51
|
Jiménez-Sánchez J, Bosque JJ, Jiménez Londoño GA, Molina-García D, Martínez Á, Pérez-Beteta J, Ortega-Sabater C, Honguero Martínez AF, García Vicente AM, Calvo GF, Pérez-García VM. Evolutionary dynamics at the tumor edge reveal metabolic imaging biomarkers. Proc Natl Acad Sci U S A 2021; 118:e2018110118. [PMID: 33536339 PMCID: PMC8017959 DOI: 10.1073/pnas.2018110118] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Accepted: 01/04/2021] [Indexed: 01/09/2023] Open
Abstract
Human cancers are biologically and morphologically heterogeneous. A variety of clonal populations emerge within these neoplasms and their interaction leads to complex spatiotemporal dynamics during tumor growth. We studied the reshaping of metabolic activity in human cancers by means of continuous and discrete mathematical models and matched the results to positron emission tomography (PET) imaging data. Our models revealed that the location of increasingly active proliferative cellular spots progressively drifted from the center of the tumor to the periphery, as a result of the competition between gradually more aggressive phenotypes. This computational finding led to the development of a metric, normalized distance from 18F-fluorodeoxyglucose (18F-FDG) hotspot to centroid (NHOC), based on the separation from the location of the activity (proliferation) hotspot to the tumor centroid. The NHOC metric can be computed for patients using 18F-FDG PET-computed tomography (PET/CT) images where the voxel of maximum uptake (standardized uptake value [SUV]max) is taken as the activity hotspot. Two datasets of 18F-FDG PET/CT images were collected, one from 61 breast cancer patients and another from 161 non-small-cell lung cancer patients. In both cohorts, survival analyses were carried out for the NHOC and for other classical PET/CT-based biomarkers, finding that the former had a high prognostic value, outperforming the latter. In summary, our work offers additional insights into the evolutionary mechanisms behind tumor progression, provides a different PET/CT-based biomarker, and reveals that an activity hotspot closer to the tumor periphery is associated to a worst patient outcome.
Collapse
Affiliation(s)
- Juan Jiménez-Sánchez
- Mathematical Oncology Laboratory, Universidad de Castilla-La Mancha, Ciudad Real, 13071, Spain
| | - Jesús J Bosque
- Mathematical Oncology Laboratory, Universidad de Castilla-La Mancha, Ciudad Real, 13071, Spain
| | | | - David Molina-García
- Mathematical Oncology Laboratory, Universidad de Castilla-La Mancha, Ciudad Real, 13071, Spain
| | - Álvaro Martínez
- Mathematical Oncology Laboratory, Universidad de Castilla-La Mancha, Ciudad Real, 13071, Spain
- Nuclear Medicine Unit, Hospital General Universitario de Ciudad Real, Ciudad Real, 13005, Spain
| | - Julián Pérez-Beteta
- Mathematical Oncology Laboratory, Universidad de Castilla-La Mancha, Ciudad Real, 13071, Spain
| | - Carmen Ortega-Sabater
- Mathematical Oncology Laboratory, Universidad de Castilla-La Mancha, Ciudad Real, 13071, Spain
| | | | - Ana M García Vicente
- Thoracic Surgery Unit, Hospital General Universitario de Albacete, Albacete, 02006, Spain
| | - Gabriel F Calvo
- Mathematical Oncology Laboratory, Universidad de Castilla-La Mancha, Ciudad Real, 13071, Spain;
| | - Víctor M Pérez-García
- Mathematical Oncology Laboratory, Universidad de Castilla-La Mancha, Ciudad Real, 13071, Spain;
| |
Collapse
|
52
|
Chang C, Zhou S, Yu H, Zhao W, Ge Y, Duan S, Wang R, Qian X, Lei B, Wang L, Liu L, Ruan M, Yan H, Sun X, Xie W. A clinically practical radiomics-clinical combined model based on PET/CT data and nomogram predicts EGFR mutation in lung adenocarcinoma. Eur Radiol 2021; 31:6259-6268. [PMID: 33544167 DOI: 10.1007/s00330-020-07676-x] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Revised: 12/09/2020] [Accepted: 12/28/2020] [Indexed: 10/22/2022]
Abstract
OBJECTIVES This study aims to develop a clinically practical model to predict EGFR mutation in lung adenocarcinoma patients according to radiomics signatures based on PET/CT and clinical risk factors. METHODS This retrospective study included 583 lung adenocarcinoma patients, including 295 (50.60%) patients with EGFR mutation and 288 (49.40%) patients without EGFR mutation. The clinical risk factors associated with lung adenocarcinoma were collected at the same time. We developed PET/CT, CT, and PET radiomics models for the prediction of EGFR mutation using multivariate logistic regression analysis, respectively. We also constructed a combined PET/CT radiomics-clinical model by nomogram analysis. The diagnostic performance and clinical net benefit of this risk-scoring model were examined via receiver operating characteristic (ROC) curve analysis while the clinical usefulness of this model was evaluated by decision curve analysis (DCA). RESULTS The ROC analysis showed predictive performance for the PET/CT radiomics model (AUC = 0.76), better than the PET model (AUC = 0.71, Delong test: Z = 3.03, p value = 0.002) and the CT model (AUC = 0.74, Delong test: Z = 1.66, p value = 0.098). Also, the PET/CT radiomics-clinical combined model has a better performance (AUC = 0.84) to predict EGFR mutation than the PET/CT radiomics model (AUC = 0.76, Delong test: D = 2.70, df = 790.81, p value < 0.001) or the clinical model (AUC = 0.81, Delong test: Z = 3.46, p value < 0.001). CONCLUSIONS We demonstrated that the combined PET/CT radiomics-clinical model has an advantage to predict EGFR mutation in lung adenocarcinoma. KEY POINTS • Radiomics from lung tumor increase the efficiency of the prediction for EGFR mutation in clinical lung adenocarcinoma on PET/CT. • A radiomic nomogram was developed to predict EGFR mutation. • Combining PET/CT radiomics-clinical model has an advantage to predict EGFR mutation.
Collapse
Affiliation(s)
- Cheng Chang
- Department of Nuclear Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University, No. 241 West Huaihai Road, Shanghai, 200030, China.,Clinical and Translational Center in Shanghai Chest Hospital, Shanghai Key Laboratory for Molecular Imaging, Shanghai University of Medicine and Health Sciences, Shanghai, 201318, China
| | - Shihong Zhou
- Department of Thoracic Surgery, Shanghai Chest Hospital, Shanghai Jiao Tong University, No. 241 West Huaihai Road, Shanghai, 200030, China
| | - Hong Yu
- Department of Radiology, Shanghai Chest Hospital, Shanghai Jiao Tong University, No. 241 West Huaihai Road, Shanghai, 200030, China
| | - Wenlu Zhao
- Department of Radiology, Second Affiliated Hospital of Soochow University, No. 1055 Sanxiang Road, Gusu District, Suzhou, 215000, Jiangsu, China
| | - Yaqiong Ge
- GE Healthcare China, Pudong New Town, No. 1, Huatuo Road, Shanghai, 210000, China
| | - Shaofeng Duan
- GE Healthcare China, Pudong New Town, No. 1, Huatuo Road, Shanghai, 210000, China
| | - Rui Wang
- Department of Thoracic Surgery, Shanghai Chest Hospital, Shanghai Jiao Tong University, No. 241 West Huaihai Road, Shanghai, 200030, China
| | - Xiaohua Qian
- Institute for Medical Imaging Technology, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, 200030, China
| | - Bei Lei
- Department of Nuclear Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University, No. 241 West Huaihai Road, Shanghai, 200030, China
| | - Lihua Wang
- Department of Nuclear Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University, No. 241 West Huaihai Road, Shanghai, 200030, China
| | - Liu Liu
- Department of Nuclear Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University, No. 241 West Huaihai Road, Shanghai, 200030, China.,Clinical and Translational Center in Shanghai Chest Hospital, Shanghai Key Laboratory for Molecular Imaging, Shanghai University of Medicine and Health Sciences, Shanghai, 201318, China
| | - Maomei Ruan
- Department of Nuclear Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University, No. 241 West Huaihai Road, Shanghai, 200030, China
| | - Hui Yan
- Department of Nuclear Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University, No. 241 West Huaihai Road, Shanghai, 200030, China
| | - Xiaoyan Sun
- Department of Nuclear Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University, No. 241 West Huaihai Road, Shanghai, 200030, China.,Clinical and Translational Center in Shanghai Chest Hospital, Shanghai Key Laboratory for Molecular Imaging, Shanghai University of Medicine and Health Sciences, Shanghai, 201318, China
| | - Wenhui Xie
- Department of Nuclear Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University, No. 241 West Huaihai Road, Shanghai, 200030, China. .,Clinical and Translational Center in Shanghai Chest Hospital, Shanghai Key Laboratory for Molecular Imaging, Shanghai University of Medicine and Health Sciences, Shanghai, 201318, China.
