1
|
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
|
2
|
Suh YJ, Han K, Kwon Y, Kim H, Lee S, Hwang SH, Kim MH, Shin HJ, Lee CY, Shim HS. Computed Tomography Radiomics for Preoperative Prediction of Spread Through Air Spaces in the Early Stage of Surgically Resected Lung Adenocarcinomas. Yonsei Med J 2024; 65:163-173. [PMID: 38373836 PMCID: PMC10896671 DOI: 10.3349/ymj.2023.0368] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 10/19/2023] [Accepted: 10/25/2023] [Indexed: 02/21/2024] Open
Abstract
PURPOSE To assess the added value of radiomics models from preoperative chest CT in predicting the presence of spread through air spaces (STAS) in the early stage of surgically resected lung adenocarcinomas using multiple validation datasets. MATERIALS AND METHODS This retrospective study included 550 early-stage surgically resected lung adenocarcinomas in 521 patients, classified into training, test, internal validation, and temporal validation sets (n=211, 90, 91, and 158, respectively). Radiomics features were extracted from the segmented tumors on preoperative chest CT, and a radiomics score (Rad-score) was calculated to predict the presence of STAS. Diagnostic performance of the conventional model and the combined model, based on a combination of conventional and radiomics features, for the diagnosis of the presence of STAS were compared using the area under the curve (AUC) of the receiver operating characteristic curve. RESULTS Rad-score was significantly higher in the STAS-positive group compared to the STAS-negative group in the training, test, internal, and temporal validation sets. The performance of the combined model was significantly higher than that of the conventional model in the training set {AUC: 0.784 [95% confidence interval (CI): 0.722-0.846] vs. AUC: 0.815 (95% CI: 0.759-0.872), p=0.042}. In the temporal validation set, the combined model showed a significantly higher AUC than that of the conventional model (p=0.001). The combined model showed a higher AUC than the conventional model in the test and internal validation sets, albeit with no statistical significance. CONCLUSION A quantitative CT radiomics model can assist in the non-invasive prediction of the presence of STAS in the early stage of lung adenocarcinomas.
Collapse
Affiliation(s)
- Young Joo Suh
- Department of Radiology, Research Institute of Radiological Science, Severance Hospital, Yonsei University College of Medicine, Seoul, Korea.
| | - Kyunghwa Han
- Department of Radiology, Research Institute of Radiological Science, Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Yonghan Kwon
- Department of Biostatistics and Computing, Yonsei University Graduate School, Seoul, Korea
| | - Hwiyoung Kim
- Department of Biomedical System Informatics, Yonsei University College of Medicine, Seoul, Korea
| | - Suji Lee
- Department of Radiology, Research Institute of Radiological Science, Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Sung Ho Hwang
- Department of Radiology, Korea University Anam Hospital, Korea University College of Medicine, Seoul, Korea
| | - Myung Hyun Kim
- Department of Radiology, Yongin Severance Hospital, Yonsei University College of Medicine, Yongin, Korea
| | - Hyun Joo Shin
- Department of Radiology, Yongin Severance Hospital, Yonsei University College of Medicine, Yongin, Korea
| | - Chang Young Lee
- Thoracic and Cardiovascular Surgery, Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Hyo Sup Shim
- Department of Pathology, Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| |
Collapse
|
3
|
Gu J, Bao S, Akemuhan R, Jia Z, Zhang Y, Huang C. Radiomics Based on Contrast-Enhanced CT for Recognizing c-Met-Positive Hepatocellular Carcinoma: a Noninvasive Approach to Predict the Outcome of Sorafenib Resistance. Mol Imaging Biol 2023; 25:1073-1083. [PMID: 37932610 DOI: 10.1007/s11307-023-01870-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2023] [Revised: 10/16/2023] [Accepted: 10/23/2023] [Indexed: 11/08/2023]
Abstract
OBJECTIVES The purpose of our project was to investigate the effectiveness of radiomic features based on contrast-enhanced computed tomography (CT) that can detect the expression of c-Met in hepatocellular carcinoma (HCC) and to validate its efficacy in predicting the outcome of sorafenib resistance. MATERIALS AND METHODS In total, 130 patients (median age, 60 years) with pathologically confirmed HCC who underwent contrast material-enhanced CT from October 2012 to July 2020 were randomly divided into a training set (n = 91) and a test set (n = 39). Radiomic features were extracted from arterial phase (AP), portal venous phase (VP) and delayed phase (DP) images of every participant's enhanced CT images. RESULTS The entire group comprised 39 Met-positive and 91 Met-negative patients. The combined model, which included the clinical factors and the radiomic features, performed well in the training (area under the curve [AUC] = 0.878) and validation (AUC = 0.851) cohorts. The nomogram, which relied on the combined model, fits well in the calibration curves. Decision curve analysis (DCA) further confirmed that the clinical valuation of the nomogram achieved comparable accuracy in c-Met prediction. Among another 20 patients with HCC who had received sorafenib, the predicted high-risk group had shorter overall survival (OS) than the predicted low-risk group (p < 0.05). CONCLUSION A multivariate model acquired from three phases (AP, VP and DP) of enhanced CT, HBV-DNA and γ glutamyl transpeptidase isoenzyme II (GGT-II) could be considered a satisfactory preoperative marker of the expression of c-Met in patients with HCC. This approach may help in overcoming sorafenib resistance in advanced HCC.
Collapse
Affiliation(s)
- Jingxiao Gu
- Department of Vascular Surgery, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, 226001, the, People's Republic of China
- Department of Radiology, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, China
| | - Shanlei Bao
- Department of Nuclear Medicine, Affiliated Hospital of Nantong University, Nantong, China
| | | | - Zhongzheng Jia
- Department of Radiology, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, China.
| | - Yu Zhang
- Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hospital of Nantong University, Nantong, China.
| | - Chen Huang
- Department of Vascular Surgery, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, 226001, the, People's Republic of China.
| |
Collapse
|
4
|
Guo C, Zhou H, Chen X, Feng Z. Computed tomography texture-based models for predicting KIT exon 11 mutation of gastrointestinal stromal tumors. Heliyon 2023; 9:e20983. [PMID: 37876490 PMCID: PMC10590931 DOI: 10.1016/j.heliyon.2023.e20983] [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: 02/21/2023] [Revised: 10/11/2023] [Accepted: 10/12/2023] [Indexed: 10/26/2023] Open
Abstract
Background KIT exon 11 mutation in gastrointestinal stromal tumors (GISTs) is associated with treatment strategies. However, few studies have shown the role of imaging-based texture analysis in KIT exon 11 mutation in GISTs. In this study, we aimed to show the association between computed tomography (CT)-based texture features and KIT exon 11 mutation. Methods Ninety-five GISTs confirmed by surgery and identified with mutational genotype of KIT were included in this study. By amplifying the samples using over-sampling technique, a total of 183 region of interest (ROI) segments were extracted from 63 patients as training cohort. The 63 new ROI segments were extracted from the 63 patients as internal validation cohort. Thirty-two patients who underwent KIT exon 11 mutation test during 2021-2023 was selected as external validation cohort. The textural parameters were evaluated both in training cohort and validation cohort. Least absolute shrinkage and selection operator (LASSO) algorithms and logistic regression analysis were used to select the discriminant features. Results Three of textural features were obtained using LASSO analysis. Logistic regression analysis showed that patients' age, tumor location and radiomics features were significantly associated with KIT exon 11 mutation (p < 0.05). A nomogram was developed based on the associated factors. The area under the curve (AUC) of clinical features, radiomics features and their combination in training cohort was 0.687 (95 % CI: 0.604-0.771), 0.829 (95 % CI: 0.768-0.890) and 0.874 (95 % CI: 0.822-0.926), respectively. The AUC of radiomics features in internal validation cohort and external cohort was 0.880 (95 % CI: 0.796-0.964) and 0.827 (95%CI: 0.667-0.987), respectively. Conclusion The CT texture-based model can be used to predict KIT exon 11 mutation in GISTs.
Collapse
Affiliation(s)
- Chuangen Guo
- Department of Radiology, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Road, Hangzhou, 310003, China
| | - Hao Zhou
- Department of Radiology, The Affiliated Hospital of Nanjing University of Chinese Medicine, 155 hanzhong Road, Nanjing, 210029, China
| | - Xiao Chen
- Department of Radiology, The Affiliated Hospital of Nanjing University of Chinese Medicine, 155 hanzhong Road, Nanjing, 210029, China
| | - Zhan Feng
- Department of Radiology, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Road, Hangzhou, 310003, China
| |
Collapse
|
5
|
Gao J, Niu R, Shi Y, Shao X, Jiang Z, Ge X, Wang Y, Shao X. The predictive value of [ 18F]FDG PET/CT radiomics combined with clinical features for EGFR mutation status in different clinical staging of lung adenocarcinoma. EJNMMI Res 2023; 13:26. [PMID: 37014500 PMCID: PMC10073367 DOI: 10.1186/s13550-023-00977-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Accepted: 03/24/2023] [Indexed: 04/05/2023] Open
Abstract
BACKGROUND This study aims to construct radiomics models based on [18F]FDG PET/CT using multiple machine learning methods to predict the EGFR mutation status of lung adenocarcinoma and evaluate whether incorporating clinical parameters can improve the performance of radiomics models. METHODS A total of 515 patients were retrospectively collected and divided into a training set (n = 404) and an independent testing set (n = 111) according to their examination time. After semi-automatic segmentation of PET/CT images, the radiomics features were extracted, and the best feature sets of CT, PET, and PET/CT modalities were screened out. Nine radiomics models were constructed using logistic regression (LR), random forest (RF), and support vector machine (SVM) methods. According to the performance in the testing set, the best model of the three modalities was kept, and its radiomics score (Rad-score) was calculated. Furthermore, combined with the valuable clinical parameters (gender, smoking history, nodule type, CEA, SCC-Ag), a joint radiomics model was built. RESULTS Compared with LR and SVM, the RF Rad-score showed the best performance among the three radiomics models of CT, PET, and PET/CT (training and testing sets AUC: 0.688, 0.666, and 0.698 vs. 0.726, 0.678, and 0.704). Among the three joint models, the PET/CT joint model performed the best (training and testing sets AUC: 0.760 vs. 0.730). The further stratified analysis found that CT_RF had the best prediction effect for stage I-II lesions (training set and testing set AUC: 0.791 vs. 0.797), while PET/CT joint model had the best prediction effect for stage III-IV lesions (training and testing sets AUC: 0.722 vs. 0.723). CONCLUSIONS Combining with clinical parameters can improve the predictive performance of PET/CT radiomics model, especially for patients with advanced lung adenocarcinoma.
Collapse
Affiliation(s)
- Jianxiong Gao
- Department of Nuclear Medicine, The Third Affiliated Hospital of Soochow University, Changzhou, 213003, China
- Institute of Clinical Translation of Nuclear Medicine and Molecular Imaging, Soochow University, Changzhou, 213003, China
| | - Rong Niu
- Department of Nuclear Medicine, The Third Affiliated Hospital of Soochow University, Changzhou, 213003, China
- Institute of Clinical Translation of Nuclear Medicine and Molecular Imaging, Soochow University, Changzhou, 213003, China
| | - Yunmei Shi
- Department of Nuclear Medicine, The Third Affiliated Hospital of Soochow University, Changzhou, 213003, China
- Institute of Clinical Translation of Nuclear Medicine and Molecular Imaging, Soochow University, Changzhou, 213003, China
| | - Xiaoliang Shao
- Department of Nuclear Medicine, The Third Affiliated Hospital of Soochow University, Changzhou, 213003, China
- Institute of Clinical Translation of Nuclear Medicine and Molecular Imaging, Soochow University, Changzhou, 213003, China
| | - Zhenxing Jiang
- Department of Radiology, The Third Affiliated Hospital of Soochow University, Changzhou, 213003, China
| | - Xinyu Ge
- Department of Nuclear Medicine, The Third Affiliated Hospital of Soochow University, Changzhou, 213003, China
- Institute of Clinical Translation of Nuclear Medicine and Molecular Imaging, Soochow University, Changzhou, 213003, China
| | - Yuetao Wang
- Department of Nuclear Medicine, The Third Affiliated Hospital of Soochow University, Changzhou, 213003, China
- Institute of Clinical Translation of Nuclear Medicine and Molecular Imaging, Soochow University, Changzhou, 213003, China
| | - Xiaonan Shao
- Department of Nuclear Medicine, The Third Affiliated Hospital of Soochow University, Changzhou, 213003, China.
- Institute of Clinical Translation of Nuclear Medicine and Molecular Imaging, Soochow University, Changzhou, 213003, China.
- Changzhou Key Laboratory of Molecular Imaging, Changzhou, 213003, China.
| |
Collapse
|
6
|
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: 6.0] [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
|
7
|
Hao P, Deng BY, Huang CT, Xu J, Zhou F, Liu ZX, Zhou W, Xu YK. Predicting anaplastic lymphoma kinase rearrangement status in patients with non-small cell lung cancer using a machine learning algorithm that combines clinical features and CT images. Front Oncol 2022; 12:994285. [PMID: 36338735 PMCID: PMC9630325 DOI: 10.3389/fonc.2022.994285] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Accepted: 09/26/2022] [Indexed: 12/01/2023] Open
Abstract
PURPOSE To develop an appropriate machine learning model for predicting anaplastic lymphoma kinase (ALK) rearrangement status in non-small cell lung cancer (NSCLC) patients using computed tomography (CT) images and clinical features. METHOD AND MATERIALS This study included 193 patients with NSCLC (154 in the training cohort, 39 in the validation cohort), 68 of whom tested positive for ALK rearrangements and 125 of whom tested negative. From the nonenhanced CT scans, 157 radiomic characteristics were extracted, and 8 clinical features were collected. Five machine learning (ML) models were assessed to find the best classification model for predicting ALK rearrangement status. A radiomic signature was developed using the least absolute shrinkage and selection operator (LASSO) algorithm. The predictive performance of the models based on radiomic features, clinical features, and their combination was assessed by receiver operating characteristic (ROC) curves. RESULTS The support vector machine (SVM) model had the highest AUC of 0.914 for classification. The clinical features model had an AUC=0.805 (95% CI 0.731-0.877) and an AUC=0.735 (95% CI 0.566-0.863) in the training and validation cohorts, respectively. The CT image-based ML model had an AUC=0.953 (95% CI 0.913-1.0) in the training cohort and an AUC=0.890 (95% CI 0.778-0.971) in the validation cohort. For predicting ALK rearrangement status, the ML model based on CT images and clinical features performed better than the model based on only clinical information or CT images, with an AUC of 0.965 (95% CI 0.826-0.882) in the primary cohort and an AUC of 0.914 (95% CI 0.804-0.893) in the validation cohort. CONCLUSION Our findings revealed that ALK rearrangement status could be accurately predicted using an ML-based classification model based on CT images and clinical data.
