, and we established a predictive model that can
distinguish cancer patients from healthy individuals based on these radiomics
features. Methods: We retrospectively collected venous-phase scans
of contrast-enhanced computed tomography (CT) images from 181 control subjects
and 85 cancer case subjects for radiomics analysis and predictive modeling. An
attending radiation oncologist delineated the pancreas for all the subjects in
the Varian Eclipse system, and we extracted 924 radiomics features using
PyRadiomics. We established a feature selection pipeline to exclude redundant or
unstable features. We randomly selected 189 cases (60 cancer and 129 control) as
the training set. The remaining 77 subjects (25 cancer and 52 control) as a test
set. We trained a Random Forest model utilizing the stable features to
distinguish the cancer patients from the healthy individuals on the training
dataset. We analyzed the performance of our best model by running 5-fold
cross-validations on the training dataset and applied our best model to the test
set. Results: We identified that 91 radiomics features are stable
against various uncertainty sources, including bin width, resampling, image
transformation, image noise, and segmentation uncertainty. Eight of the 91
features are nonredundant. Our final predictive model, using these 8 features,
has achieved a mean area under the receiver operating characteristic curve (AUC)
of 0.99 ± 0.01 on the training dataset (189 subjects) by cross-validation. The
model achieved an AUC of 0.910 on the independent test set (77 subjects) and an
accuracy of 0.935. Conclusion: CT-based radiomics analysis based on
the whole pancreas can distinguish cancer patients from healthy individuals, and
it could potentially become an early detection tool for pancreatic cancer.
Collapse
Affiliation(s)
- Shuo Wang
- Department of Radiation Oncology, University of Nebraska Medical
Center, Omaha, NE, USA,Shuo Wang, PhD, Department of Radiation
Oncology, University of Nebraska Medical Center, 986861 Nebraska Medical Center,
Omaha, NE 68198, USA. Chi Lin, MD, PhD,
Department of Radiation Oncology, University of Nebraska Medical Center, 986861
Nebraska Medical Center, Omaha, NE 68198, USA
| | - Chi Lin
- Department of Radiation Oncology, University of Nebraska Medical
Center, Omaha, NE, USA
| | - Alexander Kolomaya
- College of Medicine, University of Nebraska Medical
Center, Omaha, NE, USA
| | | | - Jeffrey Wong
- Department of Radiation Oncology, University of Nebraska Medical
Center, Omaha, NE, USA
| | - Xiaoyue Cheng
- Department of Mathematics, University of Nebraska
Omaha, Omaha, NE, USA
| | - Yu Lei
- Department of Radiation Oncology, Barrow Neurological
Institute, Phoenix, AZ, USA
| | | |
Collapse
|
38
|
Yamazaki M, Yagi T, Tominaga M, Minato K, Ishikawa H. Role of intratumoral and peritumoral CT radiomics for the prediction of EGFR gene mutation in primary lung cancer. Br J Radiol 2022; 95:20220374. [DOI: 10.1259/bjr.20220374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
Objectives To determine the added value of combining intratumoral and peritumoral CT radiomics for the prediction of epidermal growth factor receptor (EGFR) gene mutations in primary lung cancer (PLC). Methods This study included 478 patients with PLC (348 adenocarcinomas and 130 other histological types) who underwent surgical resection and EGFR gene testing. Two radiologists performed segmentation of tumors and peritumoral regions using precontrast high-resolution CT images, and 398 radiomic features (212 intra- and 186 peritumoral features) were extracted. The peritumoral region was defined as the lung parenchyma within a distance of 3 mm from the tumor border. Model performance was estimated using Random Forest, a machine-learning algorithm. Results EGFR mutations were found in 162 tumors; 161 adenocarcinomas, and one pleomorphic carcinoma. After exclusion of poorly reproducible and redundant features, 32 radiomic features remained (14 intra- and 18 peritumoral features) and were included in the model building. For predicting EGFR mutations, combining intra- and peritumoral radiomics significantly improved the performance compared to intratumoral radiomics alone (AUC [area under the receiver operating characteristic curve], 0.774 vs 0.730; p < 0.001). Even in adenocarcinomas only, adding peritumoral radiomics significantly increased performance (AUC, 0.687 vs 0.630; p < 0.001). The predictive performance using radiomics and clinical features was significantly higher than that of clinical features alone (AUC, 0.826 vs 0.777; p = 0.005). Conclusions Combining intra- and peritumoral radiomics improves the predictive accuracy of EGFR mutations and could be used to aid in decision-making of whether to perform biopsy for gene tests. Advances in knowledge Adding peritumoral to intratumoral radiomics yields greater accuracy than intratumoral radiomics alone in predicting EGFR mutations and may serve as a non-invasive method of predicting of the gene status in PLC.