| |
Collapse
|
53
|
Wang Y, Han R, Wang Q, Zheng J, Lin C, Lu C, Li L, Chen H, Jin R, He Y. Biological Significance of 18F-FDG PET/CT Maximum Standard Uptake Value for Predicting EGFR Mutation Status in Non-Small Cell Lung Cancer Patients. Int J Gen Med 2021; 14:347-356. [PMID: 33568935 PMCID: PMC7868188 DOI: 10.2147/ijgm.s287506] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Accepted: 12/31/2020] [Indexed: 12/27/2022] Open
Abstract
Purpose To investigate the potential of maximum standardized uptake value (SUVmax) in predicting epidermal growth factor receptor (EGFR) mutation status in non-small cell lung cancer (NSCLC) patients. Methods Clinical data of 311 NSCLC patients who had undergone both EGFR mutation test and 18F-FDG PET/CT scans between January 2013 and December 2017 at our hospital were retrospectively analyzed. Patients were sub-grouped by their origin of SUVmax. Univariate and multivariate analyses were performed to investigate the association between clinical factors and EGFR mutations. Receiver operating characteristic curve (ROC) analysis was performed to confirm the predictive value of clinical factors. In vitro experiments were performed to confirm the correlation between EGFR mutations and glycolysis. Results EGFR-mutant patients had higher SUVmax than the wild-type patients in both primary tumors and metastases. In the multivariate analysis, SUVmax, gender and histopathologic type were determined as independent predictors of EGFR mutation status for patients whose SUVmax were obtained from the primary tumors; while for patients whose SUVmax were obtained from the metastases, SUVmax, smoking status and histopathologic type were regarded as independent predictors. ROC analysis showed that SUVmax of the primary tumors (cut off >10.92), not of the metastases, has better predictive value than other clinical factors in predicting EGFR mutation status. The predict performance was improved after combined SUVmax with other independent predictors. In addition, our in vitro experiments demonstrated that lung cancer cells with EGFR mutations have higher aerobic glycolysis level than wild-type cells. Conclusion SUVmax of the primary tumors has the potential to serve as a biomarker to predict EGFR mutation status in NSCLC patients.
Collapse
Affiliation(s)
- Yubo Wang
- Department of Respiratory Medicine, Daping Hospital, Army Medical University, Chongqing, People's Republic of China
| | - Rui Han
- Department of Respiratory Medicine, Daping Hospital, Army Medical University, Chongqing, People's Republic of China
| | - Qiushi Wang
- Department of Pathology, Daping Hospital, Army Medical University, Chongqing, People's Republic of China
| | - Jie Zheng
- Department of Respiratory Medicine, Daping Hospital, Army Medical University, Chongqing, People's Republic of China
| | - Caiyu Lin
- Department of Respiratory Medicine, Daping Hospital, Army Medical University, Chongqing, People's Republic of China
| | - Conghua Lu
- Department of Respiratory Medicine, Daping Hospital, Army Medical University, Chongqing, People's Republic of China
| | - Li Li
- Department of Respiratory Medicine, Daping Hospital, Army Medical University, Chongqing, People's Republic of China
| | - Hengyi Chen
- Department of Respiratory Medicine, Daping Hospital, Army Medical University, Chongqing, People's Republic of China
| | - Rongbing Jin
- Department of Nuclear Medicine, Daping Hospital, Army Medical University, Chongqing, People's Republic of China
| | - Yong He
- Department of Respiratory Medicine, Daping Hospital, Army Medical University, Chongqing, People's Republic of China
| |
Collapse
|
54
|
Song Z, Liu T, Shi L, Yu Z, Shen Q, Xu M, Huang Z, Cai Z, Wang W, Xu C, Sun J, Chen M. The deep learning model combining CT image and clinicopathological information for predicting ALK fusion status and response to ALK-TKI therapy in non-small cell lung cancer patients. Eur J Nucl Med Mol Imaging 2021; 48:361-371. [PMID: 32794105 DOI: 10.1007/s00259-020-04986-6] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2020] [Accepted: 07/31/2020] [Indexed: 11/26/2022]
Abstract
PURPOSE This study aimed to investigate the deep learning model (DLM) combining computed tomography (CT) images and clinicopathological information for predicting anaplastic lymphoma kinase (ALK) fusion status in non-small cell lung cancer (NSCLC) patients. MATERIALS AND METHODS Preoperative CT images, clinicopathological information as well as the ALK fusion status from 937 patients in three hospitals were retrospectively collected to train and validate the DLM for the prediction of ALK fusion status in tumors. Another cohort of patients (n = 91) received ALK tyrosine kinase inhibitor (TKI) treatment was also included to evaluate the value of the DLM in predicting the clinical outcomes of the patients. RESULTS The performances of the DLM trained only by CT images in the primary and validation cohorts were AUC = 0.8046 (95% CI 0.7715-0.8378) and AUC = 0.7754 (95% CI 0.7199-0.8310), respectively, while the DLM trained by both CT images and clinicopathological information exhibited better performance for the prediction of ALK fusion status (AUC = 0.8540, 95% CI 0.8257-0.8823 in the primary cohort, p < 0.001; AUC = 0.8481, 95% CI 0.8036-0.8926 in the validation cohort, p < 0.001). In addition, the deep learning scores of the DLMs showed significant differences between the wild-type and ALK infusion tumors. In the ALK-target therapy cohort (n = 91), the patients predicted as ALK-positive by the DLM showed better performance of progression-free survival than the patients predicted as ALK-negative (16.8 vs. 7.5 months, p = 0.010). CONCLUSION Our findings showed that the DLM trained by both CT images and clinicopathological information could effectively predict the ALK fusion status and treatment responses of patients. For the small size of the ALK-target therapy cohort, larger data sets would be collected to further validate the performance of the model for predicting the response to ALK-TKI treatment.
Collapse
Affiliation(s)
- Zhengbo Song
- Department of Clinical Trial, Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, 310022, Zhejiang, China
| | - Tianchi Liu
- Shanghai Key Laboratory of Artificial Intelligence for Medical Image and Knowledge Graph, Shanghai, 200336, China
- YITU AI Research Institute for Healthcare, Hangzhou, 310000, Zhejiang, China
| | - Lei Shi
- Shanghai Key Laboratory of Artificial Intelligence for Medical Image and Knowledge Graph, Shanghai, 200336, China
- YITU AI Research Institute for Healthcare, Hangzhou, 310000, Zhejiang, China
| | - Zongyang Yu
- Department of Medical Oncology, 900th Hospital, Fuzhou, 350000, Fujian, China
| | - Qing Shen
- Shanghai Key Laboratory of Artificial Intelligence for Medical Image and Knowledge Graph, Shanghai, 200336, China
- YITU AI Research Institute for Healthcare, Hangzhou, 310000, Zhejiang, China
| | - Mengdi Xu
- Shanghai Key Laboratory of Artificial Intelligence for Medical Image and Knowledge Graph, Shanghai, 200336, China
- YITU AI Research Institute for Healthcare, Hangzhou, 310000, Zhejiang, China
| | - Zhangzhou Huang
- Department of Medical Oncology, Fujian Cancer Hospital, Fuzhou, 350001, China
| | - Zhijian Cai
- Institute of Immunology, Zhejiang University School of Medicine, Hangzhou, 310009, Zhejiang, China
| | - Wenxian Wang
- Department of Clinical Trial, Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, 310022, Zhejiang, China
| | - Chunwei Xu
- Department of Pathology, Fujian Cancer Hospital, Fuzhou, 350001, China
| | - Jingjing Sun
- Department of Radiology, Cancer Hospital of the University of Chinese Academy of Sciences(Zhejiang Cancer Hospital), Hangzhou, 310022, Zhejiang, China
| | - Ming Chen
- Department of Radiotherapy, Cancer Hospital of the University of Chinese Academy of Sciences(Zhejiang Cancer Hospital), Hangzhou, 310022, Zhejiang, China.