Collapse
Affiliation(s)
- Peng Hao
- Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Bo-Yu Deng
- School of Medical Information Engineering, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Chan-Tao Huang
- Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jun Xu
- Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Fang Zhou
- Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Zhe-Xing Liu
- School of Biomedical Engineering, Southern Medical Uinversity, Guangzhou, China
| | - Wu Zhou
- School of Medical Information Engineering, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yi-Kai Xu
- Nanfang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
8
|
Anagnostopoulos AK, Gaitanis A, Gkiozos I, Athanasiadis EI, Chatziioannou SN, Syrigos KN, Thanos D, Chatziioannou AN, Papanikolaou N. Radiomics/Radiogenomics in Lung Cancer: Basic Principles and Initial Clinical Results. Cancers (Basel) 2022; 14:cancers14071657. [PMID: 35406429 PMCID: PMC8997041 DOI: 10.3390/cancers14071657] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Revised: 03/14/2022] [Accepted: 03/16/2022] [Indexed: 02/01/2023] Open
Abstract
Simple Summary Radiogenomics is a promising new approach in cancer assessment, providing an evaluation of the molecular basis of imaging phenotypes after establishing associations between radiological features and molecular features at the genomic–transcriptomic–proteomic level. This review focuses on describing key aspects of radiogenomics while discussing limitations of translatability to the clinic and possible solutions to these challenges, providing the clinician with an up-to-date handbook on how to use this new tool. Abstract Lung cancer is the leading cause of cancer-related deaths worldwide, and elucidation of its complicated pathobiology has been traditionally targeted by studies incorporating genomic as well other high-throughput approaches. Recently, a collection of methods used for cancer imaging, supplemented by quantitative aspects leading towards imaging biomarker assessment termed “radiomics”, has introduced a novel dimension in cancer research. Integration of genomics and radiomics approaches, where identifying the biological basis of imaging phenotypes is feasible due to the establishment of associations between molecular features at the genomic–transcriptomic–proteomic level and radiological features, has recently emerged termed radiogenomics. This review article aims to briefly describe the main aspects of radiogenomics, while discussing its basic limitations related to lung cancer clinical applications for clinicians, researchers and patients.
Collapse
Affiliation(s)
- Athanasios K. Anagnostopoulos
- Division of Biotechnology, Center of Systems Biology, Biomedical Research Foundation of the Academy of Athens (BRFAA), 11525 Athens, Greece
- Correspondence:
| | - Anastasios Gaitanis
- Clinical and Translational Research, Center of Experimental Surgery, Biomedical Research Foundation of the Academy of Athens (BRFAA), 11527 Athens, Greece;
| | - Ioannis Gkiozos
- Third Department of Internal Medicine, “Sotiria” Hospital, National and Kapodistrian University of Athens, 11527 Athens, Greece; (I.G.); (K.N.S.)
| | - Emmanouil I. Athanasiadis
- Greek Genome Centre, Biomedical Research Foundation of the Academy of Athens (BRFAA), 11527 Athens, Greece; (E.I.A.); (D.T.)
| | - Sofia N. Chatziioannou
- Nuclear Medicine Division, Biomedical Research Foundation of the Academy of Athens (BRFAA), 11527 Athens, Greece;
| | - Konstantinos N. Syrigos
- Third Department of Internal Medicine, “Sotiria” Hospital, National and Kapodistrian University of Athens, 11527 Athens, Greece; (I.G.); (K.N.S.)
| | - Dimitris Thanos
- Greek Genome Centre, Biomedical Research Foundation of the Academy of Athens (BRFAA), 11527 Athens, Greece; (E.I.A.); (D.T.)
| | - Achilles N. Chatziioannou
- First Department of Radiology, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece;
| | - Nikolaos Papanikolaou
- Computational Clinical Imaging Group, Centre for the Unknown, Champalimaud Foundation, 1400-038 Lisbon, Portugal;
- Machine Learning Group, The Royal Marsden, London SM2 5MG, UK
- The Institute of Cancer Research, London SM2 5MG, UK
- Karolinska Institutet, 14186 Stockholm, Sweden
- Institute of Computer Science, FORTH, 70013 Heraklion, Greece
| |
Collapse
|
9
|
Yao X, Mao L, Yi K, Han Y, Li W, Xiao Y, Ji J, Wang Q, Ren K. Radiomic Signature as a Diagnostic Factor for Classification of Histologic Subtypes of Lung Cancer. JOURNAL OF MEDICAL IMAGING AND HEALTH INFORMATICS 2021. [DOI: 10.1166/jmihi.2021.3564] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
<sec> <title>Objectives:</title> To discuss the application of radiomics using Computerized Tomography (CT) analysis, for improving its diagnostic efficacy in lung, specifically in distinguishing Squamous Cell Carcinoma (SCC), lung Adenocarcinoma (ADC),
and Small Cell Lung Cancer (SCLC). </sec> <sec> <title>Methods:</title> The pathology of 189 identified cases of lung cancer was analyzed, retrospectively (60 patients with SCC, 69 patients with lung ADC and 60 patients with SCLC). A neural network was used
to determine whether the pulmonary or mediastinal window was selected to extract effective radiomic features. The key features of radiomic signature were retrieved by a Least Absolute Shrinkage and Selection Operator (LASSO) multiple logistic regression model. Next, receiver operating characteristic
curve and Area Under the Curve (AUC) analysis were used to evaluate the performance of the radiomic signature in both, training(129 patients) and validation cohorts (60 patients). </sec> <sec> <title>Results:</title> About 295 features were extracted from
a manually outlined tumor region. Features extracted from mediastinal window CT scans had a better prognostic ability than pulmonary window scans. The average accuracy for mediastinal window scans was 0.933. Our analysis revealed that the radiomic features extracted from mediastinal window
scans had the potential to build a prediction model for distinguishing between SCC, lung ADC, and SCLC. The performance of the radiomic signature to diagnose SCC and SCLC in validation cohorts proved effective, with AUC values of 0.869 and 0.859, respectively. </sec> <sec> <title>Conclusions:</title>
A unique radiomic signature was constructed as a diagnostic factor for different histologic subtypes of lung cancer. Patients with lung cancer may benefit from this proposed radiomic signature. </sec>
Collapse
Affiliation(s)
- Xiang Yao
- Department of Radiology, Xiang’an Hospital of XiaMen University, XiaMen 361000, Fujian, China
| | - Ling Mao
- The School of Economics, XiaMen University, XiaMen, Fujian, 361000, China
| | - Ke Yi
- Department of Respiratory and Critical Care Medicine, Sichuan Science City Hospital, Mianyang, Sichuan, 621000, China
| | - Yuxiao Han
- Yang Zhou University, Yangzhou, Jiangsu, 225000, China
| | - Wentao Li
- Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210000, China
| | - Yingqi Xiao
- West China School of Nursing/West China Hospital, Sichuan University, Chengdu, Sichuan, 610000, China
| | - Jun Ji
- Department of Pathology, Sunning People’s Hospital, Xuzhou, Jiangsu, 221000, China
| | - Qingqing Wang
- Department of Nephrology, Xuzhou Children’s Hospital, Xuzhou, Jiangsu, 221000, China
| | - Ke Ren
- Department of Radiology, Xiang’an Hospital of XiaMen University, XiaMen 361000, Fujian, China
| |
Collapse
|
10
|
Bracci S, Dolciami M, Trobiani C, Izzo A, Pernazza A, D'Amati G, Manganaro L, Ricci P. Quantitative CT texture analysis in predicting PD-L1 expression in locally advanced or metastatic NSCLC patients. Radiol Med 2021; 126:1425-1433. [PMID: 34373989 PMCID: PMC8558266 DOI: 10.1007/s11547-021-01399-9] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Accepted: 07/06/2021] [Indexed: 12/18/2022]
Abstract
Purpose The assessment of Programmed death-ligand 1 (PD-L1) expression has become a game changer in the treatment of patients with advanced non-small cell lung cancer (NSCLC). We aimed to investigate the ability of Radiomics applied to computed tomography (CT) in predicting PD-L1 expression in patients with advanced NSCLC. Methods By applying texture analysis, we retrospectively analyzed 72 patients with advanced NSCLC. The datasets were randomly split into a training cohort (2/3) and a validation cohort (1/3). Forty radiomic features were extracted by manually drawing tumor volumes of interest (VOIs) on baseline contrast-enhanced CT. After selecting features on the training cohort, two predictive models were created using binary logistic regression, one for PD-L1 values ≥ 50% and the other for values between 1 and 49%. The two models were analyzed with ROC curves and tested in the validation cohort. Results The Radiomic Score (Rad-Score) for PD-L1 values ≥ 50%, which consisted of Skewness and Low Gray-Level Zone Emphasis (GLZLM_LGZE), presented a cut-off value of − 0.745 with an area under the curve (AUC) of 0.811 and 0.789 in the training and validation cohort, respectively. The Rad-Score for PD-L1 values between 1 and 49% consisted of Sphericity, Skewness, Conv_Q3 and Gray Level Non-Uniformity (GLZLM_GLNU), showing a cut-off value of 0.111 with AUC of 0.763 and 0.806 in the two population, respectively. Conclusion Rad-Scores obtained from CT texture analysis could be useful for predicting PD-L1 expression and guiding the therapeutic choice in patients with advanced NSCLC.
Collapse
Affiliation(s)
- Stefano Bracci
- Department of Radiological, Oncological, and Pathological Sciences, Sapienza University of Rome, Viale Regina Elena 324, 00161, Rome, Italy
| | - Miriam Dolciami
- Department of Radiological, Oncological, and Pathological Sciences, Sapienza University of Rome, Viale Regina Elena 324, 00161, Rome, Italy
| | - Claudio Trobiani
- Department of Radiological, Oncological, and Pathological Sciences, Sapienza University of Rome, Viale Regina Elena 324, 00161, Rome, Italy
| | - Antonella Izzo
- Department of Radiological, Oncological, and Pathological Sciences, Sapienza University of Rome, Viale Regina Elena 324, 00161, Rome, Italy
| | - Angelina Pernazza
- Department of Radiological, Oncological, and Pathological Sciences, Sapienza University of Rome, Viale Regina Elena 324, 00161, Rome, Italy
| | - Giulia D'Amati
- Department of Radiological, Oncological, and Pathological Sciences, Sapienza University of Rome, Viale Regina Elena 324, 00161, Rome, Italy
| | - Lucia Manganaro
- Department of Radiological, Oncological, and Pathological Sciences, Sapienza University of Rome, Viale Regina Elena 324, 00161, Rome, Italy
| | - Paolo Ricci
- Department of Radiological, Oncological, and Pathological Sciences, Sapienza University of Rome, Viale Regina Elena 324, 00161, Rome, Italy.
| |
Collapse
|
11
|
Cucchiara F, Petrini I, Romei C, Crucitta S, Lucchesi M, Valleggi S, Scavone C, Capuano A, De Liperi A, Chella A, Danesi R, Del Re M. Combining liquid biopsy and radiomics for personalized treatment of lung cancer patients. State of the art and new perspectives. Pharmacol Res 2021; 169:105643. [PMID: 33940185 DOI: 10.1016/j.phrs.2021.105643] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Revised: 04/22/2021] [Accepted: 04/22/2021] [Indexed: 12/11/2022]
Abstract
Lung cancer has become a paradigm for precision medicine in oncology, and liquid biopsy (LB) together with radiomics may have a great potential in this scenario. They are both minimally invasive, easy to perform, and can be repeated during patient's follow-up. Also, increasing evidence suggest that LB and radiomics may provide an efficient way to screen and diagnose tumors at an early stage, including the monitoring of any change in the tumor molecular profile. This could allow treatment optimization, improvement of patients' quality of life, and healthcare-related costs reduction. Latest reports on lung cancer patients suggest a combination of these two strategies, along with cutting-edge data analysis, to decode valuable information regarding tumor type, aggressiveness, progression, and response to treatment. The approach seems more compatible with clinical practice than the current standard, and provides new diagnostic companions being able to suggest the best treatment strategy compared to conventional methods. To implement radiomics and liquid biopsy directly into clinical practice, an artificial intelligence (AI)-based system could help to link patients' clinical data together with tumor molecular profiles and imaging characteristics. AI could also solve problems and limitations related to LB and radiomics methodologies. Further work is needed, including new health policies and the access to large amounts of high-quality and well-organized data, allowing a complementary and synergistic combination of LB and imaging, to provide an attractive choice e in the personalized treatment of lung cancer.
Collapse
Affiliation(s)
- Federico Cucchiara
- Unit of Clinical Pharmacology and Pharmacogenetics, Department of Clinical and Experimental Medicine, University Hospital of Pisa, Pisa, Italy
| | - Iacopo Petrini
- Unit of Pneumology, Department of Translational Research and New Technologies in Medicine, University Hospital of Pisa, Pisa, Italy
| | - Chiara Romei
- Unit II of Radio-diagnostics, Department of Diagnostic and Imaging, University Hospital of Pisa, Pisa, Italy
| | - Stefania Crucitta
- Unit of Clinical Pharmacology and Pharmacogenetics, Department of Clinical and Experimental Medicine, University Hospital of Pisa, Pisa, Italy
| | - Maurizio Lucchesi
- Unit of Pneumology, Department of Translational Research and New Technologies in Medicine, University Hospital of Pisa, Pisa, Italy
| | - Simona Valleggi
- Unit of Pneumology, Department of Translational Research and New Technologies in Medicine, University Hospital of Pisa, Pisa, Italy
| | - Cristina Scavone
- Department of Experimental Medicine, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Annalisa Capuano
- Department of Experimental Medicine, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Annalisa De Liperi
- Unit II of Radio-diagnostics, Department of Diagnostic and Imaging, University Hospital of Pisa, Pisa, Italy
| | - Antonio Chella
- Unit of Pneumology, Department of Translational Research and New Technologies in Medicine, University Hospital of Pisa, Pisa, Italy
| | - Romano Danesi
- Unit of Clinical Pharmacology and Pharmacogenetics, Department of Clinical and Experimental Medicine, University Hospital of Pisa, Pisa, Italy.
| | - Marzia Del Re
- Unit of Clinical Pharmacology and Pharmacogenetics, Department of Clinical and Experimental Medicine, University Hospital of Pisa, Pisa, Italy
| |
Collapse
|
12
|
Zhu Y, Mao Y, Chen J, Qiu Y, Guan Y, Wang Z, He J. Value of contrast-enhanced CT texture analysis in predicting IDH mutation status of intrahepatic cholangiocarcinoma. Sci Rep 2021; 11:6933. [PMID: 33767315 PMCID: PMC7994625 DOI: 10.1038/s41598-021-86497-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2020] [Accepted: 03/16/2021] [Indexed: 12/31/2022] Open
Abstract
To explore the value of contrast-enhanced CT texture analysis in predicting isocitrate dehydrogenase (IDH) mutation status of intrahepatic cholangiocarcinomas (ICCs). Institutional review board approved this study. Contrast-enhanced CT images of 138 ICC patients (21 with IDH mutation and 117 without IDH mutation) were retrospectively reviewed. Texture analysis was performed for each lesion and compared between ICCs with and without IDH mutation. All textural features in each phase and combinations of textural features (p < 0.05) by Mann-Whitney U tests were separately used to train multiple support vector machine (SVM) classifiers. The classification generalizability and performance were evaluated using a tenfold cross-validation scheme. Among plain, arterial phase (AP), portal venous phase (VP), equilibrium phase (EP) and Sig classifiers, VP classifier showed the highest accuracy of 0.863 (sensitivity, 0.727; specificity, 0.885), with a mean area under the receiver operating characteristic curve of 0.813 in predicting IDH mutation in validation cohort. Texture features of CT images in portal venous phase could predict IDH mutation status of ICCs with SVM classifier preoperatively.