Collapse
Affiliation(s)
- Motohiko Yamazaki
- Department of Radiology and Radiation Oncology, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Takuya Yagi
- Department of Radiology and Radiation Oncology, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Masaki Tominaga
- Department of Radiology and Radiation Oncology, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Kojiro Minato
- Department of Radiology and Radiation Oncology, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Hiroyuki Ishikawa
- Department of Radiology and Radiation Oncology, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| |
Collapse
|
39
|
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: 0] [Impact Index Per Article: 0] [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
|
40
|
Chang R, Qi S, Zuo Y, Yue Y, Zhang X, Guan Y, Qian W. Predicting chemotherapy response in non-small-cell lung cancer via computed tomography radiomic features: Peritumoral, intratumoral, or combined? Front Oncol 2022; 12:915835. [PMID: 36003781 PMCID: PMC9393703 DOI: 10.3389/fonc.2022.915835] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Accepted: 07/18/2022] [Indexed: 11/15/2022] Open
Abstract
Purpose This study aims to evaluate the ability of peritumoral, intratumoral, or combined computed tomography (CT) radiomic features to predict chemotherapy response in non-small cell lung cancer (NSCLC). Methods After excluding subjects with incomplete data or other types of treatments, 272 (Dataset 1) and 43 (Dataset 2, external validation) NSCLC patients who were only treated with chemotherapy as the first-line treatment were enrolled between 2015 and 2019. All patients were divided into response and nonresponse based on the response evaluation criteria in solid tumors, version 1.1. By using 3D slicer and morphological operations in python, the intra- and peritumoral regions of lung tumors were segmented from pre-treatment CT images (unenhanced) and confirmed by two experienced radiologists. Then radiomic features (the first order, texture, shape, et al.) were extracted from the above regions of interest. The models were trained and tested in Dataset 1 and further validated in Dataset 2. The performance of models was compared using the area under curve (AUC), confusion matrix, accuracy, precision, recall, and F1-score. Results The radiomic model using features from the peritumoral region of 0–3 mm outperformed that using features from 3–6, 6–9, 9–12 mm peritumoral region, and intratumoral region (AUC: 0.95 versus 0.87, 0.86, 0.85, and 0.88). By the fusion of features from 0–3 and 3–6 mm peritumoral regions, the logistic regression model achieved the best performance, with an AUC of 0.97. This model achieved an AUC of 0.85 in the external cohort. Moreover, among the 20 selected features, seven features differed significantly between the two groups (p < 0.05). Conclusions CT radiomic features from both the peri- and intratumoral regions can predict chemotherapy response in NSCLC using machine learning models. Combined features from two peritumoral regions yielded better predictions.