| |
Collapse
|
55
|
Lu CC, Yen RF, Peng KY, Huang JY, Wu KD, Chueh JS, Lin WY. NP-59 Adrenal Scintigraphy as an Imaging Biomarker to Predict KCNJ5 Mutation in Primary Aldosteronism Patients. Front Endocrinol (Lausanne) 2021; 12:644927. [PMID: 33995277 PMCID: PMC8113947 DOI: 10.3389/fendo.2021.644927] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Accepted: 04/06/2021] [Indexed: 11/25/2022] Open
Abstract
PURPOSE Somatic KCNJ5 mutation occurs in half of unilateral primary aldosteronism (PA) and is associated with more severe phenotype. Mutation status can only be identified by tissue sample from adrenalectomy. NP-59 adrenal scintigraphy is a noninvasive functional study for disease activity assessment. This study aimed to evaluate the predictive value of NP-59 adrenal scintigraphy in somatic KCNJ5 mutation among PA patients who received adrenalectomy. METHODS Sixty-two PA patients who had NP-59 adrenal scintigraphy before adrenalectomy with available KCNJ5 mutation status were included. Two semiquantitative parameters, adrenal to liver ratio (ALR) and lesion to contralateral ratio of bilateral adrenal glands (CON) derived from NP-59 adrenal scintigraphy, of mutated and wild-type patients were compared. Cutoff values calculated by receiver-operating characteristic (ROC) analysis were used as a predictor of KCNJ5 mutation. RESULTS Twenty patients had KCNJ5 mutation and 42 patients were wild type. Patients harboring KCNJ5 mutation had both higher ALR and CON (p = 0.0031 and 0.0833, respectively) than wild-type patients. With ALR and CON cutoff of 2.10 and 1.95, the sensitivity and specificity to predict KCNJ5 mutation were 85%, 57% and 45%, 93%, respectively. Among 20 patients with KCNJ5 mutation, 16 showed G151R point mutation (KCNJ5- G151R) and 4 showed L168R point mutation (KCNJ5-L168R), which former one had significantly lower ALR (p=0.0471). CONCLUSION PA patients harboring somatic KCNJ5 mutation had significantly higher NP-59 uptake regarding to ALR and CON than those without mutation. APAs with KCNJ5-L168R point mutation showed significantly higher ALR than those with KCNJ5-G151R point mutation.
Collapse
Affiliation(s)
- Ching-Chu Lu
- Department of Nuclear Medicine, National Taiwan University Hospital, Taipei, Taiwan
- Institute of Epidemiology and Preventive Medicine, College of Public Health, National Taiwan University, Taipei, Taiwan
| | - Ruoh-Fang Yen
- Department of Nuclear Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Kang-Yung Peng
- Division of Nephrology, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Jei-Yie Huang
- Department of Nuclear Medicine, National Taiwan University Hospital, Taipei, Taiwan
- Institute of Epidemiology and Preventive Medicine, College of Public Health, National Taiwan University, Taipei, Taiwan
| | - Kwan-Dun Wu
- Division of Nephrology, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Jeff S. Chueh
- Glickman Urological and Kidney Institute, Cleveland Clinic Lerner College of Medicine, Cleveland Clinic, Cleveland, OH, United States
| | - Wan-Yu Lin
- Institute of Epidemiology and Preventive Medicine, College of Public Health, National Taiwan University, Taipei, Taiwan
- Department of Public Health, College of Public Health, National Taiwan University, Taipei, Taiwan
- *Correspondence: Wan-Yu Lin,
| |
Collapse
|
56
|
Li W, Liu H, Cheng F, Li Y, Li S, Yan J. Artificial intelligence applications for oncological positron emission tomography imaging. Eur J Radiol 2020; 134:109448. [PMID: 33307463 DOI: 10.1016/j.ejrad.2020.109448] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 10/07/2020] [Accepted: 11/26/2020] [Indexed: 12/16/2022]
Abstract
Positron emission tomography (PET), a functional and dynamic molecular imaging technique, is generally used to reveal tumors' biological behavior. Radiomics allows a high-throughput extraction of multiple features from images with artificial intelligence (AI) approaches and develops rapidly worldwide. Quantitative and objective features of medical images have been explored to recognize reliable biomarkers, with the development of PET radiomics. This paper will review the current clinical exploration of PET-based classical machine learning and deep learning methods, including disease diagnosis, the prediction of histological subtype, gene mutation status, tumor metastasis, tumor relapse, therapeutic side effects, therapeutic intervention and evaluation of prognosis. The applications of AI in oncology will be mainly discussed. The image-guided biopsy or surgery assisted by PET-based AI will be introduced as well. This paper aims to present the applications and methods of AI for PET imaging, which may offer important details for further clinical studies. Relevant precautions are put forward and future research directions are suggested.
Collapse
Affiliation(s)
- Wanting Li
- Shanxi Medical University, Taiyuan 030009, PR China; Department of Nuclear Medicine, First Hospital of Shanxi Medical University, Taiyuan 030001, PR China; Collaborative Innovation Center for Molecular Imaging, Taiyuan 030001, PR China
| | - Haiyan Liu
- Department of Nuclear Medicine, First Hospital of Shanxi Medical University, Taiyuan 030001, PR China; Collaborative Innovation Center for Molecular Imaging, Taiyuan 030001, PR China; Cellular Physiology Key Laboratory of Ministry of Education, Translational Medicine Research Center, Shanxi Medical University, Taiyuan 030001, PR China
| | - Feng Cheng
- Shanxi Medical University, Taiyuan 030009, PR China
| | - Yanhua Li
- Shanxi Medical University, Taiyuan 030009, PR China
| | - Sijin Li
- Shanxi Medical University, Taiyuan 030009, PR China; Department of Nuclear Medicine, First Hospital of Shanxi Medical University, Taiyuan 030001, PR China; Collaborative Innovation Center for Molecular Imaging, Taiyuan 030001, PR China.
| | - Jiangwei Yan
- Shanxi Medical University, Taiyuan 030009, PR China.
| |
Collapse
|
57
|
Popovic M, Talarico O, van den Hoff J, Kunin H, Zhang Z, Lafontaine D, Dogan S, Leung J, Kaye E, Czmielewski C, Mayerhoefer ME, Zanzonico P, Yaeger R, Schöder H, Humm JL, Solomon SB, Sofocleous CT, Kirov AS. KRAS mutation effects on the 2-[18F]FDG PET uptake of colorectal adenocarcinoma metastases in the liver. EJNMMI Res 2020; 10:142. [PMID: 33226505 PMCID: PMC7683631 DOI: 10.1186/s13550-020-00707-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Accepted: 09/21/2020] [Indexed: 12/14/2022] Open
Abstract
Background Deriving individual tumor genomic characteristics from patient imaging analysis is desirable. We explore the predictive value of 2-[18F]FDG uptake with regard to the KRAS mutational status of colorectal adenocarcinoma liver metastases (CLM). Methods 2-[18F]FDG PET/CT images, surgical pathology and molecular diagnostic reports of 37 patients who underwent PET/CT-guided biopsy of CLM were reviewed under an IRB-approved retrospective research protocol. Sixty CLM in 39 interventional PET scans of the 37 patients were segmented using two different auto-segmentation tools implemented in different commercially available software packages. PET standard uptake values (SUV) were corrected for: (1) partial volume effect (PVE) using cold wall-corrected contrast recovery coefficients derived from phantom spheres with variable diameter and (2) variability of arterial tracer supply and variability of uptake time after injection until start of PET scan derived from the tumor-to-blood standard uptake ratio (SUR) approach. The correlations between the KRAS mutational status and the mean, peak and maximum SUV were investigated using Student’s t test, Wilcoxon rank sum test with continuity correction, logistic regression and receiver operation characteristic (ROC) analysis.
These correlation analyses were also performed for the ratios of the mean, peak and maximum tumor uptake to the mean blood activity concentration at the time of scan: SURMEAN, SURPEAK and SURMAX, respectively. Results Fifteen patients harbored KRAS missense mutations (KRAS+), while another 3 harbored KRAS gene amplification. For 31 lesions, the mutational status was derived from the PET/CT-guided biopsy. The Student’s t test p values for separating KRAS mutant cases decreased after applying PVE correction to all uptake metrics of each lesion and when applying correction for uptake time variability to the SUR metrics. The observed correlations were strongest when both corrections were applied to SURMAX and when the patients harboring gene amplification were grouped with the wild type: p ≤ 0.001; ROC area under the curve = 0.77 and 0.75 for the two different segmentations, respectively, with a mean specificity of 0.69 and sensitivity of 0.85. Conclusion The correlations observed after applying the described corrections show potential for assigning probabilities for the KRAS missense mutation status in CLM using 2-[18F]FDG PET images.