Collapse
Affiliation(s)
- Yong Zhu
- Department of Radiology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, Jiangsu Province, China
| | - Yingfan Mao
- Department of Radiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, No. 321 Zhongshan Road, Nanjing, 210008, Jiangsu Province, China
| | - Jun Chen
- Department of Pathology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, No. 321 Zhongshan Road, Nanjing, 210008, Jiangsu Province, China
| | - Yudong Qiu
- Department of Hepatopancreatobiliary Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, No. 321 Zhongshan Road, Nanjing, 210008, Jiangsu Province, China
| | - Yue Guan
- School of Biomedical Engineering, Shanghai Jiao Tong University, No. 1954 Huashan Road, Shanghai, 200000, China
| | - Zhongqiu Wang
- Department of Radiology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, Jiangsu Province, China.
| | - Jian He
- Department of Radiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, No. 321 Zhongshan Road, Nanjing, 210008, Jiangsu Province, China.
| |
Collapse
|
13
|
Abstract
With the ongoing advances in imaging techniques, increasing volumes of anatomical and functional data are being generated as part of the routine clinical workflow. This surge of available imaging data coincides with increasing research in quantitative imaging, particularly in the domain of imaging features. An important and novel approach is radiomics, where high-dimensional image properties are extracted from routine medical images. The fundamental principle of radiomics is the hypothesis that biomedical images contain predictive information, not discernible to the human eye, that can be mined through quantitative image analysis. In this review, a general outline of radiomics and artificial intelligence (AI) will be provided, along with prominent use cases in immunotherapy (e.g. response and adverse event prediction) and targeted therapy (i.e. radiogenomics). While the increased use and development of radiomics and AI in immuno-oncology is highly promising, the technology is still in its early stages, and different challenges still need to be overcome. Nevertheless, novel AI algorithms are being constructed with an ever-increasing scope of applications.
Collapse
Affiliation(s)
- Z. Bodalal
- Department of Radiology, Netherlands Cancer Institute, Amsterdam, The Netherlands
- GROW School for Oncology and Developmental Biology, Maastricht University, Maastricht, The Netherlands
| | - I. Wamelink
- Department of Radiology, Netherlands Cancer Institute, Amsterdam, The Netherlands
- Technical Medicine, University of Twente, Enschede, The Netherlands
| | - S. Trebeschi
- Department of Radiology, Netherlands Cancer Institute, Amsterdam, The Netherlands
- GROW School for Oncology and Developmental Biology, Maastricht University, Maastricht, The Netherlands
| | - R.G.H. Beets-Tan
- Department of Radiology, Netherlands Cancer Institute, Amsterdam, The Netherlands
- GROW School for Oncology and Developmental Biology, Maastricht University, Maastricht, The Netherlands
| |
Collapse
|
14
|
Dang Y, Wang R, Qian K, Lu J, Zhang H, Zhang Y. Clinical and radiological predictors of epidermal growth factor receptor mutation in nonsmall cell lung cancer. J Appl Clin Med Phys 2020; 22:271-280. [PMID: 33314737 PMCID: PMC7856515 DOI: 10.1002/acm2.13107] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 10/26/2020] [Accepted: 10/30/2020] [Indexed: 01/06/2023] Open
Abstract
Purpose To determine the prognostic factors of epidermal growth factor receptor (EGFR) mutation status in a group of patients with nonsmall cell lung cancer (NSCLC) by analyzing their clinical and radiological features. Materials and methods Patients with NSCLC who underwent EGFR mutation detection between 2014 and 2017 were included. Clinical features and general imaging features were collected, and radiomic features were extracted from CT data by 3D Slicer software. Prognostic factors of EGFR mutation status were selected by least absolute shrinkage and selection operator (LASSO) logistic regression analysis, and receiver operating characteristic (ROC) curves were drawn for each prediction model of EGFR mutation. Results A total of 118 patients were enrolled in this study. The smoking index (P = 0.028), pleural retraction (P = 0.041), and three radiomic features were significantly associated with EGFR mutation status. The areas under the ROC curve (AUCs) for prediction models of clinical features, general imaging features, and radiomic features were 0.284, 0.703, and 0.815, respectively, and the AUC for the combined prediction model of the three models was 0.894. Finally, a nomogram was established for individualized EGFR mutation prediction. Conclusions The combination of radiomic features with clinical features and general imaging features can enable discrimination of EGFR mutation status better than the use of any group of features alone. Our study may help develop a noninvasive biomarker to identify EGFR mutation status by using a combination of the three group features.
Collapse
Affiliation(s)
- Yutao Dang
- Department of Thoracic Surgery, Xuanwu Hospital, Capital Medical University, Beijing, China.,Department of Thoracic Surgery, Shijingshan Hospital of Beijing City, Shijingshan Teaching Hospital of Capital Medical University, Beijing, China
| | - Ruotian Wang
- Department of Thoracic Surgery, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Kun Qian
- Department of Thoracic Surgery, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Jie Lu
- Department of Radiology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Haixiang Zhang
- Center for Applied Mathematics, Tianjin University, Tianjin, China
| | - Yi Zhang
- Department of Thoracic Surgery, Xuanwu Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
15
|
Wu S, Shen G, Mao J, Gao B. CT Radiomics in Predicting EGFR Mutation in Non-small Cell Lung Cancer: A Single Institutional Study. Front Oncol 2020; 10:542957. [PMID: 33117680 PMCID: PMC7576846 DOI: 10.3389/fonc.2020.542957] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2020] [Accepted: 09/03/2020] [Indexed: 12/15/2022] Open
Abstract
Objective: To evaluate the value of CT radiomics in predicting the epidermal growth factor receptor (EGFR) mutation of patients with non-small cell lung cancer (NSCLC), and combing with the clinical characteristic to construct the prediction model. Methods: Sixty-seven cases of NSCLC confirmed by pathology were enrolled. The pre-treatment chest CT enhanced images were used in Radiomics analysis. Two experienced radiologists delineated the region of interest (ROI) on open source software 3D-Slicer. The feature of ROI was extracted by Pyradiomics software package and a total of 849 features were extracted. By calculating Pearson correlation coefficient between pair-wise features and LASSO method for feature screening. The prediction model was constructed by logical regression, diagnostic efficacy of the model by the area under the receiver operating characteristic (ROC) curve was calculated. Results: Based on clinical model and the radiomics model, the AUC under the ROC was 0.8387 and 0.8815, respectively. The model combining clinical and radiomics features perfect best, the AUC under the ROC was 0.9724, the sensitivity and specificity were 85.3 and 90.9%, respectively. Conclusions: Compared with clinical features or radiomics features alone, the model constructed by combining clinical and pre-treatment chest enhanced CT features may show more utility for improved patient stratification in EGFR mutation and EGFR wild.
Collapse
Affiliation(s)
- Shanshan Wu
- Department of Radiology, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Guiquan Shen
- Department of Radiology, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Jujiang Mao
- Department of Radiology, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Bo Gao
- Department of Radiology, The Affiliated Hospital of Guizhou Medical University, Guiyang, China.,Key Laboratory of Brain Imaging, Guizhou Medical University, Guiyang, China
| |
Collapse
|
16
|
Beyond tissue biopsy: a diagnostic framework to address tumor heterogeneity in lung cancer. Curr Opin Oncol 2020; 32:68-77. [PMID: 31714259 DOI: 10.1097/cco.0000000000000598] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
PURPOSE OF REVIEW The objective of this review is to discuss the strength and limitations of tissue and liquid biopsy and functional imaging to capture spatial and temporal tumor heterogeneity either alone or as part of a diagnostic framework in non-small cell lung cancer (NSCLC). RECENT FINDINGS NSCLC displays genetic and phenotypic heterogeneity - a detailed knowledge of which is crucial to personalize treatment. Tissue biopsy often lacks spatial and temporal resolution. Thus, NSCLC needs to be characterized by complementary diagnostic methods to resolve heterogeneity. Liquid biopsy offers detection of tumor biomarkers and for example, the classification and monitoring of EGFR mutations in NSCLC. It allows repeated sampling, and therefore, appears promising to address temporal aspects of tumor heterogeneity. Functional imaging methods and emerging image analytic tools, such as radiomics capture temporal and spatial heterogeneity. Further standardization of radiomics is required to allow introduction into clinical routine. SUMMARY To augment the potential of precision therapy, improved diagnostic characterization of tumors is pivotal. We suggest a comprehensive diagnostic framework combining tissue and liquid biopsy and functional imaging to address the known aspects of spatial and temporal tumor heterogeneity on the example of NSCLC. We envision how this framework might be implemented in clinical practice.
Collapse
|
17
|
Ladwa R, Roberts KE, O'Leary C, Maggacis N, O'Byrne KJ, Miles K. Computed tomography texture analysis of response to second-line nivolumab in metastatic non-small cell lung cancer. Lung Cancer Manag 2020; 9:LMT38. [PMID: 32774469 PMCID: PMC7399606 DOI: 10.2217/lmt-2020-0002] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Objectives: Assess computed tomography texture analysis of patients likely to benefit from nivolumab. Materials & methods: Texture analysis was used to quantify heterogeneity within the largest tumor before immunotherapy. Histogram analysis was classified as hyperdense (positive skewness) or hypodense (negative skewness) and subclassified on median standard deviation value or entropy measurement. Results: 47 patients were included. At a median follow-up of 18 months, statistical significant differences in progression-free survival were observed when stratified by positive skewness with low entropy, hazard ratio: 0.43 (0.19–0.95); p = 0.036, and positive skewness with low standard deviation, hazard ratio: 0.42 (0.18–0.96); p = 0.04. Conclusion: Patients who derive a clinical benefit to Nivolumab show a computed tomography texture of a hyperdense yet homogenous tumor.
Collapse
Affiliation(s)
- Rahul Ladwa
- Department of Medical Oncology, Princess Alexandra Hospital, Brisbane, QLD, Australia.,Department of Medical Imaging, Princess Alexandra Hospital, The University of Queensland, Brisbane, QLD, Australia
| | - Kate E Roberts
- Department of Medical Oncology, Princess Alexandra Hospital, Brisbane, QLD, Australia.,Department of Medical Imaging, Princess Alexandra Hospital, The University of Queensland, Brisbane, QLD, Australia
| | - Connor O'Leary
- Department of Medical Oncology, Princess Alexandra Hospital, Brisbane, QLD, Australia.,Queensland University of Technology, Brisbane, Australia
| | - Nicole Maggacis
- Department of Medical Oncology, Princess Alexandra Hospital, Brisbane, QLD, Australia
| | - Kenneth J O'Byrne
- Department of Medical Oncology, Princess Alexandra Hospital, Brisbane, QLD, Australia.,Queensland University of Technology, Brisbane, Australia
| | - Kenneth Miles
- Department of Medical Oncology, Princess Alexandra Hospital, Brisbane, QLD, Australia.,Department of Medical Imaging, Princess Alexandra Hospital, The University of Queensland, Brisbane, QLD, Australia
| |
Collapse
|
18
|
Baseline Computed Tomography Radiomic and Genomic Assessment of Head and Neck Squamous Cell Carcinoma. J Comput Assist Tomogr 2020; 44:546-552. [DOI: 10.1097/rct.0000000000001056] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
19
|
Liu Q, Sun D, Li N, Kim J, Feng D, Huang G, Wang L, Song S. Predicting EGFR mutation subtypes in lung adenocarcinoma using 18F-FDG PET/CT radiomic features. Transl Lung Cancer Res 2020; 9:549-562. [PMID: 32676319 PMCID: PMC7354146 DOI: 10.21037/tlcr.2020.04.17] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Background Identification of epidermal growth factor receptor (EGFR) mutation types is crucial before tyrosine kinase inhibitors (TKIs) treatment. Radiomics is a new strategy to noninvasively predict the genetic status of cancer. In this study, we aimed to develop a predictive model based on 18F-fluorodeoxyglucose positron emission tomography-computed tomography (18F-FDG PET/CT) radiomic features to identify the specific EGFR mutation subtypes. Methods We retrospectively studied 18F-FDG PET/CT images of 148 patients with isolated lung lesions, which were scanned in two hospitals with different CT scan setting (slice thickness: 3 and 5 mm, respectively). The tumor regions were manually segmented on PET/CT images, and 1,570 radiomic features (1,470 from CT and 100 from PET) were extracted from the tumor regions. Seven hundred and ninety-four radiomic features insensitive to different CT settings were first selected using the Mann white U test, and collinear features were further removed from them by recursively calculating the variation inflation factor. Then, multiple supervised machine learning models were applied to identify prognostic radiomic features through: (I) a multi-variate random forest to select features of high importance in discriminating different EGFR mutation status; (II) a logistic regression model to select features of the highest predictive value of the EGFR subtypes. The EGFR mutation predicting model was constructed from prognostic radiomic features using the popular Xgboost machine-learning algorithm and validated using 3-fold cross-validation. The performance of predicting model was analyzed using the receiver operating characteristic curve (ROC) and measured with the area under the curve (AUC). Results Two sets of prognostic radiomic features were found for specific EGFR mutation subtypes: 5 radiomic features for EGFR exon 19 deletions, and 5 radiomic features for EGFR exon 21 L858R missense. The corresponding radiomic predictors achieved the prediction accuracies of 0.77 and 0.92 in terms of AUC, respectively. Combing these two predictors, the overall model for predicting EGFR mutation positivity was also constructed, and the AUC was 0.87. Conclusions In our study, we established predictive models based on radiomic analysis of 18F-FDG PET/CT images. And it achieved a satisfying prediction power in the identification of EGFR mutation status as well as the certain EGFR mutation subtypes in lung cancer.