Collapse
Affiliation(s)
- Runsheng Chang
- College of Medicine and Biological Information Engineering, Northeastern University, Shenyang, China
| | - Shouliang Qi
- College of Medicine and Biological Information Engineering, Northeastern University, Shenyang, China
- Key Laboratory of Intelligent Computing in Medical Image, Ministry of Education, Northeastern University, Shenyang, China
- *Correspondence: Shouliang Qi,
| | - Yifan Zuo
- College of Medicine and Biological Information Engineering, Northeastern University, Shenyang, China
| | - Yong Yue
- Department of Radiology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Xiaoye Zhang
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Yubao Guan
- Department of Radiology, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Wei Qian
- College of Medicine and Biological Information Engineering, Northeastern University, Shenyang, China
| |
Collapse
|
41
|
Radiomic Signatures Associated with CD8+ Tumour-Infiltrating Lymphocytes: A Systematic Review and Quality Assessment Study. Cancers (Basel) 2022; 14:cancers14153656. [PMID: 35954318 PMCID: PMC9367613 DOI: 10.3390/cancers14153656] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Revised: 07/23/2022] [Accepted: 07/25/2022] [Indexed: 02/04/2023] Open
Abstract
The tumour immune microenvironment influences the efficacy of immune checkpoint inhibitors. Within this microenvironment are CD8-expressing tumour-infiltrating lymphocytes (CD8+ TILs), which are an important mediator and marker of anti-tumour response. In practice, the assessment of CD8+ TILs via tissue sampling involves logistical challenges. Radiomics, the high-throughput extraction of features from medical images, may offer a novel and non-invasive alternative. We performed a systematic review of the available literature reporting radiomic signatures associated with CD8+ TILs. We also aimed to evaluate the methodological quality of the identified studies using the Radiomics Quality Score (RQS) tool, and the risk of bias and applicability with the Quality Assessment of Diagnostic Accuracy Studies (QUADAS-2) tool. Articles were searched from inception until 31 December 2021, in three electronic databases, and screened against eligibility criteria. Twenty-seven articles were included. A wide variety of cancers have been studied. The reported radiomic signatures were heterogeneous, with very limited reproducibility between studies of the same cancer group. The overall quality of studies was found to be less than desirable (mean RQS = 33.3%), indicating a need for technical maturation. Some potential avenues for further investigation are also discussed.
Collapse
|
42
|
Kothari G, Woon B, Patrick CJ, Korte J, Wee L, Hanna GG, Kron T, Hardcastle N, Siva S. The impact of inter-observer variation in delineation on robustness of radiomics features in non-small cell lung cancer. Sci Rep 2022; 12:12822. [PMID: 35896707 PMCID: PMC9329346 DOI: 10.1038/s41598-022-16520-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Accepted: 07/11/2022] [Indexed: 11/25/2022] Open
Abstract
Artificial intelligence and radiomics have the potential to revolutionise cancer prognostication and personalised treatment. Manual outlining of the tumour volume for extraction of radiomics features (RF) is a subjective process. This study investigates robustness of RF to inter-observer variation (IOV) in contouring in lung cancer. We utilised two public imaging datasets: ‘NSCLC-Radiomics’ and ‘NSCLC-Radiomics-Interobserver1’ (‘Interobserver’). For ‘NSCLC-Radiomics’, we created an additional set of manual contours for 92 patients, and for ‘Interobserver’, there were five manual and five semi-automated contours available for 20 patients. Dice coefficients (DC) were calculated for contours. 1113 RF were extracted including shape, first order and texture features. Intraclass correlation coefficient (ICC) was computed to assess robustness of RF to IOV. Cox regression analysis for overall survival (OS) was performed with a previously published radiomics signature. The median DC ranged from 0.81 (‘NSCLC-Radiomics’) to 0.85 (‘Interobserver’—semi-automated). The median ICC for the ‘NSCLC-Radiomics’, ‘Interobserver’ (manual) and ‘Interobserver’ (semi-automated) were 0.90, 0.88 and 0.93 respectively. The ICC varied by feature type and was lower for first order and gray level co-occurrence matrix (GLCM) features. Shape features had a lower median ICC in the ‘NSCLC-Radiomics’ dataset compared to the ‘Interobserver’ dataset. Survival analysis showed similar separation of curves for three of four RF apart from ‘original_shape_Compactness2’, a feature with low ICC (0.61). The majority of RF are robust to IOV, with first order, GLCM and shape features being the least robust. Semi-automated contouring improves feature stability. Decreased robustness of a feature is significant as it may impact upon the features’ prognostic capability.