Collapse
Affiliation(s)
- M Popovic
- Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA.,Cornell University, Ithaca, NY, 14850, USA
| | - O Talarico
- Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA.,Vassar Brothers Medical Center, Poughkeepsie, NY, 12601, USA.,Lebedev Physical Institute RAS, Moscow, Russia, 119991
| | - J van den Hoff
- Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf, 01328, Dresden, Germany
| | - H Kunin
- Department of Radiology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA
| | - Z Zhang
- Department of Epidemiology & Biostatistics, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA
| | - D Lafontaine
- Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA
| | - S Dogan
- Department of Pathology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA
| | - J Leung
- Technology Division, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA
| | - E Kaye
- Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA
| | - C Czmielewski
- Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA
| | - M E Mayerhoefer
- Department of Radiology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA
| | - P Zanzonico
- Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA
| | - R Yaeger
- Department of Medicine, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA
| | - H Schöder
- Department of Radiology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA
| | - J L Humm
- Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA
| | - S B Solomon
- Department of Radiology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA
| | - C T Sofocleous
- Department of Radiology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA
| | - A S Kirov
- Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA.
| |
Collapse
|
58
|
Galván-Tejada CE, Villagrana-Bañuelos KE, Zanella-Calzada LA, Moreno-Báez A, Luna-García H, Celaya-Padilla JM, Galván-Tejada JI, Gamboa-Rosales H. Univariate Analysis of Short-Chain Fatty Acids Related to Sudden Infant Death Syndrome. Diagnostics (Basel) 2020; 10:E896. [PMID: 33147746 PMCID: PMC7693700 DOI: 10.3390/diagnostics10110896] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 10/12/2020] [Accepted: 10/19/2020] [Indexed: 11/29/2022] Open
Abstract
Sudden infant death syndrome (SIDS) is defined as the death of a child under one year of age, during sleep, without apparent cause, after exhaustive investigation, so it is a diagnosis of exclusion. SIDS is the principal cause of death in industrialized countries. Inborn errors of metabolism (IEM) have been related to SIDS. These errors are a group of conditions characterized by the accumulation of toxic substances usually produced by an enzyme defect and there are thousands of them and included are the disorders of the β-oxidation cycle, similarly to what can affect the metabolism of different types of fatty acid chain (within these, short chain fatty acids (SCFAs)). In this work, an analysis of postmortem SCFAs profiles of children who died due to SIDS is proposed. Initially, a set of features containing SCFAs information, obtained from the NIH Common Fund's National Metabolomics Data Repository (NMDR) is submitted to an univariate analysis, developing a model based on the relationship between each feature and the binary output (death due to SIDS or not), obtaining 11 univariate models. Then, each model is validated, calculating their receiver operating characteristic curve (ROC curve) and area under the ROC curve (AUC) value. For those features whose models presented an AUC value higher than 0.650, a new multivariate model is constructed, in order to validate its behavior in comparison to the univariate models. In addition, a comparison between this multivariate model and a model developed based on the whole set of features is finally performed. From the results, it can be observed that each SCFA which comprises of the SFCAs profile, has a relationship with SIDS and could help in risk identification.
Collapse
Affiliation(s)
- Carlos E. Galván-Tejada
- Unidad Académica de Ingeniería Eléctrica, Universidad Autónoma de Zacatecas, Jardín Juarez 147, Centro, Zacatecas 98000, Mexico; (C.E.G.-T.); (K.E.V.-B.); (A.M.-B.); (H.L.-G.); (J.M.C.-P.); (J.I.G.-T.)
| | - Karen E. Villagrana-Bañuelos
- Unidad Académica de Ingeniería Eléctrica, Universidad Autónoma de Zacatecas, Jardín Juarez 147, Centro, Zacatecas 98000, Mexico; (C.E.G.-T.); (K.E.V.-B.); (A.M.-B.); (H.L.-G.); (J.M.C.-P.); (J.I.G.-T.)
| | | | - Arturo Moreno-Báez
- Unidad Académica de Ingeniería Eléctrica, Universidad Autónoma de Zacatecas, Jardín Juarez 147, Centro, Zacatecas 98000, Mexico; (C.E.G.-T.); (K.E.V.-B.); (A.M.-B.); (H.L.-G.); (J.M.C.-P.); (J.I.G.-T.)
| | - Huizilopoztli Luna-García
- Unidad Académica de Ingeniería Eléctrica, Universidad Autónoma de Zacatecas, Jardín Juarez 147, Centro, Zacatecas 98000, Mexico; (C.E.G.-T.); (K.E.V.-B.); (A.M.-B.); (H.L.-G.); (J.M.C.-P.); (J.I.G.-T.)
| | - Jose M. Celaya-Padilla
- Unidad Académica de Ingeniería Eléctrica, Universidad Autónoma de Zacatecas, Jardín Juarez 147, Centro, Zacatecas 98000, Mexico; (C.E.G.-T.); (K.E.V.-B.); (A.M.-B.); (H.L.-G.); (J.M.C.-P.); (J.I.G.-T.)
| | - Jorge I. Galván-Tejada
- Unidad Académica de Ingeniería Eléctrica, Universidad Autónoma de Zacatecas, Jardín Juarez 147, Centro, Zacatecas 98000, Mexico; (C.E.G.-T.); (K.E.V.-B.); (A.M.-B.); (H.L.-G.); (J.M.C.-P.); (J.I.G.-T.)
| | - Hamurabi Gamboa-Rosales
- Unidad Académica de Ingeniería Eléctrica, Universidad Autónoma de Zacatecas, Jardín Juarez 147, Centro, Zacatecas 98000, Mexico; (C.E.G.-T.); (K.E.V.-B.); (A.M.-B.); (H.L.-G.); (J.M.C.-P.); (J.I.G.-T.)
| |
Collapse
|
59
|
Abstract
Radiomics describes the extraction of multiple features from medical images, including molecular imaging modalities, that with bioinformatic approaches, provide additional clinically relevant information that may be invisible to the human eye. This information may complement standard radiological interpretation with data that may better characterize a disease or that may provide predictive or prognostic information. Progressing from predefined image features, often describing heterogeneity of voxel intensities within a volume of interest, there is increasing use of machine learning to classify disease characteristics and deep learning methods based on artificial neural networks that can learn features without a priori definition and without the need for preprocessing of images. There have been advances in standardization and harmonization of methods to a level that should support multicenter studies. However, in this relatively early phase of research in the field, there are limited aspects that have been adopted into routine practice. Most of the reports in the molecular imaging field describe radiomic approaches in cancer using 18F-fluorodeoxyglucose positron emission tomography (18F-FDG-PET). In this review, we will describe radiomics in molecular imaging and summarize the pertinent literature in lung cancer where reports are most prevalent and mature.
Collapse
Affiliation(s)
- Gary J R Cook
- Cancer Imaging Department, School of Biomedical Engineering and Imaging Sciences, King's College London, London, UK; King's College London & Guy's and St Thomas' PET Centre, St Thomas' Hospital, London, UK.
| | - Vicky Goh
- Cancer Imaging Department, School of Biomedical Engineering and Imaging Sciences, King's College London, London, UK; Radiology Department, Guy's and St Thomas' Hospitals NHS Trust, London, UK
| |
Collapse
|
60
|
de la Pinta C, Barrios-Campo N, Sevillano D. Radiomics in lung cancer for oncologists. J Clin Transl Res 2020; 6:127-134. [PMID: 33521373 PMCID: PMC7837741] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 04/12/2020] [Accepted: 06/08/2020] [Indexed: 11/26/2022] Open
Abstract
UNLABELLED Radiomics has revolutionized the world of medical imaging. The aim of this review is to guide oncologists in radiomics and its applications in diagnosis, prediction of response and damage, prediction of survival, and prognosis in lung cancer. In this review, we analyzed published literature on PubMed and MEDLINE with papers published in the last 10 years. We included papers in English language with information about radiomics features and diagnostic, predictive, and prognosis of radiomics in lung cancer. All citations were evaluated for relevant content and validation. RELEVANCE FOR PATIENTS The evolution of technology allows the development of computer algorithms that facilitate the diagnosis and evaluation of response after different oncological treatments and their non-invasive follow-up.