Collapse
Affiliation(s)
- Qiufang Liu
- Department of Nuclear Medicine, Fudan University Shanghai Cancer Center, Shanghai Medical College, Fudan University, Shanghai 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China.,SJTU-USYD Joint Research Alliance for Translational Medicine, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Dazhen Sun
- SJTU-USYD Joint Research Alliance for Translational Medicine, Shanghai Jiao Tong University, Shanghai 200240, China.,Department of Automation, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Nan Li
- Department of Nuclear Medicine, Fudan University Shanghai Cancer Center, Shanghai Medical College, Fudan University, Shanghai 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Jinman Kim
- SJTU-USYD Joint Research Alliance for Translational Medicine, Shanghai Jiao Tong University, Shanghai 200240, China.,Biomedical and Multimedia Information Technology Research Group, School of Computer Science, University of Sydney, Sydney, Australia.,Shanghai Key Laboratory for Molecular Imaging, Shanghai University of Medicine and Health Sciences, Shanghai 201318, China
| | - Dagan Feng
- SJTU-USYD Joint Research Alliance for Translational Medicine, Shanghai Jiao Tong University, Shanghai 200240, China.,Biomedical and Multimedia Information Technology Research Group, School of Computer Science, University of Sydney, Sydney, Australia.,Shanghai Key Laboratory for Molecular Imaging, Shanghai University of Medicine and Health Sciences, Shanghai 201318, China
| | - Gang Huang
- SJTU-USYD Joint Research Alliance for Translational Medicine, Shanghai Jiao Tong University, Shanghai 200240, China.,Shanghai Key Laboratory for Molecular Imaging, Shanghai University of Medicine and Health Sciences, Shanghai 201318, China.,Department of Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Lisheng Wang
- SJTU-USYD Joint Research Alliance for Translational Medicine, Shanghai Jiao Tong University, Shanghai 200240, China.,Department of Automation, Shanghai Jiao Tong University, Shanghai 200240, China.,Shanghai Key Laboratory for Molecular Imaging, Shanghai University of Medicine and Health Sciences, Shanghai 201318, China
| | - Shaoli Song
- Department of Nuclear Medicine, Fudan University Shanghai Cancer Center, Shanghai Medical College, Fudan University, Shanghai 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China.,SJTU-USYD Joint Research Alliance for Translational Medicine, Shanghai Jiao Tong University, Shanghai 200240, China.,Shanghai Key Laboratory for Molecular Imaging, Shanghai University of Medicine and Health Sciences, Shanghai 201318, China
| |
Collapse
|
20
|
Dercle L, Fronheiser M, Lu L, Du S, Hayes W, Leung DK, Roy A, Wilkerson J, Guo P, Fojo AT, Schwartz LH, Zhao B. Identification of Non–Small Cell Lung Cancer Sensitive to Systemic Cancer Therapies Using Radiomics. Clin Cancer Res 2020; 26:2151-2162. [DOI: 10.1158/1078-0432.ccr-19-2942] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2019] [Revised: 11/27/2019] [Accepted: 01/22/2020] [Indexed: 11/16/2022]
|
21
|
Lu X, Li M, Zhang H, Hua S, Meng F, Yang H, Li X, Cao D. A novel radiomic nomogram for predicting epidermal growth factor receptor mutation in peripheral lung adenocarcinoma. Phys Med Biol 2020; 65:055012. [PMID: 31978901 DOI: 10.1088/1361-6560/ab6f98] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
To predict the epidermal growth factor receptor (EGFR) mutation status in patients with lung adenocarcinoma using quantitative radiomic biomarkers and semantic features. We analyzed the computed tomography (CT) images and medical record data of 104 patients with lung adenocarcinoma who underwent surgical excision and EGFR mutation detection from 2016 to 2018 at our center. CT radiomic and semantic features that reflect the tumors' heterogeneity and phenotype were extracted from preoperative non-enhanced CT scans. The least absolute shrinkage and selection operator method was applied to select the most distinguishable features. Three logistic regression models were built to predict the EGFR mutation status by combining the CT semantic with clinicopathological characteristics, using the radiomic features alone, and by combining the radiomic and clinicopathological features. Receiver operating characteristic (ROC) curve analysis was performed using five-fold cross-validation and the mean area under the curve (AUC) values were calculated and compared between the models to obtain the optimal model for predicting EGFR mutation. Furthermore, radiomic nomograms were constructed to demonstrate the performance of the model. In total, 1025 radiomic features were extracted and reduced to 13 features as the most important predictors to build the radiomic signature. The combined radiomic and clinicopathological features model was developed based on the radiomic signature, sex, smoking, vascular infiltration, and pathohistological type. The AUC was 0.90 ± 0.02 for the training, 0.88 ± 0.11 for the verification, and 0.894 for the test dataset. This model was superior to the other prediction models that used the combined CT semantic and clinicopathological features (AUC for the test dataset: 0.768) and radiomic features alone (AUC for the test dataset: 0.837). The prediction model built by radiomic biomarkers and clinicopathological features, including the radiomic signature, sex, smoking, vascular infiltration, and pathological type, outperformed the other two models and could effectively predict the EGFR mutation status in patients with peripheral lung adenocarcinoma. The radiomic nomogram of this model is expected to become an effective biomarker for patients with lung adenocarcinoma requiring adjuvant targeted treatment.
Collapse
Affiliation(s)
- Xiaoqian Lu
- Department of Radiology, the First Hospital of Jilin University, 130021 Changchun, People's Republic of China. These authors contributed equally to this work
| | | | | | | | | | | | | | | |
Collapse
|
22
|
Nair JKR, Saeed UA, McDougall CC, Sabri A, Kovacina B, Raidu BVS, Khokhar RA, Probst S, Hirsh V, Chankowsky J, Van Kempen LC, Taylor J. Radiogenomic Models Using Machine Learning Techniques to Predict EGFR Mutations in Non-Small Cell Lung Cancer. Can Assoc Radiol J 2020; 72:109-119. [PMID: 32063026 DOI: 10.1177/0846537119899526] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND The purpose of this study was to build radiogenomics models from texture signatures derived from computed tomography (CT) and 18F-FDG PET-CT (FDG PET-CT) images of non-small cell lung cancer (NSCLC) with and without epidermal growth factor receptor (EGFR) mutations. METHODS Fifty patients diagnosed with NSCLC between 2011 and 2015 and with known EGFR mutation status were retrospectively identified. Texture features extracted from pretreatment CT and FDG PET-CT images by manual contouring of the primary tumor were used to develop multivariate logistic regression (LR) models to predict EGFR mutations in exon 19 and exon 20. RESULTS An LR model evaluating FDG PET-texture features was able to differentiate EGFR mutant from wild type with an area under the curve (AUC), sensitivity, specificity, and accuracy of 0.87, 0.76, 0.66, and 0.71, respectively. The model derived from CT texture features had an AUC, sensitivity, specificity, and accuracy of 0.83, 0.84, 0.73, and 0.78, respectively. FDG PET-texture features that could discriminate between mutations in EGFR exon 19 and 21 demonstrated AUC, sensitivity, specificity, and accuracy of 0.86, 0.84, 0.73, and 0.78, respectively. Based on CT texture features, the AUC, sensitivity, specificity, and accuracy were 0.75, 0.81, 0.69, and 0.75, respectively. CONCLUSION Non-small cell lung cancer texture analysis using FGD-PET and CT images can identify tumors with mutations in EGFR. Imaging signatures could be valuable for pretreatment assessment and prognosis in precision therapy.
Collapse
Affiliation(s)
- Jay Kumar Raghavan Nair
- Department of Radiology, 54473McGill University Health Centre, Montreal, Québec, Canada.,Department of Radiology, McMaster University Faculty of Health Sciences, Hamilton, Ontario, Canada.,Department of Radiology, 2129University of Calgary, Calgary, Alberta, Canada
| | - Umar Abid Saeed
- Department of Radiology, 54473McGill University Health Centre, Montreal, Québec, Canada.,Department of Radiology, 2129University of Calgary, Calgary, Alberta, Canada
| | - Connor C McDougall
- Department of Mechanical Engineering, 2129University of Calgary, Calgary, Alberta, Canada
| | - Ali Sabri
- Department of Radiology, McMaster University, Hamilton, Ontario, Canada.,Department of Radiology, Jewish General Hospital, Montreal, Québec, Canada
| | - Bojan Kovacina
- Department of Radiology, Jewish General Hospital, Montreal, Québec, Canada
| | - B V S Raidu
- Raidu Analysts and Associates, Mumbai, India
| | - Riaz Ahmed Khokhar
- Department of Radiology, 54473McGill University Health Centre, Montreal, Québec, Canada.,Department of Surgery, Khokhar Medical Centre, Rawalpindi, Pakistan
| | - Stephan Probst
- Department of Nuclear Medicine, Jewish General Hospital, Québec, Montreal, Canada
| | - Vera Hirsh
- Department of Oncology, 5620McGill University Health Centre, Montreal, Québec, Canada
| | - Jeffrey Chankowsky
- Department of Radiology, 54473McGill University Health Centre, Montreal, Québec, Canada
| | - Léon C Van Kempen
- Department of Pathology, 10173University Medical Center Groningen, University of Groningen, Groningen, the Netherlands.,Department of Pathology, Jewish General Hospital, Montreal, Québec, Canada
| | - Jana Taylor
- Department of Radiology, 54473McGill University Health Centre, Montreal, Québec, Canada
| |
Collapse
|
23
|
Yoon J, Suh YJ, Han K, Cho H, Lee HJ, Hur J, Choi BW. Utility of CT radiomics for prediction of PD-L1 expression in advanced lung adenocarcinomas. Thorac Cancer 2020; 11:993-1004. [PMID: 32043309 PMCID: PMC7113038 DOI: 10.1111/1759-7714.13352] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Revised: 01/20/2020] [Accepted: 01/21/2020] [Indexed: 12/23/2022] Open
Abstract
Background We aimed to assess if quantitative radiomic features can predict programmed death ligand 1 (PD‐L1) expression in advanced stage lung adenocarcinoma. Methods This retrospective study included 153 patients who had advanced stage (>IIIA by TNM classification) lung adenocarcinoma with pretreatment thin section computed tomography (CT) images and PD‐L1 expression test results in their pathology reports. Clinicopathological data were collected from electronic medical records. Visual analysis and radiomic feature extraction of the tumor from pretreatment CT were performed. We constructed two models for multivariate logistic regression analysis (one based on clinical variables, and the other based on a combination of clinical variables and radiomic features), and compared c‐statistics of the receiver operating characteristic curves of each model to identify the model with the higher predictability. Results Among 153 patients, 53 patients were classified as PD‐L1 positive and 100 patients as PD‐L1 negative. There was no significant difference in clinical characteristics or imaging findings on visual analysis between the two groups (P > 0.05 for all). Rad‐score by radiomic analysis was higher in the PD‐L1 positive group than in the PD‐L1 negative group with a statistical significance (−0.378 ± 1.537 vs. −1.171 ± 0.822, P = 0.0008). A prediction model that uses clinical variables and CT radiomic features showed higher performance compared to a prediction model that uses clinical variables only (c‐statistic = 0.646 vs. 0.550, P = 0.0299). Conclusions Quantitative CT radiomic features can predict PD‐L1 expression in advanced stage lung adenocarcinoma. A prediction model composed of clinical variables and CT radiomic features may facilitate noninvasive assessment of PD‐L1 expression. Key points Significant findings of the study Quantitative CT radiomic features can help predict PD‐L1 expression, whereas none of the qualitative imaging findings is associated with PD‐L1 positivity. What this study adds A prediction model composed of clinical variables and CT radiomic features may facilitate noninvasive assessment of PD‐L1 expression.
Collapse
Affiliation(s)
- Jiyoung Yoon
- Department of Radiology, Research Institute of Radiological Science, Severance Hospital, Yonsei University College of Medicine, Seoul, South Korea
| | - Young Joo Suh
- Department of Radiology, Research Institute of Radiological Science, Severance Hospital, Yonsei University College of Medicine, Seoul, South Korea
| | - Kyunghwa Han
- Department of Radiology, Research Institute of Radiological Science, Severance Hospital, Yonsei University College of Medicine, Seoul, South Korea
| | - Hyoun Cho
- Department of Radiology, Research Institute of Radiological Science, Severance Hospital, Yonsei University College of Medicine, Seoul, South Korea
| | - Hye-Jeong Lee
- Department of Radiology, Research Institute of Radiological Science, Severance Hospital, Yonsei University College of Medicine, Seoul, South Korea
| | - Jin Hur
- Department of Radiology, Research Institute of Radiological Science, Severance Hospital, Yonsei University College of Medicine, Seoul, South Korea
| | - Byoung Wook Choi
- Department of Radiology, Research Institute of Radiological Science, Severance Hospital, Yonsei University College of Medicine, Seoul, South Korea
| |
Collapse
|
24
|
Chen C, Guo X, Wang J, Guo W, Ma X, Xu J. The Diagnostic Value of Radiomics-Based Machine Learning in Predicting the Grade of Meningiomas Using Conventional Magnetic Resonance Imaging: A Preliminary Study. Front Oncol 2019; 9:1338. [PMID: 31867272 PMCID: PMC6908490 DOI: 10.3389/fonc.2019.01338] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Accepted: 11/15/2019] [Indexed: 02/05/2023] Open
Abstract
Objective: The purpose of the current study is to investigate whether texture analysis-based machine learning algorithms could help devise a non-invasive imaging biomarker for accurate classification of meningiomas using machine learning algorithms. Method: The study cohort was established from the hospital database by reviewing the medical records. Patients were selected if they underwent meningioma resection in the neurosurgery department between January 2015 and December 2018. A total number of 40 texture parameters were extracted from pretreatment postcontrast T1-weighted (T1C) images based on six matrixes. Three feature selection methods were adopted, namely, distance correlation, least absolute shrinkage and selection operator (LASSO), and gradient boosting decision tree (GBDT). Multiclass classification methods of linear discriminant analysis (LDA) and support vector machine (SVM) algorithms were employed to establish the classification models. The diagnostic performances of models were evaluated with confusion matrix based on which the areas under the curve, accuracy, and Kappa value of models were calculated. Result: Confusion matrix showed that the LDA-based models represented better diagnostic performances than SVM-based models. The highest accuracy among LDA-based models was 75.6%, shown in the combination of Lasso + LDA. The optimal models for SVM-based models was Lasso+SVM, with accuracy of 59.0% in the testing group. One of the SVM-based models, GBDT+SVM, was overfitting, suggesting that this model was not suitable for application. Conclusion: Machine learning algorithms with texture features extracted from T1C images could potentially serve as the assistant imaging biomarkers for presurgically grading meningiomas.
Collapse
Affiliation(s)
- Chaoyue Chen
- Department of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, China.,State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, China.,Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, China
| | - Xinyi Guo
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, China.,West China School of Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Jian Wang
- School of Computer Science, Nanjing University of Science and Technology, Nanjing, China
| | - Wen Guo
- Department of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, China.,State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, China.,Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, China.,West China School of Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Xuelei Ma
- Department of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, China.,State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, China
| | - Jianguo Xu
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
25
|
Value of pre-therapy 18F-FDG PET/CT radiomics in predicting EGFR mutation status in patients with non-small cell lung cancer. Eur J Nucl Med Mol Imaging 2019; 47:1137-1146. [PMID: 31728587 DOI: 10.1007/s00259-019-04592-1] [Citation(s) in RCA: 129] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Accepted: 10/22/2019] [Indexed: 12/25/2022]
Abstract
PURPOSE To assess the predictive power of pre-therapy 18F-FDG PET/CT-based radiomic features for epidermal growth factor receptor (EGFR) mutation status in non-small cell lung cancer. METHODS Two hundred and forty-eight lung cancer patients underwent pre-therapy diagnostic 18F-FDG PET/CT scans and were tested for genetic mutations. The LIFEx package was used to extract 47 PET and 45 CT radiomic features reflecting tumor heterogeneity and phenotype. The least absolute shrinkage and selection operator (LASSO) algorithm was used to select radiomic features and develop a radiomics signature. We compared the predictive performance of models established by radiomics signature, clinical variables, and their combinations using receiver operating curves (ROCs). In addition, a nomogram based on the radiomics signature score (rad-score) and clinical variables was developed. RESULTS The patients were divided into a training set (n = 175) and a validation set (n = 73). Ten radiomic features were selected to build the radiomics signature model. The model showed a significant ability to discriminate between EGFR mutation and EGFR wild type, with area under the ROC curve (AUC) equal to 0.79 in the training set, and 0.85 in the validation set, compared with 0.75 and 0.69 for the clinical model. When clinical variables and radiomics signature were combined, the AUC increased to 0.86 (95% CI [0.80-0.91]) in the training set and 0.87 (95% CI [0.79-0.95]) in the validation set, thus showing better performance in the prediction of EGFR mutations. CONCLUSION The PET/CT-based radiomic features showed good performance in predicting EGFR mutation in non-small cell lung cancer, providing a useful method for the choice of targeted therapy in a clinical setting.