Collapse
Affiliation(s)
- Gargi Kothari
- Department of Radiation Oncology, Peter MacCallum Cancer Centre, Victorian Comprehensive Cancer Centre Building, 305 Grattan Street, Melbourne, VIC, 3000, Australia. .,Sir Peter MacCallum Department of Oncology, Peter MacCallum Cancer Centre, University of Melbourne, Melbourne, VIC, Australia.
| | - Beverley Woon
- Sir Peter MacCallum Department of Oncology, Peter MacCallum Cancer Centre, University of Melbourne, Melbourne, VIC, Australia.,Department of Radiology, Cancer Imaging, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - Cameron J Patrick
- Statistical Consulting Centre, University of Melbourne, Parkville, Australia
| | - James Korte
- Department of Physical Sciences, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia.,Department of Biomedical Engineering, School of Chemical and Biomedical Engineering, University of Melbourne, Melbourne, VIC, Australia
| | - Leonard Wee
- Department of Radiotherapy (MAASTRO), GROW School of Oncology, Maastricht University Medical Centre+, Maastricht, The Netherlands.,Clinical Data Science, Maastricht University, Maastricht, The Netherlands
| | - Gerard G Hanna
- Department of Radiation Oncology, Peter MacCallum Cancer Centre, Victorian Comprehensive Cancer Centre Building, 305 Grattan Street, Melbourne, VIC, 3000, Australia.,Sir Peter MacCallum Department of Oncology, Peter MacCallum Cancer Centre, University of Melbourne, Melbourne, VIC, Australia
| | - Tomas Kron
- Sir Peter MacCallum Department of Oncology, Peter MacCallum Cancer Centre, University of Melbourne, Melbourne, VIC, Australia.,Department of Physical Sciences, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia.,Centre for Medical Radiation Physics, University of Wollongong, Wollongong, NSW, Australia
| | - Nicholas Hardcastle
- Sir Peter MacCallum Department of Oncology, Peter MacCallum Cancer Centre, University of Melbourne, Melbourne, VIC, Australia.,Department of Physical Sciences, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia.,Centre for Medical Radiation Physics, University of Wollongong, Wollongong, NSW, Australia
| | - Shankar Siva
- Department of Radiation Oncology, Peter MacCallum Cancer Centre, Victorian Comprehensive Cancer Centre Building, 305 Grattan Street, Melbourne, VIC, 3000, Australia.,Sir Peter MacCallum Department of Oncology, Peter MacCallum Cancer Centre, University of Melbourne, Melbourne, VIC, Australia
| |
Collapse
|
43
|
Radiomics for Detection of the EGFR Mutation in Liver Metastatic NSCLC. Acad Radiol 2022; 30:1039-1046. [PMID: 35907759 DOI: 10.1016/j.acra.2022.06.016] [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: 05/11/2022] [Revised: 06/20/2022] [Accepted: 06/23/2022] [Indexed: 12/09/2022]
Abstract
RATIONALE AND OBJECTIVES The research aims to investigate whether MRI radiomics on hepatic metastasis from primary nonsmall cell lung cancer (NSCLC) can be used to differentiate patients with epidermal growth factor receptor (EGFR) mutations from those with EGFR wild-type, and develop a prediction model based on combination of primary tumor and the metastasis. MATERIALS AND METHODS A total of 130 patients were enrolled between Aug. 2017 and Dec. 2021, all pathologically confirmed harboring hepatic metastasis from primary NSCLC. The pyradiomics was used to extract radiomics features from intra- and peritumoral areas of both primary tumor and metastasis. The least absolute shrinkage and selection operator (LASSO) regression was applied to identify most predictive features and to develop radiomics signatures (RSs) for prediction of the EGFR mutation status. The receiver operating characteristic (ROC) curve analysis was performed to assess the prediction capability of the developed RSs. RESULTS A RS-Primary and a RS-Metastasis were derived from the primary tumor and metastasis, respectively. The RS-Combine by combination of the primary tumor and metastasis achieved the highest prediction performance in the training (AUCs, RS-Primary vs. RS-Metastasis vs. RS-Combine, 0.826 vs. 0.821 vs. 0.908) and testing (AUCs, RS-Primary vs. RS-Metastasis vs. RS-Combine, 0.760 vs. 0.791 vs. 0.884) set. The smoking status showed significant difference between EGFR mutant and wild-type groups (p < 0.05) in the training set. CONCLUSION The study indicates that hepatic metastasis-based radiomics can be used to detect the EGFR mutation. The developed multiorgan combined radiomics signature may be helpful to guide individual treatment strategies for patients with metastatic NSCLC.