Collapse
Affiliation(s)
| | - Nuria Barrios-Campo
- Department of Biomedical Engineering, Madrid Polytechnic University, Madrid, Spain
| | - David Sevillano
- Department of Medical Physics, Ramón y Cajal Hospital, Madrid, Spain
| |
Collapse
|
61
|
Mu W, Jiang L, Zhang J, Shi Y, Gray JE, Tunali I, Gao C, Sun Y, Tian J, Zhao X, Sun X, Gillies RJ, Schabath MB. Non-invasive decision support for NSCLC treatment using PET/CT radiomics. Nat Commun 2020; 11:5228. [PMID: 33067442 PMCID: PMC7567795 DOI: 10.1038/s41467-020-19116-x] [Citation(s) in RCA: 166] [Impact Index Per Article: 33.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Accepted: 09/14/2020] [Indexed: 12/26/2022] Open
Abstract
Two major treatment strategies employed in non-small cell lung cancer, NSCLC, are tyrosine kinase inhibitors, TKIs, and immune checkpoint inhibitors, ICIs. The choice of strategy is based on heterogeneous biomarkers that can dynamically change during therapy. Thus, there is a compelling need to identify comprehensive biomarkers that can be used longitudinally to help guide therapy choice. Herein, we report a 18F-FDG-PET/CT-based deep learning model, which demonstrates high accuracy in EGFR mutation status prediction across patient cohorts from different institutions. A deep learning score (EGFR-DLS) was significantly and positively associated with longer progression free survival (PFS) in patients treated with EGFR-TKIs, while EGFR-DLS is significantly and negatively associated with higher durable clinical benefit, reduced hyperprogression, and longer PFS among patients treated with ICIs. Thus, the EGFR-DLS provides a non-invasive method for precise quantification of EGFR mutation status in NSCLC patients, which is promising to identify NSCLC patients sensitive to EGFR-TKI or ICI-treatments.
Collapse
Affiliation(s)
- Wei Mu
- Department of Cancer Physiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Lei Jiang
- Department of Nuclear Medicine, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - JianYuan Zhang
- Department of Nuclear Medicine, the Fourth Hospital of Hebei Medical University, Hebei, China
- Department of Nuclear Medicine, Baoding No.1 Central Hospital, Baoding, Hebei, China
| | - Yu Shi
- Department of Cancer Physiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Jhanelle E Gray
- Department of Thoracic Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Ilke Tunali
- Department of Cancer Physiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Chao Gao
- NHC and CAMS Key Laboratory of Molecular Probe and Targeted Theranostics, Molecular Imaging Research Center (MIRC), Harbin Medical University, Harbin, Heilongjiang, China
- TOF-PET/CT/MR center, the Fourth Hospital of Harbin Medical University, Harbin Medical University, Harbin, Heilongjiang, China
| | - Yingying Sun
- NHC and CAMS Key Laboratory of Molecular Probe and Targeted Theranostics, Molecular Imaging Research Center (MIRC), Harbin Medical University, Harbin, Heilongjiang, China
- TOF-PET/CT/MR center, the Fourth Hospital of Harbin Medical University, Harbin Medical University, Harbin, Heilongjiang, China
| | - Jie Tian
- Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, School of Medicine, Beihang University, Beijing, China
- CAS Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing, China
| | - Xinming Zhao
- Department of Nuclear Medicine, the Fourth Hospital of Hebei Medical University, Hebei, China.
| | - Xilin Sun
- NHC and CAMS Key Laboratory of Molecular Probe and Targeted Theranostics, Molecular Imaging Research Center (MIRC), Harbin Medical University, Harbin, Heilongjiang, China.
- TOF-PET/CT/MR center, the Fourth Hospital of Harbin Medical University, Harbin Medical University, Harbin, Heilongjiang, China.
| | - Robert J Gillies
- Department of Cancer Physiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA.
| | - Matthew B Schabath
- Department of Thoracic Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA.
- Department of Cancer Epidemiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA.
| |
Collapse
|
62
|
Zhang M, Bao Y, Rui W, Shangguan C, Liu J, Xu J, Lin X, Zhang M, Huang X, Zhou Y, Qu Q, Meng H, Qian D, Li B. Performance of 18F-FDG PET/CT Radiomics for Predicting EGFR Mutation Status in Patients With Non-Small Cell Lung Cancer. Front Oncol 2020; 10:568857. [PMID: 33134170 PMCID: PMC7578399 DOI: 10.3389/fonc.2020.568857] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Accepted: 09/22/2020] [Indexed: 01/01/2023] Open
Abstract
Objective To assess the performance of pretreatment 18F-fluorodeoxyglucose positron emission tomography/computed tomography (18F-FDG PET/CT) radiomics features for predicting EGFR mutation status in patients with non-small cell lung cancer (NSCLC). Patients and Methods We enrolled total 173 patients with histologically proven NSCLC who underwent preoperative 18F-FDG PET/CT. Tumor tissues of all patients were tested for EGFR mutation status. A PET/CT radiomics prediction model was established through multi-step feature selection. The predictive performances of radiomics model, clinical features and conventional PET-derived semi-quantitative parameters were compared using receiver operating curves (ROCs) analysis. Results Four CT and two PET radiomics features were finally selected to build the PET/CT radiomics model. Compared with area under the ROC curve (AUC) equal to 0.664, 0.683 and 0.662 for clinical features, maximum standardized uptake values (SUVmax) and total lesion glycolysis (TLG), the PET/CT radiomics model showed better performance to discriminate between EGFR positive and negative mutations with the AUC of 0.769 and the accuracy of 67.06% after 10-fold cross-validation. The combined model, based on the PET/CT radiomics and clinical feature (gender) further improved the AUC to 0.827 and the accuracy to 75.29%. Only one PET radiomics feature demonstrated significant but low predictive ability (AUC = 0.661) for differentiating 19 Del from 21 L858R mutation subtypes. Conclusions EGFR mutations status in patients with NSCLC could be well predicted by the combined model based on 18F-FDG PET/CT radiomics and clinical feature, providing an alternative useful method for the selection of targeted therapy.
Collapse
Affiliation(s)
- Min Zhang
- Department of Nuclear Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yiming Bao
- Institute of Medical Robotics, Shanghai Jiao Tong University, Shanghai, China
| | - Weiwei Rui
- Department of Pathology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chengfang Shangguan
- Department of Oncology, Rujin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jiajun Liu
- Department of Nuclear Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jianwei Xu
- Institute of Medical Robotics, Shanghai Jiao Tong University, Shanghai, China
| | - Xiaozhu Lin
- Department of Nuclear Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Miao Zhang
- Department of Nuclear Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xinyun Huang
- Department of Nuclear Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yilei Zhou
- Department of Nuclear Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qian Qu
- Department of Nuclear Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hongping Meng
- Department of Nuclear Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Dahong Qian
- Department of Nuclear Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Institute of Medical Robotics, Shanghai Jiao Tong University, Shanghai, China
| | - Biao Li
- Department of Nuclear Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
63
|
Value of interim 18F-FDG PET/CT for predicting progression-free survival in stage ⅢB/IV EGFR-mutant non-small-cell lung cancer patients with EGFR-TKI therapy. Lung Cancer 2020; 149:137-143. [PMID: 33011375 DOI: 10.1016/j.lungcan.2020.09.020] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 09/11/2020] [Accepted: 09/23/2020] [Indexed: 11/23/2022]
Abstract
OBJECTIVES We retrospectively investigated the prognostic value of FDG-PET performed for patients with Stage ⅢB/IV EGFR-mutant non-small-cell lung cancer (NSCLC) receiving EGFR tyrosine kinase inhibitor (TKI) therapy. METHODS A total of 78 patients newly diagnosed with Stage ⅢB/IV EGFR-mutant NSCLC who received baseline and interim PET/CT examination and were treated with EGFR-TKI therapy were included. Interim PET was performed after 4-6 weeks of treatment. Cox proportional hazards regression analysis was used to assess the association between quantitative 18F-FDG PET/CT parameters, other clinicopathological factors and progression-free survival (PFS), non-durable clinical benefit (non-DCB). Five interim PET variables were analyzed in this study in the prediction of non-DCB. RESULTS The one-year and two-year progression-free survival rates of the patients were 33.9% (28.6-39.2%) and 20.7% (16.1-25.3%), respectively. Multivariable analysis indicated that interim PET relevant factors ΔSUVmax (p = 0.002, p = 0.014) and ΔSUVmean (p = 0.000, p = 0.030) were independent risk factors for predicting the PFS or non-DCB of patients receiving EGFR-TKI treatment. The optimal cutoff values of the parameters in the tumor survival analyses were 56.74% for ΔSUVmax (p = 0.002) and 36.48% for ΔSUVmean (p = 0.001). ΔSUVmax had the highest diagnostic value in the prediction of non-DCB. The one-year progression-free survival rates (95% confidence intervals) of patients with ΔSUVmax ≥ 56.74% and ΔSUVmax <56.74% were 59.5% (44.2-74.8%) and 5.7% (0.0-13.3%), respectively (p = 0.000). CONCLUSION An early PET scan after 4-6 weeks can effectively predict the PFS and non-DCB of patients with Stage ⅢB/IV EGFR-mutant NSCLC receiving EGFR-TKI therapy.