Collapse
|
26
|
Deng Y, Soule E, Cui E, Samuel A, Shah S, Lall C, Sundaram C, Sandrasegaran K. Usefulness of CT texture analysis in differentiating benign and malignant renal tumours. Clin Radiol 2019; 75:108-115. [PMID: 31668402 DOI: 10.1016/j.crad.2019.09.131] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2019] [Accepted: 09/12/2019] [Indexed: 12/22/2022]
Abstract
AIM To elucidate visually imperceptible differences between benign and malignant renal tumours using computed tomography texture analysis (CTTA) using filtration histogram based parameters. MATERIALS AND METHODS A retrospective study was performed by texture analysis of pretreatment contrast-enhanced CT examinations in 354 histopathologically confirmed renal cell carcinomas (RCCs) and 147 benign renal tumours. A region-of-interest was drawn encompassing the largest cross-section of the tumour on venous phase axial CT. CTTA features of entropy, kurtosis, mean positive pixel density, and skewness at different spatial filters were calculated and compared in an attempt to differentiate benign lesions from malignancy. RESULTS Entropy with fine spatial filter was significantly higher in RCC than benign renal tumours (p=0.022). Entropy with fine and medium filters was higher in RCC than lipid-poor angiomyolipoma (p=0.050 and 0.052, respectively). Entropy >5.62 had high specificity of 85.7%, but low sensitivity of 31.3%, respectively, for predicting RCC. CONCLUSIONS Differences in entropy were helpful in differentiating RCC from lipid-poor angiomyolipoma, and chromophobe RCC from oncocytoma. This technique may be useful to differentiate lesions that appear equivocal on visual assessment or alter management in poor surgical candidates.
Collapse
Affiliation(s)
- Y Deng
- Department of Radiology, First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China; Department of Radiology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - E Soule
- Department of Radiology, University of Florida College of Medicine, Jacksonville, FL, USA
| | - E Cui
- Department of Radiology, Indiana University School of Medicine, Indianapolis, IN, USA; Department of Radiology, Jiangmen Central Hospital, Affiliated Jiangmen Hospital of Sun YAT-SEN University, Jiangmen, China
| | - A Samuel
- Department of Radiology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - S Shah
- Department of Radiology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - C Lall
- Department of Radiology, University of Florida College of Medicine, Jacksonville, FL, USA
| | - C Sundaram
- Department of Urology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - K Sandrasegaran
- Department of Radiology, Indiana University School of Medicine, Indianapolis, IN, USA; Department of Radiology, Mayo Clinic, Phoenix, AZ, USA.
| |
Collapse
|
27
|
Bogowicz M, Vuong D, Huellner MW, Pavic M, Andratschke N, Gabrys HS, Guckenberger M, Tanadini-Lang S. CT radiomics and PET radiomics: ready for clinical implementation? THE QUARTERLY JOURNAL OF NUCLEAR MEDICINE AND MOLECULAR IMAGING : OFFICIAL PUBLICATION OF THE ITALIAN ASSOCIATION OF NUCLEAR MEDICINE (AIMN) [AND] THE INTERNATIONAL ASSOCIATION OF RADIOPHARMACOLOGY (IAR), [AND] SECTION OF THE SOCIETY OF RADIOPHARMACEUTICAL CHEMISTRY AND BIOLOGY 2019; 63:355-370. [PMID: 31527578 DOI: 10.23736/s1824-4785.19.03192-3] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
INTRODUCTION Today, rapid technical and clinical developments result in an increasing number of treatment options for oncological diseases. Thus, decision support systems are needed to offer the right treatment to the right patient. Imaging biomarkers hold great promise in patient-individual treatment guidance. Routinely performed for diagnosis and staging, imaging datasets are expected to hold more information than used in the clinical practice. Radiomics describes the extraction of a large number of meaningful quantitative features from medical images, such as computed tomography (CT) and positron emission tomography (PET). Due to the non-invasive nature and ability to capture 3D image-based heterogeneity, radiomic features are potential surrogate markers of the cancer phenotype. Several radiomic studies are published per day, owing to encouraging results of many radiomics-based patient outcome models. Despite this comparably large number of studies, radiomics is mainly studied in proof of principle concept. Hence, a translation of radiomics from a hot topic research field into an essential clinical decision-making tool is lacking, but of high clinical interest. EVIDENCE ACQUISITION Herein, we present a literature review addressing the clinical evidence of CT and PET radiomics. An extensive literature review was conducted in PubMed, including papers on robustness and clinical applications. EVIDENCE SYNTHESIS We summarize image-modality related influences on the robustness of radiomic features and provide an overview of clinical evidence reported in the literature. Today, more evidence has been provided for CT imaging, however, PET imaging offers the promise of direct imaging of biological processes and functions. We provide a summary of future research directions, which needs to be addressed in order to successfully introduce radiomics into clinical medicine. In comparison to CT, more focus should be directed towards harmonization of PET acquisition and reconstruction protocols, which is important for transferable modelling. CONCLUSIONS Both CT and PET radiomics are promising pre-treatment and intra-treatment biomarkers for outcome prediction. Most studies are performed in retrospective setting, however their validation in prospective data collections is ongoing.
Collapse
Affiliation(s)
- Marta Bogowicz
- Department of Radiation Oncology, University Hospital Zurich and University of Zurich, Zurich, Switzerland -
| | - Diem Vuong
- Department of Radiation Oncology, University Hospital Zurich and University of Zurich, Zurich, Switzerland
| | - Martin W Huellner
- Department of Nuclear Medicine, University Hospital Zurich and University of Zurich, Zurich, Switzerland
| | - Matea Pavic
- Department of Radiation Oncology, University Hospital Zurich and University of Zurich, Zurich, Switzerland
| | - Nicolaus Andratschke
- Department of Radiation Oncology, University Hospital Zurich and University of Zurich, Zurich, Switzerland
| | - Hubert S Gabrys
- Department of Radiation Oncology, University Hospital Zurich and University of Zurich, Zurich, Switzerland
| | - Matthias Guckenberger
- Department of Radiation Oncology, University Hospital Zurich and University of Zurich, Zurich, Switzerland
| | - Stephanie Tanadini-Lang
- Department of Radiation Oncology, University Hospital Zurich and University of Zurich, Zurich, Switzerland
| |
Collapse
|
28
|
Association of radiomic features with epidermal growth factor receptor mutation status in non-small cell lung cancer and survival treated with tyrosine kinase inhibitors. Nucl Med Commun 2019; 40:1091-1098. [PMID: 31469811 DOI: 10.1097/mnm.0000000000001076] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Since the discovery of the fact that tyrosine kinase inhibitors could improve progression-free survival for patients with advanced non-small cell lung cancer compared with traditional chemotherapy, it has been extremely important to identify epidermal growth factor receptor mutation status in treatment stratification. Although lack of sufficient biopsy samples limit the precise detection of epidermal growth factor receptor mutation status in clinical practice, and it is difficult to identify the sensitive patients who confer favorable response to tyrosine kinase inhibitors. An increasing number of scholars tried to deal with these problems using methods based on the non-invasive imaging including computed tomography and PET to find the association with epidermal growth factor receptor mutation status and survival treated with tyrosine kinase inhibitor in non-small cell lung cancer. Although the conclusions have not reached a consensus, quantitative and high-throughput radiomics have brought us a new direction and might successfully help identify patients undergoing tyrosine kinase inhibitors who could get significant benefits.
Collapse
|
29
|
Shen TX, Liu L, Li WH, Fu P, Xu K, Jiang YQ, Pan F, Guo Y, Zhang MC. CT imaging-based histogram features for prediction of EGFR mutation status of bone metastases in patients with primary lung adenocarcinoma. Cancer Imaging 2019; 19:34. [PMID: 31174617 PMCID: PMC6556025 DOI: 10.1186/s40644-019-0221-9] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2018] [Accepted: 05/26/2019] [Indexed: 12/13/2022] Open
Abstract
OBJECTIVE To identify imaging markers that reflect the epidermal growth factor receptor (EGFR) mutation status by comparing computed tomography (CT) imaging-based histogram features between bone metastases with and without EGFR mutation in patients with primary lung adenocarcinoma. MATERIALS AND METHODS This retrospective study included 57 patients, with pathologically confirmed bone metastasis of primary lung adenocarcinoma. EGFR mutation status of bone metastases was confirmed by gene detection. The CT imaging of the metastatic bone lesions which were obtained between June 2014 and December 2017 were collected and analyzed. A total of 42 CT imaging-based histogram features were automatically extracted. Feature selection was conducted using Student's t-test, Mann-Whitney U test, single-factor logistic regression analysis and Spearman correlation analysis. A receiver operating characteristic (ROC) curve was plotted to compare the effectiveness of features in distinguishing between EGFR(+) and EGFR(-) groups. DeLong's test was used to analyze the differences between the area under the curve (AUC) values. RESULTS Three histogram features, namely range, skewness, and quantile 0.975 were significantly associated with EGFR mutation status. After combining these three features and combining range and skewness, we obtained the same AUC values, sensitivity and specificity. Meanwhile, the highest AUC value was achieved (AUC 0.783), which also had a higher sensitivity (0.708) and specificity (0.788). The differences between AUC values of the three features and their various combinations were statistically insignificant. CONCLUSION CT imaging-based histogram features of bone metastases with and without EGFR mutation in patients with primary lung adenocarcinoma were identified, and they may contribute to diagnosis and prediction of EGFR mutation status.
Collapse
Affiliation(s)
- Tong-Xu Shen
- Department of Radiology, China-Japan Union Hospital of Jilin University, NO.126 Xiantai Street, Changchun, 130033, Jilin, China
| | - Lin Liu
- Department of Radiology, China-Japan Union Hospital of Jilin University, NO.126 Xiantai Street, Changchun, 130033, Jilin, China
| | - Wen-Hui Li
- College of Computer Science and Technology, Jilin University, NO.2699 Qianjin Street, Changchun, 130012, Jilin, China
| | - Ping Fu
- Department of Ultrasound, China-Japan Union Hospital of Jilin University, NO.126 Xiantai Street, Changchun, 130033, Jilin, China
| | - Kai Xu
- Department of Radiology, China-Japan Union Hospital of Jilin University, NO.126 Xiantai Street, Changchun, 130033, Jilin, China
| | - Yu-Qing Jiang
- Department of Radiology, China-Japan Union Hospital of Jilin University, NO.126 Xiantai Street, Changchun, 130033, Jilin, China
| | - Feng Pan
- Department of Radiology, China-Japan Union Hospital of Jilin University, NO.126 Xiantai Street, Changchun, 130033, Jilin, China
| | - Yan Guo
- GE Healthcare, China, NO.69 Heping North Street, Shenyang, 110000, Liaoning, China
| | - Meng-Chao Zhang
- Department of Radiology, China-Japan Union Hospital of Jilin University, NO.126 Xiantai Street, Changchun, 130033, Jilin, China.
| |
Collapse
|
30
|
Bodalal Z, Trebeschi S, Nguyen-Kim TDL, Schats W, Beets-Tan R. Radiogenomics: bridging imaging and genomics. Abdom Radiol (NY) 2019; 44:1960-1984. [PMID: 31049614 DOI: 10.1007/s00261-019-02028-w] [Citation(s) in RCA: 184] [Impact Index Per Article: 30.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
From diagnostics to prognosis to response prediction, new applications for radiomics are rapidly being developed. One of the fastest evolving branches involves linking imaging phenotypes to the tumor genetic profile, a field commonly referred to as "radiogenomics." In this review, a general outline of radiogenomic literature concerning prominent mutations across different tumor sites will be provided. The field of radiogenomics originates from image processing techniques developed decades ago; however, many technical and clinical challenges still need to be addressed. Nevertheless, increasingly accurate and robust radiogenomic models are being presented and the future appears to be bright.
Collapse
|
31
|
Erdal BS, Prevedello LM, Qian S, Demirer M, Little K, Ryu J, O'Donnell T, White RD. Radiology and Enterprise Medical Imaging Extensions (REMIX). J Digit Imaging 2019; 31:91-106. [PMID: 28840365 PMCID: PMC5788816 DOI: 10.1007/s10278-017-0010-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Radiology and Enterprise Medical Imaging Extensions (REMIX) is a platform originally designed to both support the medical imaging-driven clinical and clinical research operational needs of Department of Radiology of The Ohio State University Wexner Medical Center. REMIX accommodates the storage and handling of “big imaging data,” as needed for large multi-disciplinary cancer-focused programs. The evolving REMIX platform contains an array of integrated tools/software packages for the following: (1) server and storage management; (2) image reconstruction; (3) digital pathology; (4) de-identification; (5) business intelligence; (6) texture analysis; and (7) artificial intelligence. These capabilities, along with documentation and guidance, explaining how to interact with a commercial system (e.g., PACS, EHR, commercial database) that currently exists in clinical environments, are to be made freely available.