Collapse
|
44
|
Radiogenomic analysis reveals tumor heterogeneity of triple-negative breast cancer. Cell Rep Med 2022; 3:100694. [PMID: 35858585 PMCID: PMC9381418 DOI: 10.1016/j.xcrm.2022.100694] [Citation(s) in RCA: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Revised: 06/07/2022] [Accepted: 06/23/2022] [Indexed: 11/04/2022]
Abstract
Triple-negative breast cancer (TNBC) is a subset of breast cancer with an adverse prognosis and significant tumor heterogeneity. Here, we extract quantitative radiomic features from contrast-enhanced magnetic resonance images to construct a breast cancer radiomic dataset (n = 860) and a TNBC radiogenomic dataset (n = 202). We develop and validate radiomic signatures that can fairly differentiate TNBC from other breast cancer subtypes and distinguish molecular subtypes within TNBC. A radiomic feature that captures peritumoral heterogeneity is determined to be a prognostic factor for recurrence-free survival (p = 0.01) and overall survival (p = 0.004) in TNBC. Combined with the established matching TNBC transcriptomic and metabolomic data, we demonstrate that peritumoral heterogeneity is associated with immune suppression and upregulated fatty acid synthesis in tumor samples. Collectively, this multi-omic dataset serves as a useful public resource to promote precise subtyping of TNBC and helps to understand the biological significance of radiomics. A radiomic signature identifies TNBC from other subtypes of breast cancer Radiomic features are predictive of TNBC molecular subtypes A radiomic feature reflecting peritumoral heterogeneity indicates TNBC prognosis Peritumoral heterogeneity correlates with metabolic and immune abnormalities in TNBC
Collapse
|
45
|
Liang HY, Yang SF, Zou HM, Hou F, Duan LS, Huang CC, Xu JX, Liu SL, Hao DP, Wang HX. Deep Learning Radiomics Nomogram to Predict Lung Metastasis in Soft-Tissue Sarcoma: A Multi-Center Study. Front Oncol 2022; 12:897676. [PMID: 35814362 PMCID: PMC9265249 DOI: 10.3389/fonc.2022.897676] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Accepted: 05/18/2022] [Indexed: 11/13/2022] Open
Abstract
Objectives To build and evaluate a deep learning radiomics nomogram (DLRN) for preoperative prediction of lung metastasis (LM) status in patients with soft tissue sarcoma (STS). Methods In total, 242 patients with STS (training set, n=116; external validation set, n=126) who underwent magnetic resonance imaging were retrospectively enrolled in this study. We identified independent predictors for LM-status and evaluated their performance. The minimum redundancy maximum relevance (mRMR) method and least absolute shrinkage and selection operator (LASSO) algorithm were adopted to screen radiomics features. Logistic regression, decision tree, random forest, support vector machine (SVM), and adaptive boosting classifiers were compared for their ability to predict LM. To overcome the imbalanced distribution of the LM data, we retrained each machine-learning classifier using the synthetic minority over-sampling technique (SMOTE). A DLRN combining the independent clinical predictors with the best performing radiomics prediction signature (mRMR+LASSO+SVM+SMOTE) was established. Area under the receiver operating characteristics curve (AUC), calibration curves, and decision curve analysis (DCA) were used to assess the performance and clinical applicability of the models. Result Comparisons of the AUC values applied to the external validation set revealed that the DLRN model (AUC=0.833) showed better prediction performance than the clinical model (AUC=0.664) and radiomics model (AUC=0.799). The calibration curves indicated good calibration efficiency and the DCA showed the DLRN model to have greater clinical applicability than the other two models. Conclusion The DLRN was shown to be an accurate and efficient tool for LM-status prediction in STS.