Collapse
|
64
|
Seol HY, Kim YS, Kim SJ. Predictive value of 18F-fluorodeoxyglucose positron emission tomography/computed tomography for PD-L1 expression in non-small cell lung cancer: A systematic review and meta-analysis. Thorac Cancer 2020; 11:3260-3268. [PMID: 32951338 PMCID: PMC7605997 DOI: 10.1111/1759-7714.13664] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Revised: 09/02/2020] [Accepted: 09/02/2020] [Indexed: 12/26/2022] Open
Abstract
Background The purpose of the current study was to investigate the predictive value of 18F‐fluorodeoxyglucose positron emission tomography/computed tomography (18F‐FDG PET/CT) for programmed death ligand 1 (PD‐L1) in non‐small cell lung cancer (NSCLC) patients through a systematic review and meta‐analysis. Methods The PubMed, Cochrane, and EMBASE database, from the earliest available date of indexing through 30 April 2020, were searched for studies evaluating the diagnostic performance of 18F‐FDG PET/CT for prediction of PD‐L1 expression in NSCLC patients. Results Across six studies (1739 patients), the pooled sensitivity for 18F‐FDG PET/CT was 0.72 (95% CI: 0.58–0.82) with heterogeneity (I2 = 90.9, P < 0.001) and a pooled specificity of 0.69 (95% CI: 0.64–0.74) with heterogeneity (I2 = 77.9, P < 0.001). Likelihood ratio (LR) syntheses gave an overall positive likelihood ratio (LR +) of 2.3 (95% CI: 1.8–2.9) and negative likelihood ratio (LR‐) of 0.41 (95% CI: 0.26–0.63). The pooled diagnostic odds ratio (DOR) was six (95% CI: 3–11). Hierarchical summary receiver operating characteristic (ROC) curve indicated that the area under the curve was 0.74 (95% CI: 0.70–0.78). Conclusions The current meta‐analysis showed a moderate sensitivity and specificity of 18F‐FDG PET/CT for the prediction of PD‐L1 expression in NSCLC patients. The DOR was low and the likelihood ratio scatter‐gram indicated that 18F‐FDG PET/CT might not be useful for the prediction of PD‐L1 expression in NSCLC patients and not for its exclusion. Key points Significant findings of the study The current meta‐analysis showed a moderate sensitivity and specificity of 18F‐FDG PET/CT for the prediction of PD‐L1 expression in NSCLC patients. The DOR was low and the likelihood ratio scattergram indicated that 18F‐FDG PET/CT might not be useful for the prediction of PD‐L1 expression in NSCLC patients and not for its exclusion. What this study adds This study concluded that the role of 18F‐FDG PET/CT in predicting tumor expression of PD‐L1 should be further elucidated.
Collapse
Affiliation(s)
- Hee Yun Seol
- Department of Internal Medicine, Pusan National University Yangsan Hospital, Yangsan, South Korea
| | - Yun Seong Kim
- Department of Internal Medicine, Pusan National University Yangsan Hospital, Yangsan, South Korea
| | - Seong-Jang Kim
- Department of Nuclear Medicine, College of Medicine, Pusan National University, Yangsan, South Korea.,Department of Nuclear Medicine, Pusan National University Yangsan Hospital, Yangsan, South Korea.,BioMedical Research Institute for Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan, South Korea
| |
Collapse
|
65
|
CT Radiomics in Colorectal Cancer: Detection of KRAS Mutation Using Texture Analysis and Machine Learning. APPLIED SCIENCES-BASEL 2020. [DOI: 10.3390/app10186214] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
In this work, by using descriptive techniques, the characteristics of the texture of the CT (computed tomography) image of patients with colorectal cancer were extracted and, subsequently, classified in KRAS+ or KRAS-. This was accomplished by using different classifiers, such as Support Vector Machine (SVM), Grading Boosting Machine (GBM), Neural Networks (NNET), and Random Forest (RF). Texture analysis can provide a quantitative assessment of tumour heterogeneity by analysing both the distribution and relationship between the pixels in the image. The objective of this research is to demonstrate that CT-based Radiomics can predict the presence of mutation in the KRAS gene in colorectal cancer. This is a retrospective study, with 47 patients from the University Hospital, with a confirmatory pathological analysis of KRAS mutation. The highest accuracy and kappa achieved were 83% and 64.7%, respectively, with a sensitivity of 88.9% and a specificity of 75.0%, achieved by the NNET classifier using the texture feature vectors combining wavelet transform and Haralick coefficients. The fact of being able to identify the genetic expression of a tumour without having to perform either a biopsy or a genetic test is a great advantage, because it prevents invasive procedures that involve complications and may present biases in the sample. As well, it leads towards a more personalized and effective treatment.
Collapse
|
66
|
Liu G, Xu Z, Ge Y, Jiang B, Groen H, Vliegenthart R, Xie X. 3D radiomics predicts EGFR mutation, exon-19 deletion and exon-21 L858R mutation in lung adenocarcinoma. Transl Lung Cancer Res 2020; 9:1212-1224. [PMID: 32953499 PMCID: PMC7481623 DOI: 10.21037/tlcr-20-122] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2020] [Accepted: 06/11/2020] [Indexed: 12/11/2022]
Abstract
BACKGROUND To establish a radiomic approach to identify epidermal growth factor receptor (EGFR) mutation status in lung adenocarcinoma patients based on CT images, and to distinguish exon-19 deletion and exon-21 L858R mutation. METHODS Two hundred sixty-three patients who underwent pre-surgical contrast-enhanced CT and molecular testing were included, and randomly divided into the training (80%) and test (20%) cohort. Tumor images were three-dimensionally segmented to extract 1,672 radiomic features. Clinical features (age, gender, and smoking history) were added to build classification models together with radiomic features. Subsequently, the top-10 most relevant features were used to establish classifiers. For the classifying tasks including EGFR mutation, exon-19 deletion, and exon-21 L858R mutation, four logistic regression models were established for each task. RESULTS The training and test cohort consisted of 210 and 53 patients, respectively. Among the established models, the highest accuracy and sensitivity among the four models were 75.5% (61.7-86.2%) and 92.9% (76.5-99.1%) to classify EGFR mutation, respectively. The highest specificity values were 86.7% (69.3-96.2%) and 70.4% (49.8-86.3%) to classify exon-19 deletion and exon-21 L858R mutation, respectively. CONCLUSIONS CT radiomics can sensitively identify the presence of EGFR mutation, and increase the certainty of distinguishing exon-19 deletion and exon-21 L858R mutation in lung adenocarcinoma patients. CT radiomics may become a helpful non-invasive biomarker to select EGFR mutation patients for invasive sampling.
Collapse
Affiliation(s)
- Guixue Liu
- Department of Radiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhihan Xu
- Siemens Healthineers Ltd, Shanghai, China
| | | | - Beibei Jiang
- Department of Radiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Harry Groen
- Department of Lung Diseases, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9700RB Groningen, The Netherlands
| | - Rozemarijn Vliegenthart
- Department of Radiology, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9700RB Groningen, The Netherlands
| | - Xueqian Xie
- Department of Radiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
67
|
Yao G, Zhou Y, Gu Y, Wang Z, Yang M, Sun J, Luo Q, Zhao H. Value of combining PET/CT and clinicopathological features in predicting EGFR mutation in Lung Adenocarcinoma with Bone Metastasis. J Cancer 2020; 11:5511-5517. [PMID: 32742498 PMCID: PMC7391209 DOI: 10.7150/jca.46414] [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] [Received: 03/27/2020] [Accepted: 06/29/2020] [Indexed: 12/22/2022] Open
Abstract
Purpose: Epidermal growth factor receptor (EGFR) mutation is the most common target for precision treatment in metastatic lung adenocarcinoma. We investigated the predictive role of 18F-FDG PET/CT and clinicopathological features for EGFR mutations in lung adenocarcinoma with bone metastasis. Methods: Seventy-five lung adenocarcinoma patients with histologically confirmed bone metastasis were included. They all received EGFR status test and PET/CT before systemic treatment. The differences of maximum standardized uptake value (SUVmax) in primary tumor (pSUVmax), regional lymph node (nSUVmax) and bone metastasis (bmSUVmax) between different EGFR status groups were compared, alongside with common clinicopathological features. Multivariate logistic regression analysis was performed to evaluate predictors of EGFR mutations. Results: EGFR mutations were found in 37 patients (49.3%). EGFR mutations were more common in females, non-smokers, expression of Thyroid Transcription Factor-1 (TTF-1) and NaspinA. Low bmSUVmax was significantly associated with EGFR mutations, while no significant difference was observed in pSUVmax and nSUVmax. Multivariate analysis showed that bmSUVmax ≤7, non-smoking, expression of TTF-1 were predictors of EGFR mutations. The area under the curve (AUC) of receiver operating characteristic (ROC) curve was 0.84 for the combination of the three factors. Conclusion: Low bmSUVmax is more frequently in EGFR mutations, and bmSUVmax is an independent predictor of EGFR mutations. Combining bmSUVmax with other clinicopathological features could forecast the EGFR status in lung adenocarcinoma with unavailable EGFR gene testing.