Collapse
Affiliation(s)
- Barbaros S Erdal
- Radiology Department, The Ohio State University Wexner Medical Center, 395 W 12th Ave, Columbus, OH, 43210, USA.
| | - Luciano M Prevedello
- Radiology Department, The Ohio State University Wexner Medical Center, 395 W 12th Ave, Columbus, OH, 43210, USA
| | - Songyue Qian
- Radiology Department, The Ohio State University Wexner Medical Center, 395 W 12th Ave, Columbus, OH, 43210, USA
| | - Mutlu Demirer
- Radiology Department, The Ohio State University Wexner Medical Center, 395 W 12th Ave, Columbus, OH, 43210, USA
| | - Kevin Little
- Radiology Department, The Ohio State University Wexner Medical Center, 395 W 12th Ave, Columbus, OH, 43210, USA
| | - John Ryu
- Radiology Department, The Ohio State University Wexner Medical Center, 395 W 12th Ave, Columbus, OH, 43210, USA
| | - Thomas O'Donnell
- Siemens Medical Solutions USA, Inc, 40 Liberty Boulevard, Malvern, PA, 19355, USA
| | - Richard D White
- Radiology Department, The Ohio State University Wexner Medical Center, 395 W 12th Ave, Columbus, OH, 43210, USA
| |
Collapse
|
32
|
Schofield R, Ganeshan B, Fontana M, Nasis A, Castelletti S, Rosmini S, Treibel TA, Manisty C, Endozo R, Groves A, Moon JC. Texture analysis of cardiovascular magnetic resonance cine images differentiates aetiologies of left ventricular hypertrophy. Clin Radiol 2019; 74:140-149. [PMID: 30527518 DOI: 10.1016/j.crad.2018.09.016] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Accepted: 09/26/2018] [Indexed: 10/28/2022]
Abstract
AIM To investigate whether unenhanced cardiovascular magnetic resonance (CMR) balanced steady state free precession (bSSFP) cine images could be analysed using textural analysis (TA) software to differentiate different aetiologies of disease causing increased myocardial wall thickness (left ventricular hypertrophy [LVH]) and indicate the severity of myocardial tissue abnormality. MATERIALS AND METHODS A mid short axis unenhanced cine frame of 216 patients comprising 50 cases of hypertrophic cardiomyopathy (HCM; predominantly Left ventricular outflow tract obstruction [LVOTO] subtype), 52 cases of cardiac amyloid (CA; predominantly AL: light chain subtype), 68 cases of aortic stenosis (AS), 15 hypertensive patients with LVH (HTN+LVH), and 31 healthy volunteers (HV) underwent TA of the CMR cine images (CMRTA) using TexRAD (TexRAD Ltd, Cambridge, UK). Among the HV, 16/31 were scanned twice to form a test-retest reproducibility cohort. CMRTA comprised a filtration-histogram technique to extract and quantify features using six parameters. RESULTS Test-retest analysis in the HV showed a medium filter (3 mm) was the most reproducible (intra-class correlation of 0.9 for kurtosis and skewness and 0.8 for mean and SD). Disease cohorts were statistically different (p<0.001) to HV for all parameters. Pairwise comparisons of CMRTA parameters showed kurtosis and skewness was consistently significant in ranking the degree of difference from HV (greatest to least): CA, HCM, LVH+HTN, AS (p<0.001). Similarly, mean, standard deviation, entropy, and mean positive pixel (MPP) were consistent in ranking degree of difference from HV: HCM, CA, AS and HTN+LVH. CONCLUSION Radiomic features of bSSFP CMR data sets derived using TA show promise in discriminating between the aetiologies of LVH.
Collapse
Affiliation(s)
- R Schofield
- Bart's Heart Centre, London, UK; Institute of Cardiovascular Science, University College London, UK.
| | - B Ganeshan
- Institute of Nuclear Medicine, University College London, UK
| | - M Fontana
- National Amyloid Centre, Royal Free Hospital, London, UK
| | - A Nasis
- Monash Cardiovascular Research Centre, Monash University Department of Medicine (MMC), Melbourne, Australia
| | | | | | - T A Treibel
- Bart's Heart Centre, London, UK; Institute of Cardiovascular Science, University College London, UK
| | - C Manisty
- Bart's Heart Centre, London, UK; Institute of Cardiovascular Science, University College London, UK
| | - R Endozo
- Institute of Nuclear Medicine, University College London, UK
| | - A Groves
- Institute of Nuclear Medicine, University College London, UK
| | - J C Moon
- Bart's Heart Centre, London, UK; Institute of Cardiovascular Science, University College London, UK
| |
Collapse
|
33
|
Li Y, Lu L, Xiao M, Dercle L, Huang Y, Zhang Z, Schwartz LH, Li D, Zhao B. CT Slice Thickness and Convolution Kernel Affect Performance of a Radiomic Model for Predicting EGFR Status in Non-Small Cell Lung Cancer: A Preliminary Study. Sci Rep 2018; 8:17913. [PMID: 30559455 PMCID: PMC6297245 DOI: 10.1038/s41598-018-36421-0] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Accepted: 11/20/2018] [Indexed: 12/25/2022] Open
Abstract
We evaluated whether the optimal selection of CT reconstruction settings enables the construction of a radiomics model to predict epidermal growth factor receptor (EGFR) mutation status in primary lung adenocarcinoma (LAC) using standard of care CT images. Fifty-one patients (EGFR:wildtype = 23:28) with LACs of clinical stage I/II/IIIA were included in the analysis. The LACs were segmented in four conditions, two slice thicknesses (Thin: 1 mm; Thick: 5 mm) and two convolution kernels (Sharp: B70f/B70s; Smooth: B30f/B31f/B31s), which constituted four groups: (1) Thin-Sharp, (2) Thin-Smooth, (3) Thick-Sharp, and (4) Thick-Smooth. Machine learning algorithms selected and combined 1,695 quantitative image features to build prediction models. The performance of prediction models was assessed by calculating the area under the curve (AUC). The best prediction model yielded AUC (95%CI) = 0.83 (0.68, 0.92) using the Thin-Smooth reconstruction setting. The AUC of models using thick slices was significantly lower than that of thin slices (P < 10-3), whereas the impact of reconstruction kernel was not significant. Our study showed that the optimal prediction of EGFR mutational status in early stage LACs was achieved by using thin CT-scan slices, independently of convolution kernels. Results from the prediction model suggest that tumor heterogeneity is associated with EGFR mutation.
Collapse
Affiliation(s)
- Yajun Li
- Department of Radiology, the Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China.
| | - Lin Lu
- Department of Radiology, Columbia University Medical Center, New York, NY, 10039, USA
| | - Manjun Xiao
- Department of Radiology, the Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China
| | - Laurent Dercle
- Department of Radiology, Columbia University Medical Center, New York, NY, 10039, USA
- Gustave Roussy, Université Paris-Saclay, Inserm, UMR1015, Paris, France
| | - Yue Huang
- Department of Surgery, University of Missouri, Columbia, MO, USA
| | - Zishu Zhang
- Department of Radiology, the Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China
| | - Lawrence H Schwartz
- Department of Radiology, Columbia University Medical Center, New York, NY, 10039, USA
| | - Daiqiang Li
- Department of Pathology, the Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China
| | - Binsheng Zhao
- Department of Radiology, Columbia University Medical Center, New York, NY, 10039, USA
| |
Collapse
|
34
|
Mei D, Luo Y, Wang Y, Gong J. CT texture analysis of lung adenocarcinoma: can Radiomic features be surrogate biomarkers for EGFR mutation statuses. Cancer Imaging 2018; 18:52. [PMID: 30547844 PMCID: PMC6295009 DOI: 10.1186/s40644-018-0184-2] [Citation(s) in RCA: 64] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Accepted: 12/03/2018] [Indexed: 12/03/2022] Open
Abstract
Objective To investigate whether radiomic features can be surrogate biomarkers for epidermal growth factor receptor (EGFR) mutation statuses. Materials and methods Two hundred ninety six consecutive patients, who underwent CT examinations before operation within 3 months and had EGFR mutations tested, were enrolled in this retrospective study. CT texture features were extracted using an open-source software with whole volume segmentation. The association between CT texture features and EGFR mutation statuses were analyzed. Results In the 296 patients, there were 151 patients with EGFR mutations (51%). Logistic analysis identified that lower age (Odds Ratio[OR]: 0.968,95% confidence interval [CI]:0.946~0.990, p = 0.005) and a radiomic feature named GreyLevelNonuniformityNormalized (OR: 0.012, 95% CI:0.000~0.352, p = 0.01) were predictors for exon 19 mutation; higher age (OR: 1.027, 95%CI:1.003~1.052,p = 0.025), female sex (OR: 2.189, 95%CI:1.264~3.791, p = 0.005) and a radiomic feature named Maximum2DDiameterColumn (OR: 0.968, 95%CI:0.946~0.990], p = 0.005) for exon 21 mutation; and female sex (OR: 1.883,95%CI:1.064~3.329, p = 0.030), non-smoking status (OR: 2.070, 95%CI:1.090~3.929, p = 0.026) and a radiomic feature termed SizeZone NonUniformityNormalized (OR: 0.010, 95% CI:0.0001~0.852, p = 0.042) for EGFR mutations. Areas under the curve (AUCs) of combination with clinical and radiomic features to predict exon 19 mutation, exon 21 mutation and EGFR mutations were 0.655, 0.675 and 0.664, respectively. Conclusion Several radiomic features are associated with EGFR mutation statuses of lung adenocarcinoma. Combination with clinical files, moderate diagnostic performance can be obtained to predict EGFR mutation status of lung adenocarcinoma. Radiomic features might harbor potential surrogate biomarkers for identification of EGRF mutation statuses.
Collapse
Affiliation(s)
- Dongdong Mei
- Department of Radiology, Shenzhen People's Hospital, the Second Clinical Medical College, Jinan University, Shenzhen, 518020, Guangdong, China
| | - Yan Luo
- Department of Radiology, Shenzhen People's Hospital, the Second Clinical Medical College, Jinan University, Shenzhen, 518020, Guangdong, China
| | - Yan Wang
- Department of Radiology and Biomedical Imaging, University of California San Francisco, 185 Berry St, Suite 350, San Francisco, CA, 94107, USA
| | - Jingshan Gong
- Department of Radiology, Shenzhen People's Hospital, the Second Clinical Medical College, Jinan University, Shenzhen, 518020, Guangdong, China.
| |
Collapse
|
35
|
Rizzo S, Savoldi F, Rossi D, Bellomi M. Radiogenomics as association between non-invasive imaging features and molecular genomics of lung cancer. ANNALS OF TRANSLATIONAL MEDICINE 2018; 6:447. [PMID: 30603635 DOI: 10.21037/atm.2018.11.17] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
- Stefania Rizzo
- Division of Radiology, IEO, European Institute of Oncology IRCCS, Milan, IT, Italy
| | - Filippo Savoldi
- Postgraduate School in Radiodiagnostics, Università degli Studi di Milano, Milan, IT, Italy
| | - Duccio Rossi
- Postgraduate School in Radiodiagnostics, Università degli Studi di Milano, Milan, IT, Italy
| | - Massimo Bellomi
- Division of Radiology, IEO, European Institute of Oncology IRCCS, Milan, IT, Italy.,Department of Oncology and Hemato-Oncology, Università degli Studi di Milano, Milan, IT, Italy
| |
Collapse
|
36
|
Ahn SJ, Kim JH, Lee SM, Park SJ, Han JK. CT reconstruction algorithms affect histogram and texture analysis: evidence for liver parenchyma, focal solid liver lesions, and renal cysts. Eur Radiol 2018; 29:4008-4015. [PMID: 30456584 DOI: 10.1007/s00330-018-5829-9] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Revised: 09/20/2018] [Accepted: 10/12/2018] [Indexed: 01/17/2023]
Abstract
PURPOSE To determine the effects of different reconstruction algorithms on histogram and texture features in different targets. MATERIALS AND METHODS Among 3620 patients, 480 had normal liver parenchyma, 494 had focal solid liver lesions (metastases = 259; hepatocellular carcinoma = 99; hemangioma = 78; abscess = 32; and cholangiocarcinoma = 26), and 488 had renal cysts. CT images were reconstructed with filtered back-projection (FBP), hybrid iterative reconstruction (HIR), and iterative model reconstruction (IMR) algorithms. Computerized histogram and texture analyses were performed by extracting 11 features. RESULTS Different reconstruction algorithms had distinct, significant effects. IMR had a greater effect than HIR. For instance, IMR had a significant effect on five features of liver parenchyma, nine features of focal liver lesions, and four features of renal cysts on portal-phase scans and four, eight, and four features, respectively, on precontrast scans (p < 0.05). Meanwhile, different algorithms had a greater effect on focal liver lesions (six in HIR and nine in IMR on portal-phase, three in HIR, and eight in IMR on precontrast scans) than on liver parenchyma or cysts. The mean attenuation and standard deviation were not affected by the reconstruction algorithm (p > .05). Most parameters showed good or excellent intra- and interobserver agreement, with intraclass correlation coefficients ranging from 0.634 to 0.972. CONCLUSIONS Different reconstruction algorithms affect histogram and texture features. Reconstruction algorithms showed stronger effects in focal liver lesions than in liver parenchyma or renal cysts. KEY POINTS • Imaging heterogeneities influenced the quantification of image features. • Different reconstruction algorithms had a significant effect on histogram and texture features. • Solid liver lesions were more affected than liver parenchyma or cysts.
Collapse
Affiliation(s)
- Su Joa Ahn
- Department of Radiology, Seoul National University Hospital, 101 Daehangno, Jongno-gu, Seoul, 110-744, Republic of Korea
| | - Jung Hoon Kim
- Department of Radiology, Seoul National University Hospital, 101 Daehangno, Jongno-gu, Seoul, 110-744, Republic of Korea. .,Department of Radiology and Institute of Radiation Medicine, Seoul National University College of Medicine, 101 Daehang-no, Chongno-gu, Seoul, 110-744, Korea.
| | - Sang Min Lee
- Department of Radiology, Hallym University Sacred Heart Hospital, 22, Gwanpyeong-ro 170beon-gil, Anyang-si, 431-796, Korea
| | - Sang Joon Park
- Department of Radiology, Seoul National University Hospital, 101 Daehangno, Jongno-gu, Seoul, 110-744, Republic of Korea
| | - Joon Koo Han
- Department of Radiology, Seoul National University Hospital, 101 Daehangno, Jongno-gu, Seoul, 110-744, Republic of Korea.,Department of Radiology and Institute of Radiation Medicine, Seoul National University College of Medicine, 101 Daehang-no, Chongno-gu, Seoul, 110-744, Korea
| |
Collapse
|
37
|
Baliyan V, Kordbacheh H, Parameswaran B, Ganeshan B, Sahani D, Kambadakone A. Virtual monoenergetic imaging in rapid kVp-switching dual-energy CT (DECT) of the abdomen: impact on CT texture analysis. Abdom Radiol (NY) 2018; 43:2693-2701. [PMID: 29541830 DOI: 10.1007/s00261-018-1527-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
PURPOSE To study the impact of keV levels of virtual monoenergetic images generated from rapid kVp-switching dual-energy CT (rsDECT) on CT texture analysis (CTTA). METHODS This study included 30 consecutive patients (59.3 ± 12 years; range 34-77 years; 17M:13F) who underwent portal venous phase abdominal CT on a rsDECT scanner. Axial 5-mm monoenergetic images at 5 energy levels (40/50/60/70/80 keV) were created and CTTA of liver was performed. CTTA comprised a filtration-histogram technique with different spatial scale filter (SSF) values (0-6). CTTA quantification at each SSF value included histogram-based statistical parameters such as mean intensity, standard deviation (SD), entropy, mean of positive pixels (MPP), skewness, and kurtosis. The values were compared using repeated measures ANOVA. RESULTS Among the different CTTA metrics, mean intensity (at SSF > 0), skewness, and kurtosis did not show variability whereas entropy, MPP, and SD varied with different keV levels. There was no change in skewness and kurtosis values for all 6 filters (p > 0.05). Mean intensity showed no change for filters 2-6 (p > 0.05). Mean intensity at SSF = 0 i.e., mean attenuations were 91.2 ± 2.9, 108.7 ± 3.6, 136.1 ± 4.7, 179.8 ± 6.9, and 250.5 ± 10.1 HU for 80, 70, 60, 50, and 40 keV images, respectively demonstrating significant variability (decrease) with increasing keV levels (p < 0.001). Entropy, MPP, and SD values showed a statistically significant decrease with increasing keV of monoenergetic images on all 6 filters (p < 0.001). CONCLUSION The energy levels of monoenergetic images have variable impact on the different CTTA parameters, with no significant change in skewness, kurtosis, and filtered mean intensity whereas significant decrease in mean attenuation, entropy, MPP, and SD values with increasing energy levels.