Collapse
Affiliation(s)
- Hao-yu Liang
- Department of Radiology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Shi-feng Yang
- Department of Radiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Hong-mei Zou
- Department of Radiology, The Third People’s Hospital of Qingdao, Qingdao, China
| | - Feng Hou
- Department of Pathology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Li-sha Duan
- Department of Radiology, The Third Hospital of Hebei Medical University, Shijiazhuang, China
| | - Chen-cui Huang
- Department of Research Collaboration, Research and Development (R&D) Center, Beijing Deepwise & League of Philosophy Doctor (PHD) Technology Co., Ltd, Beijing, China
| | - Jing-xu Xu
- Department of Research Collaboration, Research and Development (R&D) Center, Beijing Deepwise & League of Philosophy Doctor (PHD) Technology Co., Ltd, Beijing, China
| | - Shun-li Liu
- Department of Radiology, The Affiliated Hospital of Qingdao University, Qingdao, China
- *Correspondence: Shun-li Liu, ; Da-peng Hao, ; He-xiang Wang,
| | - Da-peng Hao
- Department of Radiology, The Affiliated Hospital of Qingdao University, Qingdao, China
- *Correspondence: Shun-li Liu, ; Da-peng Hao, ; He-xiang Wang,
| | - He-xiang Wang
- Department of Radiology, The Affiliated Hospital of Qingdao University, Qingdao, China
- *Correspondence: Shun-li Liu, ; Da-peng Hao, ; He-xiang Wang,
| |
Collapse
|
46
|
Abdollahi H, Chin E, Clark H, Hyde DE, Thomas S, Wu J, Uribe CF, Rahmim A. Radiomics-guided radiation therapy: opportunities and challenges. Phys Med Biol 2022; 67. [DOI: 10.1088/1361-6560/ac6fab] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Accepted: 05/13/2022] [Indexed: 11/11/2022]
Abstract
Abstract
Radiomics is an advanced image-processing framework, which extracts image features and considers them as biomarkers towards personalized medicine. Applications include disease detection, diagnosis, prognosis, and therapy response assessment/prediction. As radiation therapy aims for further individualized treatments, radiomics could play a critical role in various steps before, during and after treatment. Elucidation of the concept of radiomics-guided radiation therapy (RGRT) is the aim of this review, attempting to highlight opportunities and challenges underlying the use of radiomics to guide clinicians and physicists towards more effective radiation treatments. This work identifies the value of RGRT in various steps of radiotherapy from patient selection to follow-up, and subsequently provides recommendations to improve future radiotherapy using quantitative imaging features.
Collapse
|
47
|
Takehana K, Sakamoto R, Fujimoto K, Matsuo Y, Nakajima N, Yoshizawa A, Menju T, Nakamura M, Yamada R, Mizowaki T, Nakamoto Y. Peritumoral radiomics features on preoperative thin-slice CT images can predict the spread through air spaces of lung adenocarcinoma. Sci Rep 2022; 12:10323. [PMID: 35725754 PMCID: PMC9209514 DOI: 10.1038/s41598-022-14400-w] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Accepted: 06/06/2022] [Indexed: 11/16/2022] Open
Abstract
The spread through air spaces (STAS) is recognized as a negative prognostic factor in patients with early-stage lung adenocarcinoma. The present study aimed to develop a machine learning model for the prediction of STAS using peritumoral radiomics features extracted from preoperative CT imaging. A total of 339 patients who underwent lobectomy or limited resection for lung adenocarcinoma were included. The patients were randomly divided (3:2) into training and test cohorts. Two prediction models were created using the training cohort: a conventional model based on the tumor consolidation/tumor (C/T) ratio and a machine learning model based on peritumoral radiomics features. The areas under the curve for the two models in the testing cohort were 0.70 and 0.76, respectively (P = 0.045). The cumulative incidence of recurrence (CIR) was significantly higher in the STAS high-risk group when using the radiomics model than that in the low-risk group (44% vs. 4% at 5 years; P = 0.002) in patients who underwent limited resection in the testing cohort. In contrast, the 5-year CIR was not significantly different among patients who underwent lobectomy (17% vs. 11%; P = 0.469). In conclusion, the machine learning model for STAS prediction based on peritumoral radiomics features performed better than the C/T ratio model.