Collapse
Affiliation(s)
- Guangyu Yao
- Department of Internal Oncology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200030, China
| | - Yiyi Zhou
- Department of Internal Oncology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200030, China
| | - Yifeng Gu
- Department of Radiology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200030, China
| | - Zhiyu Wang
- Department of Internal Oncology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200030, China
| | - Mengdi Yang
- Department of Internal Oncology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200030, China
| | - Jing Sun
- Department of Internal Oncology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200030, China
| | - Quanyong Luo
- Department of Nuclear Medicine, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200030, China
| | - Hui Zhao
- Department of Internal Oncology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200030, China
| |
Collapse
|
68
|
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
|
69
|
Park YJ, Shin MH, Moon SH. Radiogenomics Based on PET Imaging. Nucl Med Mol Imaging 2020; 54:128-138. [PMID: 32582396 DOI: 10.1007/s13139-020-00642-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2020] [Revised: 04/02/2020] [Accepted: 04/30/2020] [Indexed: 02/07/2023] Open
Abstract
Radiogenomics or imaging genomics is a novel omics strategy of associating imaging data with genetic information, which has the potential to advance personalized medicine. Imaging features extracted from PET or PET/CT enable assessment of in vivo functional and physiological activity and provide comprehensive tumor information non-invasively. However, PET features are considered secondary to features on conventional imaging, and there has not yet been a review of the radiogenomic approach using PET features. This review article summarizes the current state of PET-based radiogenomic research for cancer, which discusses some of its limitations and directions for future study.
Collapse
Affiliation(s)
- Yong-Jin Park
- Department of Nuclear Medicine and Molecular Imaging, Samsung Medical Center, Seoul, Republic of Korea
| | - Mu Heon Shin
- Department of Nuclear Medicine and Molecular Imaging, Samsung Medical Center, Seoul, Republic of Korea
| | - Seung Hwan Moon
- Department of Nuclear Medicine and Molecular Imaging, Samsung Medical Center, Seoul, Republic of Korea
| |
Collapse
|
70
|
Avanzo M, Stancanello J, Pirrone G, Sartor G. Radiomics and deep learning in lung cancer. Strahlenther Onkol 2020; 196:879-887. [PMID: 32367456 DOI: 10.1007/s00066-020-01625-9] [Citation(s) in RCA: 130] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Accepted: 04/15/2020] [Indexed: 02/07/2023]
Abstract
Lung malignancies have been extensively characterized through radiomics and deep learning. By providing a three-dimensional characterization of the lesion, models based on radiomic features from computed tomography (CT) and positron-emission tomography (PET) have been developed to detect nodules, distinguish malignant from benign lesions, characterize their histology, stage, and genotype. Deep learning models have been applied to automatically segment organs at risk in lung cancer radiotherapy, stratify patients according to the risk for local and distant recurrence, and identify patients candidate for molecular targeted therapy and immunotherapy. Moreover, radiomics has also been applied successfully to predict side effects such as radiation- and immunotherapy-induced pneumonitis and differentiate lung injury from recurrence. Radiomics could also untap the potential for further use of the cone beam CT acquired for treatment image guidance, four-dimensional CT, and dose-volume data from radiotherapy treatment plans. Radiomics is expected to increasingly affect the clinical practice of treatment of lung tumors, optimizing the end-to-end diagnosis-treatment-follow-up chain. The main goal of this article is to provide an update on the current status of lung cancer radiomics.
Collapse
Affiliation(s)
- Michele Avanzo
- Department of Medical Physics, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Via F. Gallini 2, 33081, Aviano, PN, Italy.
| | | | - Giovanni Pirrone
- Department of Medical Physics, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Via F. Gallini 2, 33081, Aviano, PN, Italy
| | - Giovanna Sartor
- Department of Medical Physics, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Via F. Gallini 2, 33081, Aviano, PN, Italy
| |
Collapse
|
71
|
Mu W, Tunali I, Gray JE, Qi J, Schabath MB, Gillies RJ. Radiomics of 18F-FDG PET/CT images predicts clinical benefit of advanced NSCLC patients to checkpoint blockade immunotherapy. Eur J Nucl Med Mol Imaging 2020; 47:1168-1182. [PMID: 31807885 PMCID: PMC8663718 DOI: 10.1007/s00259-019-04625-9] [Citation(s) in RCA: 122] [Impact Index Per Article: 24.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Accepted: 11/18/2019] [Indexed: 01/04/2023]
Abstract
INTRODUCTION Immunotherapy has improved outcomes for patients with non-small cell lung cancer (NSCLC), yet durable clinical benefit (DCB) is experienced in only a fraction of patients. Here, we test the hypothesis that radiomics features from baseline pretreatment 18F-FDG PET/CT scans can predict clinical outcomes of NSCLC patients treated with checkpoint blockade immunotherapy. METHODS This study included 194 patients with histologically confirmed stage IIIB-IV NSCLC with pretreatment PET/CT images. Radiomics features were extracted from PET, CT, and PET+CT fusion images based on minimum Kullback-Leibler divergence (KLD) criteria. The radiomics features from 99 retrospective patients were used to train a multiparametric radiomics signature (mpRS) to predict DCB using an improved least absolute shrinkage and selection operator (LASSO) method, which was subsequently validated in both retrospective (N = 47) and prospective test cohorts (N = 48). Using these cohorts, the mpRS was also used to predict progression-free survival (PFS) and overall survival (OS) by training nomogram models using multivariable Cox regression analyses with additional clinical characteristics incorporated. RESULTS The mpRS could predict patients who will receive DCB, with areas under receiver operating characteristic curves (AUCs) of 0.86 (95%CI 0.79-0.94), 0.83 (95%CI 0.71-0.94), and 0.81 (95%CI 0.68-0.92) in the training, retrospective test, and prospective test cohorts, respectively. In the same three cohorts, respectively, nomogram models achieved C-indices of 0.74 (95%CI 0.68-0.80), 0.74 (95%CI 0.66-0.82), and 0.77 (95%CI 0.69-0.84) to predict PFS and C-indices of 0.83 (95%CI 0.77-0.88), 0.83 (95%CI 0.71-0.94), and 0.80 (95%CI 0.69-0.91) to predict OS. CONCLUSION PET/CT-based signature can be used prior to initiation of immunotherapy to identify NSCLC patients most likely to benefit from immunotherapy. As such, these data may be leveraged to improve more precise and individualized decision support in the treatment of patients with advanced NSCLC.