Collapse
Affiliation(s)
- Vinit Baliyan
- Department of Radiology, Massachusetts General Hospital, White 270, 55 Fruit Street, Boston, MA, 02114, USA
| | - Hamed Kordbacheh
- Department of Radiology, Massachusetts General Hospital, White 270, 55 Fruit Street, Boston, MA, 02114, USA
| | | | - Balaji Ganeshan
- Institute of Nuclear Medicine, University College London, London, UK
| | - Dushyant Sahani
- Department of Radiology, Massachusetts General Hospital, White 270, 55 Fruit Street, Boston, MA, 02114, USA
| | - Avinash Kambadakone
- Department of Radiology, Massachusetts General Hospital, White 270, 55 Fruit Street, Boston, MA, 02114, USA.
| |
Collapse
|
38
|
Liu S, Shi H, Ji C, Guan W, Chen L, Sun Y, Tang L, Guan Y, Li W, Ge Y, He J, Liu S, Zhou Z. CT textural analysis of gastric cancer: correlations with immunohistochemical biomarkers. Sci Rep 2018; 8:11844. [PMID: 30087428 PMCID: PMC6081398 DOI: 10.1038/s41598-018-30352-6] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Accepted: 07/27/2018] [Indexed: 02/06/2023] Open
Abstract
To investigate the ability of CT texture analysis to assess and predict the expression statuses of E-cadherin, Ki67, VEGFR2 and EGFR in gastric cancers, the enhanced CT images of 139 patients with gastric cancer were retrospectively reviewed. The region of interest was manually drawn along the margin of the lesion on the largest slice in the arterial and venous phases, which yielded a series of texture parameters. Our results showed that the standard deviation, width, entropy, entropy (H), correlation and contrast from the arterial and venous phases were significantly correlated with the E-cadherin expression level in gastric cancers (all P < 0.05). The skewness from the arterial phase and the mean and autocorrelation from the venous phase were negatively correlated with the Ki67 expression level in gastric cancers (all P < 0.05). The width, entropy and contrast from the venous phase were positively correlated with the VEGFR2 expression level in gastric cancers (all P < 0.05). No significant correlation was found between the texture features and EGFR expression level. CT texture analysis, which had areas under the receiver operating characteristic curve (AUCs) ranging from 0.612 to 0.715, holds promise in predicting E-cadherin, Ki67 and VEGFR2 expression levels in gastric cancers.
Collapse
Affiliation(s)
- Shunli Liu
- Department of Radiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, 210008, China
| | - Hua Shi
- Department of Radiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, 210008, China
| | - Changfeng Ji
- Department of Radiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, 210008, China
| | - Wenxian Guan
- Department of Gastrointestinal Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, 210008, China
| | - Ling Chen
- Department of Pathology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, 210008, China
| | - Yingshi Sun
- Department of Radiology, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Lei Tang
- Department of Radiology, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Yue Guan
- School of Electronic Science and Engineering, Nanjing University, Nanjing, 210046, China
| | - Weifeng Li
- School of Electronic Science and Engineering, Nanjing University, Nanjing, 210046, China
| | - Yun Ge
- School of Electronic Science and Engineering, Nanjing University, Nanjing, 210046, China
| | - Jian He
- Department of Radiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, 210008, China.
| | - Song Liu
- Department of Radiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, 210008, China.
| | - Zhengyang Zhou
- Department of Radiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, 210008, China.
| |
Collapse
|
39
|
CT texture analysis can be a potential tool to differentiate gastrointestinal stromal tumors without KIT exon 11 mutation. Eur J Radiol 2018; 107:90-97. [PMID: 30292279 DOI: 10.1016/j.ejrad.2018.07.025] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Revised: 07/23/2018] [Accepted: 07/25/2018] [Indexed: 02/06/2023]
Abstract
OBJECTIVE To evaluate CT texture analysis as a tool to differentiate gastrointestinal stromal tumors (GISTs) without KIT exon 11 mutation. MATERIALS AND METHODS This study consisted of a study group of 69 GISTs and a validation group of 17 GISTs. Clinical information of the patients were collected and analyzed. Two-dimensional and three-dimensional texture analysis was performed. The textural parameters were evaluated in the study group and were validated in the validation group. The repeatability of the textural parameters on the single region of interest (single-ROI), double-ROI, and whole volume of interest (whole-VOI) was analyzed. The independent predictor for the GIST genotypes was analyzed with logistic regression models. The support vector machine (SVM) classifiers were also trained and 6-fold cross validation ROC curves were computed. Subjective heterogeneity scores of each lesion on enhanced CT images were given by radiologists and the corresponding difference of the heterogeneity rating was evaluated. RESULTS The non-gastric location, lower CD34_stain level and higher textural parameter standard Deviation (stdDeviation) were associated with the GISTs without KIT exon 11 mutation in the study group. The cross validation SVM classifiers achieved with combination of stdDeviation, anatomic location and CD34_stain level demonstrated medium to good prediction efficiency (AUC = 0.864-0.904) regarding the GIST genotypes. The stdDeviation was an independent predictor of GISTs without KIT exon 11 mutation, and had a medium correlation with the GIST genotypes in the study group (AUC = 0.726-0.750). The stdDeviation showed good performance (AUC = 0.904-0.962) when validated in the validation group. The double-ROIs improved the performances of single-ROIs, decreasing the variances of single-ROIs brought by section-selection, and demonstrating excellent agreements between ROIs and whole-VOI. Subjective heterogeneity scores had no statistically significant differences between GIST genotypes. CONCLUSION CT texture analysis can potentially help to differentiate GISTs without KIT exon 11 mutation from those GISTs with KIT exon 11 mutation on enhanced CT images.
Collapse
|
40
|
Suh YJ, Lee HJ, Kim YJ, Kim KG, Kim H, Jeon YK, Kim YT. Computed tomography characteristics of lung adenocarcinomas with epidermal growth factor receptor mutation: A propensity score matching study. Lung Cancer 2018; 123:52-59. [PMID: 30089595 DOI: 10.1016/j.lungcan.2018.06.030] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Revised: 06/13/2018] [Accepted: 06/27/2018] [Indexed: 02/04/2023]
Abstract
OBJECTIVES We investigated the relationship between computed tomography (CT) characteristics and epidermal growth factor receptor (EGFR) mutations in a large Asian cohort who received surgical resection of invasive lung adenocarcinoma. MATERIALS AND METHODS We retrospectively included 864 patients (524 with EGFR mutation and 340 with EGFR wild-type) who received surgical resections for invasive lung adenocarcinomas. After applying propensity score matching, 312 patients with mutated EGFR were matched with 312 patients with wild-type EGFR. CT characteristics, predominant histologic subtype, and CT measurement parameters (volume and estimated diameter of the total tumor and inner solid portion and ground-glass opacity [GGO] proportion) were compared within matched pairs. RESULTS Tumors in the EGFR mutation group showed higher proportions of pure ground-glass nodules (4.1% vs 1.3%), GGO-predominant (23.7% vs 14.7%), and solid-predominant part-solid nodules (37.2% vs 31.7%) CT characteristics, whereas EGFR wild-type tumors predominantly presented as pure solid nodules (34.6% vs 52.2%, P < 0.0001). EGFR mutation tumors more frequently had a lepidic-predominant subtype than did EGFR wild-type tumors (20.2% and 11.9%; P < 0.0001), and showed a smaller whole tumor size and solid portion (P < 0.0001) with a higher GGO proportion (P < 0.0001). Tumors with exon 21 missense mutations showed the highest GGO proportion and the smallest inner solid portion size, followed by tumors harboring an exon 19 deletion, compared with EGFR wild-type tumors (posthoc P < 0.01). CONCLUSION Adenocarcinomas with EGFR mutations had a higher GGO proportion than those with wild-type EGFR after matching of clinical variables. Lesions with an exon 21 mutation had a higher GGO proportion than lesions with other mutations.
Collapse
Affiliation(s)
- Young Joo Suh
- Department of Radiology, Seoul National University Hospital, 101 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea; Department of Radiology, Seoul National University College of Medicine, 103 Daehak-ro, Jongnogu, Seoul, 03080, Republic of Korea; Department of Radiology, Severance Hospital, Yonsei University College of Medicine, Republic of Korea
| | - Hyun-Ju Lee
- Department of Radiology, Seoul National University Hospital, 101 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea; Department of Radiology, Seoul National University College of Medicine, 103 Daehak-ro, Jongnogu, Seoul, 03080, Republic of Korea.
| | - Young Jae Kim
- Department of Biomedical Engineering, Gachon University College of Medicine, Incheon, Republic of Korea
| | - Kwang Gi Kim
- Department of Biomedical Engineering, Gachon University College of Medicine, Incheon, Republic of Korea
| | - Heekyung Kim
- Department of Radiology, Seoul National University Hospital, 101 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea; Department of Radiology, Seoul National University College of Medicine, 103 Daehak-ro, Jongnogu, Seoul, 03080, Republic of Korea
| | - Yoon Kyung Jeon
- Department of Pathology, Seoul National University Hospital, Seoul, Republic of Korea
| | - Young Tae Kim
- Department of Thoracic and Cardiovascular Surgery, Seoul National University Hospital, Cancer Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
41
|
Lee SW, Park H, Lee HY, Sohn I, Lee SH, Kang J, Sun JM, Ahn MJ. Deciphering Clinicoradiologic Phenotype for Thymidylate Synthase Expression Status in Patients with Advanced Lung Adenocarcinoma Using a Radiomics Approach. Sci Rep 2018; 8:8968. [PMID: 29895834 PMCID: PMC5997691 DOI: 10.1038/s41598-018-27273-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2017] [Accepted: 05/31/2018] [Indexed: 01/08/2023] Open
Abstract
We aimed to identify predictive clinicoradiologic characteristics of thymidylate synthase (TS) expression status in advanced non-squamous non-small cell lung cancer patients. We reviewed clinicoradiologic features of 169 patients stratified into TS-negative (n = 84) and TS-positive (n = 85) groups, including quantitative CT radiomic features of both primary lung and metastatic lesions from initial CT and PET. Clinical factors including age and smoking history were significantly associated with TS as well as radiomic features. The predictive performance for dichotomizing TS expression status was slightly higher when imaging features of primary lung lesions were added compared to the model based solely on the clinical features, but without statistical significance (10-fold cross-validated AUC = 0.619 and 0.581, respectively; P = 0.425). The predictive performance of clinicoradiologic parameters slightly increased with primary lung lesions only compared to the inclusion of metastatic lesions, but without statistical significance (10-fold cross-validated AUC = 0.619 and 0.554, respectively; P = 0.203). Overall survival was prolonged in the TS-negative group compared to the TS-positive group (P = 0.001). TS-negativity is a potential prognostic biomarker, and our study presents that although CT radiomic features have potential for predicting TS expression status, clinical significance is uncertain. The addition of radiomic features to clinical factors did not show significant improvement in predicting TS-negativity.
Collapse
Affiliation(s)
- So Won Lee
- Department of Radiology and Center for Imaging Science, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea.,Department of Radiology, CHA Gangnam Medical Center, CHA University School of Medicine, Seoul, Korea
| | - Hyunjin Park
- School of Electronic and Electrical Engineering, Sungkyunkwan University, Suwon, Korea.,Center for Neuroscience Imaging Research, Institute for Basic Science, Suwon, Korea
| | - Ho Yun Lee
- Department of Radiology and Center for Imaging Science, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea.
| | - Insuk Sohn
- Statistics and Data Center, Samsung Medical Center, Seoul, Korea
| | - Seung-Hak Lee
- Department of Electronic Electrical and Computer Engineering, Sungkyunkwan University, Suwon, Korea
| | - Jun Kang
- Department of Pathology, Inchun St. Mary's Hospital, College of Medicine, Catholic University of Korea, Inchun, Korea
| | - Jong-Mu Sun
- Division of Hematology-Oncology, Department of Internal Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Myung-Ju Ahn
- Division of Hematology-Oncology, Department of Internal Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| |
Collapse
|
42
|
Rastogi A, Maheshwari S, Shinagare AB, Baheti AD. Computed Tomography Advances in Oncoimaging. Semin Roentgenol 2018; 53:147-156. [PMID: 29861006 DOI: 10.1053/j.ro.2018.02.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Ashita Rastogi
- Department of Radiodiagnosis, Tata Memorial Centre, Mumbai, India
| | - Sharad Maheshwari
- Department of Radiology, Kokilaben Dhirubhai Ambani Hospital, Mumbai, India
| | - Atul B Shinagare
- Department of Radiology, Harvard Medical School, Dana-Farber Cancer Institute, Boston, MA
| | - Akshay D Baheti
- Department of Radiodiagnosis, Tata Memorial Centre, Mumbai, India.
| |
Collapse
|
43
|
|
44
|
Cao Y, Xu H. A new predictive scoring system based on clinical data and computed tomography features for diagnosing EGFR-mutated lung adenocarcinoma. ACTA ACUST UNITED AC 2018; 25:e132-e138. [PMID: 29719437 DOI: 10.3747/co.25.3805] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Background We aimed to develop a new EGFR mutation-predictive scoring system to use in screening for EGFR-mutated lung adenocarcinomas (lacs). Methods The study enrolled 279 patients with lac, including 121 patients with EGFR wild-type tumours and 158 with EGFR-mutated tumours. The Student t-test, chi-square test, or Fisher exact test was applied to discriminate clinical and computed tomography (ct) features between the two groups. Using a principal component analysis (pca) model, we derived predictive coefficients for the presence of EGFR mutation in lac. Results The EGFR mutation-predictive score includes sex, smoking history, homogeneity, ground-glass opacity (ggo) on imaging, and the presence of pericardial effusion. The pca predictive model took this form: [Formula: see text]Model scores ranged from 79 to 147. The area under the receiver operating characteristic curve was 0.752 [95% confidence interval (ci): 0.697 to 0.801] in the lac population at the optimal cut-off value of 109, and the sensitivity and specificity were 68.4% (95% ci: 60.5% to 75.5%) and 74.4% (95% ci: 65.6% to 81.9%) respectively. Conclusions The EGFR mutation risk scoring system based on clinical data and ct features is noninvasive and user-friendly. The model appears to frame a positive predictive value and was able to determine the value of repeating a biopsy if tissue is limited.