Collapse
Affiliation(s)
- Keiichi Takehana
- Department of Radiation Oncology and Image-Applied Therapy, Graduate School of Medicine, Kyoto University, 54 Shogoinkawahara-cho, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Ryo Sakamoto
- Department of Diagnostic Imaging and Nuclear Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Koji Fujimoto
- Department of Diagnostic Imaging and Nuclear Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Yukinori Matsuo
- Department of Radiation Oncology and Image-Applied Therapy, Graduate School of Medicine, Kyoto University, 54 Shogoinkawahara-cho, Sakyo-ku, Kyoto, 606-8507, Japan.
| | - Naoki Nakajima
- Department of Diagnostic Pathology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Akihiko Yoshizawa
- Department of Diagnostic Pathology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Toshi Menju
- Department of Thoracic Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Mitsuhiro Nakamura
- Division of Medical Physics, Department of Information Technology and Medical Engineering, Human Health Sciences, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Ryo Yamada
- Department of Statistical Genetics, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Takashi Mizowaki
- Department of Radiation Oncology and Image-Applied Therapy, Graduate School of Medicine, Kyoto University, 54 Shogoinkawahara-cho, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Yuji Nakamoto
- Department of Diagnostic Imaging and Nuclear Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| |
Collapse
|
48
|
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: 2.0] [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
|
49
|
Wang F, Tan BF, Poh SS, Siow TR, Lim FLWT, Yip CSP, Wang MLC, Nei W, Tan HQ. Predicting outcomes for locally advanced rectal cancer treated with neoadjuvant chemoradiation with CT-based radiomics. Sci Rep 2022; 12:6167. [PMID: 35418656 PMCID: PMC9008122 DOI: 10.1038/s41598-022-10175-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2021] [Accepted: 03/31/2022] [Indexed: 12/24/2022] Open
Abstract
A feasibility study was performed to determine if CT-based radiomics could play an augmentative role in predicting neoadjuvant rectal score (NAR), locoregional failure free survival (LRFFS), distant metastasis free survival (DMFS), disease free survival (DFS) and overall survival (OS) in locally advanced rectal cancer (LARC). The NAR score, which takes into account the pathological tumour and nodal stage as well as clinical tumour stage, is a validated surrogate endpoint used for early determination of treatment response whereby a low NAR score (< 8) has been correlated with better outcomes and high NAR score (> 16) has been correlated with poorer outcomes. CT images of 191 patients with LARC were used in this study. Primary tumour (GTV) and mesorectum (CTV) were contoured separately and radiomics features were extracted from both segments. Two NAR models (NAR > 16 and NAR < 8) models were constructed using Least Absolute Shrinkage and Selection Operator (LASSO) and the survival models were constructed using regularized Cox regressions. Area under curve (AUC) and time-dependent AUC were used to quantify the performance of the LASSO and Cox regression respectively, using ten folds cross validations. The NAR > 16 and NAR < 8 models have an average AUCs of 0.68 ± 0.13 and 0.59 ± 0.14 respectively. There are statistically significant differences between the clinical and combined model for LRFFS (from 0.68 ± 0.04 to 0.72 ± 0.04), DMFS (from 0.68 ± 0.05 to 0.70 ± 0.05) and OS (from 0.64 ± 0.06 to 0.66 ± 0.06). CTV radiomics features were also found to be more important than GTV features in the NAR prediction model. The most important clinical features are age and CEA for NAR > 16 and NAR < 8 models respectively, while the most significant clinical features are age, surgical margin and NAR score across all the four survival models.