Collapse
Affiliation(s)
- Y Cao
- Department of Radiology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, P.R.C
| | - H Xu
- Department of Radiology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, P.R.C
| |
Collapse
|
45
|
Jansen RW, van Amstel P, Martens RM, Kooi IE, Wesseling P, de Langen AJ, Menke-Van der Houven van Oordt CW, Jansen BHE, Moll AC, Dorsman JC, Castelijns JA, de Graaf P, de Jong MC. Non-invasive tumor genotyping using radiogenomic biomarkers, a systematic review and oncology-wide pathway analysis. Oncotarget 2018; 9:20134-20155. [PMID: 29732009 PMCID: PMC5929452 DOI: 10.18632/oncotarget.24893] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2017] [Accepted: 02/26/2018] [Indexed: 12/12/2022] Open
Abstract
With targeted treatments playing an increasing role in oncology, the need arises for fast non-invasive genotyping in clinical practice. Radiogenomics is a rapidly evolving field of research aimed at identifying imaging biomarkers useful for non-invasive genotyping. Radiogenomic genotyping has the advantage that it can capture tumor heterogeneity, can be performed repeatedly for treatment monitoring, and can be performed in malignancies for which biopsy is not available. In this systematic review of 187 included articles, we compiled a database of radiogenomic associations and unraveled networks of imaging groups and gene pathways oncology-wide. Results indicated that ill-defined tumor margins and tumor heterogeneity can potentially be used as imaging biomarkers for 1p/19q codeletion in glioma, relevant for prognosis and disease profiling. In non-small cell lung cancer, FDG-PET uptake and CT-ground-glass-opacity features were associated with treatment-informing traits including EGFR-mutations and ALK-rearrangements. Oncology-wide gene pathway analysis revealed an association between contrast enhancement (imaging) and the targetable VEGF-signalling pathway. Although the need of independent validation remains a concern, radiogenomic biomarkers showed potential for prognosis prediction and targeted treatment selection. Quantitative imaging enhanced the potential of multiparametric radiogenomic models. A wealth of data has been compiled for guiding future research towards robust non-invasive genomic profiling.
Collapse
Affiliation(s)
- Robin W Jansen
- Department of Radiology and Nuclear Medicine, VU University Medical Center, Amsterdam, The Netherlands
| | - Paul van Amstel
- Department of Radiology and Nuclear Medicine, VU University Medical Center, Amsterdam, The Netherlands
| | - Roland M Martens
- Department of Radiology and Nuclear Medicine, VU University Medical Center, Amsterdam, The Netherlands
| | - Irsan E Kooi
- Department of Clinical Genetics, VU University Medical Center, Amsterdam, The Netherlands
| | - Pieter Wesseling
- Department of Pathology, VU University Medical Center, Amsterdam, The Netherlands.,Department of Pathology, Princess Máxima Center for Pediatric Oncology and University Medical Center Utrecht, Utrecht, The Netherlands
| | - Adrianus J de Langen
- Department of Respiratory Diseases, VU University Medical Center, Amsterdam, The Netherlands
| | | | - Bernard H E Jansen
- Department of Radiology and Nuclear Medicine, VU University Medical Center, Amsterdam, The Netherlands
| | - Annette C Moll
- Department of Ophthalmology, VU University Medical Center, Amsterdam, The Netherlands
| | - Josephine C Dorsman
- Department of Clinical Genetics, VU University Medical Center, Amsterdam, The Netherlands
| | - Jonas A Castelijns
- Department of Radiology and Nuclear Medicine, VU University Medical Center, Amsterdam, The Netherlands
| | - Pim de Graaf
- Department of Radiology and Nuclear Medicine, VU University Medical Center, Amsterdam, The Netherlands
| | - Marcus C de Jong
- Department of Radiology and Nuclear Medicine, VU University Medical Center, Amsterdam, The Netherlands
| |
Collapse
|
46
|
Bak SH, Park H, Lee HY, Kim Y, Kim HL, Jung SH, Kim H, Kim J, Park K. Imaging genotyping of functional signaling pathways in lung squamous cell carcinoma using a radiomics approach. Sci Rep 2018; 8:3284. [PMID: 29459639 PMCID: PMC5818618 DOI: 10.1038/s41598-018-21706-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2017] [Accepted: 02/09/2018] [Indexed: 01/06/2023] Open
Abstract
Imaging features can be useful for identifying distinct genomic differences and have predictive power for certain phenotypes attributed to genomic mutations. We aimed to identify predictive imaging biomarkers that underpin genomic alterations and clinical outcomes in lung squamous cell carcinoma (SQCC) using a radiomics approach. In 57 patients with lung SQCC who underwent preoperative computed tomography (CT) and whole-exome DNA sequencing, 63 quantitative imaging features were extracted from CT and 73 clinicoradiological features including imaging features were classified into 8 categories: clinical, global, histogram-based, lung cancer-specific, shape, local, regional, and emphysema. Mutational profiles for core signaling pathways of lung SQCC were classified into five categories: redox stress, apoptosis, proliferation, differentiation, and chromatin remodelers. Range and right lung volume was significantly associated with alternation of apoptosis and proliferation pathway (p = 0.03, and p = 0.03). Energy was associated with the redox stress pathway (p = 0.06). None of the clinicoradiological features showed any significant association with the alteration of differentiation and chromatin remodelers pathway. This study showed that radiomic features indicating five different functional pathways of lung SQCC were different form one another. Radiomics approaches to lung SQCC have the potential to noninvasively predict alterations in core signaling pathways and clinical outcome.
Collapse
Affiliation(s)
- So Hyeon Bak
- Department of Radiology and Center for Imaging Science, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea.,Department of Radiology, Kangwon National University Hospital, Chuncheon, Korea
| | - Hyunjin Park
- School of Electronic and Electrical Engineering, Sungkyunkwan University, Suwon, Korea.,Center for Neuroscience Imaging Research (CNIR), Institute for Basic Science, Suwon, Korea
| | - Ho Yun Lee
- Department of Radiology and Center for Imaging Science, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea.
| | - Youngwook Kim
- Samsung Advanced Institute for Health Sciences and Technology, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Hyung-Lae Kim
- Department of Biochemistry, School of Medicine, Ewha Womans University, Seoul, Korea
| | - Sin-Ho Jung
- Statistics and Data Center, Research Institute for Future Medicine, Samsung Medical Center, Seoul, Korea
| | - Hyeseung Kim
- Statistics and Data Center, Research Institute for Future Medicine, Samsung Medical Center, Seoul, Korea
| | - Jonghoon Kim
- Department of Electronic Electrical and Computer Engineering, Sungkyunkwan University, Suwon, Korea
| | - Keunchil Park
- Division of Hematology/Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea.
| |
Collapse
|
47
|
Jakola AS, Zhang YH, Skjulsvik AJ, Solheim O, Bø HK, Berntsen EM, Reinertsen I, Gulati S, Förander P, Brismar TB. Quantitative texture analysis in the prediction of IDH status in low-grade gliomas. Clin Neurol Neurosurg 2018; 164:114-120. [DOI: 10.1016/j.clineuro.2017.12.007] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2017] [Revised: 11/30/2017] [Accepted: 12/04/2017] [Indexed: 01/17/2023]
|
48
|
Lubner MG, Smith AD, Sandrasegaran K, Sahani DV, Pickhardt PJ. CT Texture Analysis: Definitions, Applications, Biologic Correlates, and Challenges. Radiographics 2017; 37:1483-1503. [PMID: 28898189 DOI: 10.1148/rg.2017170056] [Citation(s) in RCA: 564] [Impact Index Per Article: 70.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
This review discusses potential oncologic and nononcologic applications of CT texture analysis ( CTTA CT texture analysis ), an emerging area of "radiomics" that extracts, analyzes, and interprets quantitative imaging features. CTTA CT texture analysis allows objective assessment of lesion and organ heterogeneity beyond what is possible with subjective visual interpretation and may reflect information about the tissue microenvironment. CTTA CT texture analysis has shown promise in lesion characterization, such as differentiating benign from malignant or more biologically aggressive lesions. Pretreatment CT texture features are associated with histopathologic correlates such as tumor grade, tumor cellular processes such as hypoxia or angiogenesis, and genetic features such as KRAS or epidermal growth factor receptor (EGFR) mutation status. In addition, and likely as a result, these CT texture features have been linked to prognosis and clinical outcomes in some tumor types. CTTA CT texture analysis has also been used to assess response to therapy, with decreases in tumor heterogeneity generally associated with pathologic response and improved outcomes. A variety of nononcologic applications of CTTA CT texture analysis are emerging, particularly quantifying fibrosis in the liver and lung. Although CTTA CT texture analysis seems to be a promising imaging biomarker, there is marked variability in methods, parameters reported, and strength of associations with biologic correlates. Before CTTA CT texture analysis can be considered for widespread clinical implementation, standardization of tumor segmentation and measurement techniques, image filtration and postprocessing techniques, and methods for mathematically handling multiple tumors and time points is needed, in addition to identification of key texture parameters among hundreds of potential candidates, continued investigation and external validation of histopathologic correlates, and structured reporting of findings. ©RSNA, 2017.
Collapse
Affiliation(s)
- Meghan G Lubner
- From the Department of Radiology, University of Wisconsin School of Medicine and Public Health, E3/311 Clinical Sciences Center, 600 Highland Ave, Madison, WI 35792 (M.G.L., P.J.P.); Department of Radiology, University of Mississippi Medical Center, Jackson, Miss (A.D.S.); Department of Radiology, Indiana University School of Medicine, Indianapolis, Ind (K.S.); and Department of Radiology, Harvard Medical School, Boston, Mass (D.V.S.)
| | - Andrew D Smith
- From the Department of Radiology, University of Wisconsin School of Medicine and Public Health, E3/311 Clinical Sciences Center, 600 Highland Ave, Madison, WI 35792 (M.G.L., P.J.P.); Department of Radiology, University of Mississippi Medical Center, Jackson, Miss (A.D.S.); Department of Radiology, Indiana University School of Medicine, Indianapolis, Ind (K.S.); and Department of Radiology, Harvard Medical School, Boston, Mass (D.V.S.)
| | - Kumar Sandrasegaran
- From the Department of Radiology, University of Wisconsin School of Medicine and Public Health, E3/311 Clinical Sciences Center, 600 Highland Ave, Madison, WI 35792 (M.G.L., P.J.P.); Department of Radiology, University of Mississippi Medical Center, Jackson, Miss (A.D.S.); Department of Radiology, Indiana University School of Medicine, Indianapolis, Ind (K.S.); and Department of Radiology, Harvard Medical School, Boston, Mass (D.V.S.)
| | - Dushyant V Sahani
- From the Department of Radiology, University of Wisconsin School of Medicine and Public Health, E3/311 Clinical Sciences Center, 600 Highland Ave, Madison, WI 35792 (M.G.L., P.J.P.); Department of Radiology, University of Mississippi Medical Center, Jackson, Miss (A.D.S.); Department of Radiology, Indiana University School of Medicine, Indianapolis, Ind (K.S.); and Department of Radiology, Harvard Medical School, Boston, Mass (D.V.S.)
| | - Perry J Pickhardt
- From the Department of Radiology, University of Wisconsin School of Medicine and Public Health, E3/311 Clinical Sciences Center, 600 Highland Ave, Madison, WI 35792 (M.G.L., P.J.P.); Department of Radiology, University of Mississippi Medical Center, Jackson, Miss (A.D.S.); Department of Radiology, Indiana University School of Medicine, Indianapolis, Ind (K.S.); and Department of Radiology, Harvard Medical School, Boston, Mass (D.V.S.)
| |
Collapse
|
49
|
Zhang L, Chen B, Liu X, Song J, Fang M, Hu C, Dong D, Li W, Tian J. Quantitative Biomarkers for Prediction of Epidermal Growth Factor Receptor Mutation in Non-Small Cell Lung Cancer. Transl Oncol 2017; 11:94-101. [PMID: 29216508 PMCID: PMC6002350 DOI: 10.1016/j.tranon.2017.10.012] [Citation(s) in RCA: 92] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2017] [Revised: 10/25/2017] [Accepted: 10/30/2017] [Indexed: 02/05/2023] Open
Abstract
OBJECTIVES To predict epidermal growth factor receptor (EGFR) mutation status using quantitative radiomic biomarkers and representative clinical variables. METHODS The study included 180 patients diagnosed as of non-small cell lung cancer (NSCLC) with their pre-therapy computed tomography (CT) scans. Using a radiomic method, 485 features that reflect the heterogeneity and phenotype of tumors were extracted. Afterwards, these radiomic features were used for predicting epidermal growth factor receptor (EGFR) mutation status by a least absolute shrinkage and selection operator (LASSO) based on multivariable logistic regression. As a result, we found that radiomic features have prognostic ability in EGFR mutation status prediction. In addition, we used radiomic nomogram and calibration curve to test the performance of the model. RESULTS Multivariate analysis revealed that the radiomic features had the potential to build a prediction model for EGFR mutation. The area under the receiver operating characteristic curve (AUC) for the training cohort was 0.8618, and the AUC for the validation cohort was 0.8725, which were superior to prediction model that used clinical variables alone. CONCLUSION Radiomic features are better predictors of EGFR mutation status than conventional semantic CT image features or clinical variables to help doctors to decide who need EGFR tyrosine kinase inhibitor (TKI) treatment.
Collapse
Affiliation(s)
- Liwen Zhang
- School of automation, Harbin University of Science and Technology, Harbin, Heilongjiang, 150080, China; CAS Key Lab of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing, 100190, China
| | - Bojiang Chen
- Department of respiratory and critical care medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Xia Liu
- School of automation, Harbin University of Science and Technology, Harbin, Heilongjiang, 150080, China
| | - Jiangdian Song
- School of Medical Informatics, China Medical University, Shenyang, Liaoning 110122, China
| | - Mengjie Fang
- CAS Key Lab of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing, 100190, China
| | - Chaoen Hu
- CAS Key Lab of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing, 100190, China
| | - Di Dong
- CAS Key Lab of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing, 100190, China; University of Chinese Academy of Sciences, Beijing, 100049, China.
| | - Weimin Li
- Department of respiratory and critical care medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China.
| | - Jie Tian
- CAS Key Lab of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing, 100190, China; University of Chinese Academy of Sciences, Beijing, 100049, China.
| |
Collapse
|
50
|
Kim Y, Oh D, Hwang D. Small-scale noise-like moiré pattern caused by detector sensitivity inhomogeneity in computed tomography. OPTICS EXPRESS 2017; 25:27127-27145. [PMID: 29092193 DOI: 10.1364/oe.25.027127] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/10/2017] [Accepted: 10/05/2017] [Indexed: 06/07/2023]
Abstract
We report a new type of moiré pattern caused by inhomogeneous detector sensitivity in computed tomography. Defects in one or a few detector bins or miscalibrated detectors induce well-known ring artifacts. When detector sensitivity is not homogenous over all detector bins, these ring artifacts occur everywhere as distributed rings in reconstructed images and may cause a moiré pattern when combined with insufficient view sampling, which induces a noise-like pattern or a subtle texture in the reconstructed images. Complete correction of the inhomogeneity in detectors can remove the pattern and improve image quality. This paper describes several properties of moiré patterns caused by detector sensitivity inhomogeneity.
Collapse
|