Collapse
Affiliation(s)
- Fuqiang Wang
- Division of Radiation Oncology, National Cancer Centre Singapore, Singapore, Singapore.
| | - Boon Fei Tan
- Division of Radiation Oncology, National Cancer Centre Singapore, Singapore, Singapore
| | - Sharon Shuxian Poh
- Division of Radiation Oncology, National Cancer Centre Singapore, Singapore, Singapore
| | - Tian Rui Siow
- Division of Radiation Oncology, National Cancer Centre Singapore, Singapore, Singapore
| | | | - Connie Siew Poh Yip
- Division of Radiation Oncology, National Cancer Centre Singapore, Singapore, Singapore
| | | | - Wenlong Nei
- Division of Radiation Oncology, National Cancer Centre Singapore, Singapore, Singapore
| | - Hong Qi Tan
- Division of Radiation Oncology, National Cancer Centre Singapore, Singapore, Singapore.
| |
Collapse
|
50
|
Davey A, van Herk M, Faivre-Finn C, McWilliam A. Radial Data Mining to Identify Density-Dose Interactions That Predict Distant Failure Following SABR. Front Oncol 2022; 12:838155. [PMID: 35356210 PMCID: PMC8959483 DOI: 10.3389/fonc.2022.838155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Accepted: 02/11/2022] [Indexed: 11/13/2022] Open
Abstract
Purpose Lower dose outside the planned treatment area in lung stereotactic radiotherapy has been linked to increased risk of distant metastasis (DM) possibly due to underdosage of microscopic disease (MDE). Independently, tumour density on pretreatment computed tomography (CT) has been linked to risk of MDE. No studies have investigated the interaction between imaging biomarkers and incidental dose. The interaction would showcase whether the impact of dose on outcome is dependent on imaging and, hence, if imaging could inform which patients require dose escalation outside the gross tumour volume (GTV). We propose an image-based data mining methodology to investigate density-dose interactions radially from the GTV to predict DM with no a priori assumption on location. Methods Dose and density were quantified in 1-mm annuli around the GTV for 199 patients with early-stage lung cancer treated with 60 Gy in 5 fractions. Each annulus was summarised by three density and three dose parameters. For parameter combinations, Cox regressions were performed including a dose-density interaction in independent annuli. Heatmaps were created that described improvement in DM prediction due to the interaction. Regions of significant improvement were identified and studied in overall outcome models. Results Dose-density interactions were identified that significantly improved prediction for over 50% of bootstrap resamples. Dose and density parameters were not significant when the interaction was omitted. Tumour density variance and high peritumour density were associated with DM for patients with more cold spots (less than 30-Gy EQD2) and non-uniform dose about 3 cm outside of the GTV. Associations identified were independent of the mean GTV dose. Conclusions Patients with high tumour variance and peritumour density have increased risk of DM if there is a low and non-uniform dose outside the GTV. The dose regions are independent of tumour dose, suggesting that incidental dose may play an important role in controlling occult disease. Understanding such interactions is key to identifying patients who will benefit from dose-escalation. The methodology presented allowed spatial dose-density interactions to be studied at the exploratory stage for the first time. This could accelerate the clinical implementation of imaging biomarkers by demonstrating the impact of incidental dose for tumours of varying characteristics in routine data.
Collapse
Affiliation(s)
- Angela Davey
- Division of Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom
| | - Marcel van Herk
- Division of Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom.,Department of Radiotherapy Related Research, The Christie National Health Service (NHS) Foundation Trust, Manchester, United Kingdom
| | - Corinne Faivre-Finn
- Division of Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom.,Department of Radiotherapy Related Research, The Christie National Health Service (NHS) Foundation Trust, Manchester, United Kingdom.,Department of Clinical Oncology, The Christie National Health Service (NHS) Foundation Trust, Manchester, United Kingdom
| | - Alan McWilliam
- Division of Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom.,Department of Radiotherapy Related Research, The Christie National Health Service (NHS) Foundation Trust, Manchester, United Kingdom
| |
Collapse
|