Collapse
Affiliation(s)
- Wei Mu
- Department of Cancer Physiology, H. Lee Moffitt Cancer Center and Research Institute, 12902 Magnolia Drive, Tampa, FL, 33612, USA
| | - Ilke Tunali
- Department of Cancer Physiology, H. Lee Moffitt Cancer Center and Research Institute, 12902 Magnolia Drive, Tampa, FL, 33612, USA
| | - Jhanelle E Gray
- Department of Thoracic Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Jin Qi
- Department of Cancer Physiology, H. Lee Moffitt Cancer Center and Research Institute, 12902 Magnolia Drive, Tampa, FL, 33612, USA
| | - Matthew B Schabath
- Department of Thoracic Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
- Department of Cancer Epidemiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Robert J Gillies
- Department of Cancer Physiology, H. Lee Moffitt Cancer Center and Research Institute, 12902 Magnolia Drive, Tampa, FL, 33612, USA.
| |
Collapse
|
72
|
Huemer F, Leisch M, Geisberger R, Melchardt T, Rinnerthaler G, Zaborsky N, Greil R. Combination Strategies for Immune-Checkpoint Blockade and Response Prediction by Artificial Intelligence. Int J Mol Sci 2020; 21:E2856. [PMID: 32325898 PMCID: PMC7215892 DOI: 10.3390/ijms21082856] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Revised: 04/14/2020] [Accepted: 04/15/2020] [Indexed: 12/23/2022] Open
Abstract
The therapeutic concept of unleashing a pre-existing immune response against the tumor by the application of immune-checkpoint inhibitors (ICI) has resulted in long-term survival in advanced cancer patient subgroups. However, the majority of patients do not benefit from single-agent ICI and therefore new combination strategies are eagerly necessitated. In addition to conventional chemotherapy, kinase inhibitors as well as tumor-specific vaccinations are extensively investigated in combination with ICI to augment therapy responses. An unprecedented clinical outcome with chimeric antigen receptor (CAR-)T cell therapy has led to the approval for relapsed/refractory diffuse large B cell lymphoma and B cell acute lymphoblastic leukemia whereas response rates in solid tumors are unsatisfactory. Immune-checkpoints negatively impact CAR-T cell therapy in hematologic and solid malignancies and as a consequence provide a therapeutic target to overcome resistance. Established biomarkers such as programmed death ligand 1 (PD-L1) and tumor mutational burden (TMB) help to select patients who will benefit most from ICI, however, biomarker negativity does not exclude responses. Investigating alterations in the antigen presenting pathway as well as radiomics have the potential to determine tumor immunogenicity and response to ICI. Within this review we summarize the literature about specific combination partners for ICI and the applicability of artificial intelligence to predict ICI therapy responses.
Collapse
Affiliation(s)
- Florian Huemer
- Department of Internal Medicine III with Haematology, Medical Oncology, Haemostaseology, Infectiology and Rheumatology, Oncologic Center, Paracelsus Medical University, 5020 Salzburg, Austria; (F.H.); (M.L.); (T.M.); (G.R.)
| | - Michael Leisch
- Department of Internal Medicine III with Haematology, Medical Oncology, Haemostaseology, Infectiology and Rheumatology, Oncologic Center, Paracelsus Medical University, 5020 Salzburg, Austria; (F.H.); (M.L.); (T.M.); (G.R.)
| | - Roland Geisberger
- Salzburg Cancer Research Institute-Laboratory for Immunological and Molecular Cancer Research (SCRI-LIMCR), 5020 Salzburg, Austria; (R.G.); (N.Z.)
| | - Thomas Melchardt
- Department of Internal Medicine III with Haematology, Medical Oncology, Haemostaseology, Infectiology and Rheumatology, Oncologic Center, Paracelsus Medical University, 5020 Salzburg, Austria; (F.H.); (M.L.); (T.M.); (G.R.)
| | - Gabriel Rinnerthaler
- Department of Internal Medicine III with Haematology, Medical Oncology, Haemostaseology, Infectiology and Rheumatology, Oncologic Center, Paracelsus Medical University, 5020 Salzburg, Austria; (F.H.); (M.L.); (T.M.); (G.R.)
- Cancer Cluster Salzburg, 5020 Salzburg, Austria
| | - Nadja Zaborsky
- Salzburg Cancer Research Institute-Laboratory for Immunological and Molecular Cancer Research (SCRI-LIMCR), 5020 Salzburg, Austria; (R.G.); (N.Z.)
- Cancer Cluster Salzburg, 5020 Salzburg, Austria
| | - Richard Greil
- Department of Internal Medicine III with Haematology, Medical Oncology, Haemostaseology, Infectiology and Rheumatology, Oncologic Center, Paracelsus Medical University, 5020 Salzburg, Austria; (F.H.); (M.L.); (T.M.); (G.R.)
- Salzburg Cancer Research Institute-Laboratory for Immunological and Molecular Cancer Research (SCRI-LIMCR), 5020 Salzburg, Austria; (R.G.); (N.Z.)
- Cancer Cluster Salzburg, 5020 Salzburg, Austria
| |
Collapse
|
73
|
Tixier F, Cheze-le-Rest C, Schick U, Simon B, Dufour X, Key S, Pradier O, Aubry M, Hatt M, Corcos L, Visvikis D. Transcriptomics in cancer revealed by Positron Emission Tomography radiomics. Sci Rep 2020; 10:5660. [PMID: 32221360 PMCID: PMC7101432 DOI: 10.1038/s41598-020-62414-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Accepted: 03/13/2020] [Indexed: 11/09/2022] Open
Abstract
Metabolic images from Positron Emission Tomography (PET) are used routinely for diagnosis, follow-up or treatment planning purposes of cancer patients. In this study we aimed at determining if radiomic features extracted from 18F-Fluoro Deoxy Glucose (FDG) PET images could mirror tumor transcriptomics. In this study we analyzed 45 patients with locally advanced head and neck cancer (H&N) that underwent FDG-PET scans at the time of diagnosis and transcriptome analysis using RNAs from both cancer and healthy tissues on microarrays. Association between PET radiomics and transcriptomics was carried out with the Genomica software and a functional annotation was used to associate PET radiomics, gene expression and altered biological pathways. We identified relationships between PET radiomics and genes involved in cell-cycle, disease, DNA repair, extracellular matrix organization, immune system, metabolism or signal transduction pathways, according to the Reactome classification. Our results suggest that these FDG PET radiomic features could be used to infer tissue gene expression and cellular pathway activity in H&N cancers. These observations strengthen the value of radiomics as a promising approach to personalize treatments through targeting tumor-specific molecular processes.
Collapse
Affiliation(s)
- Florent Tixier
- Department of Nuclear Medicine, Poitiers University Hospital, Poitiers, France.
- LaTIM, INSERM, UMR 1101, Univ Brest, Brest, France.
| | - Catherine Cheze-le-Rest
- Department of Nuclear Medicine, Poitiers University Hospital, Poitiers, France
- LaTIM, INSERM, UMR 1101, Univ Brest, Brest, France
| | - Ulrike Schick
- LaTIM, INSERM, UMR 1101, Univ Brest, Brest, France
- Radiation Oncology Department, University Hospital, Brest, France
| | - Brigitte Simon
- INSERM, UMR 1078, Université de Brest, Génétique Génomique Fonctionnelle et Biotechnologies, Etablissement Français du Sang, Brest, France
| | - Xavier Dufour
- Head and Neck Department, Poitiers University Hospital, Poitiers, France
| | - Stéphane Key
- Radiation Oncology Department, University Hospital, Brest, France
| | - Olivier Pradier
- Radiation Oncology Department, University Hospital, Brest, France
| | - Marc Aubry
- CNRS, UMR 6290, IGDR, Université de Rennes 1, Rennes, France
| | - Mathieu Hatt
- LaTIM, INSERM, UMR 1101, Univ Brest, Brest, France
| | - Laurent Corcos
- INSERM, UMR 1078, Université de Brest, Génétique Génomique Fonctionnelle et Biotechnologies, Etablissement Français du Sang, Brest, France
| | | |
Collapse
|
74
|
Konert T, Everitt S, La Fontaine MD, van de Kamer JB, MacManus MP, Vogel WV, Callahan J, Sonke JJ. Robust, independent and relevant prognostic 18F-fluorodeoxyglucose positron emission tomography radiomics features in non-small cell lung cancer: Are there any? PLoS One 2020; 15:e0228793. [PMID: 32097418 PMCID: PMC7041813 DOI: 10.1371/journal.pone.0228793] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2019] [Accepted: 01/22/2020] [Indexed: 12/14/2022] Open
Abstract
In locally advanced lung cancer, established baseline clinical variables show limited prognostic accuracy and 18F-fluorodeoxyglucose positron emission tomography (FDG PET) radiomics features may increase accuracy for optimal treatment selection. Their robustness and added value relative to current clinical factors are unknown. Hence, we identify robust and independent PET radiomics features that may have complementary value in predicting survival endpoints. A 4D PET dataset (n = 70) was used for assessing the repeatability (Bland-Altman analysis) and independence of PET radiomics features (Spearman rank: |ρ|<0.5). Two 3D PET datasets combined (n = 252) were used for training and validation of an elastic net regularized generalized logistic regression model (GLM) based on a selection of clinical and robust independent PET radiomics features (GLMall). The fitted model performance was externally validated (n = 40). The performance of GLMall (measured with area under the receiver operating characteristic curve, AUC) was highest in predicting 2-year overall survival (0.66±0.07). No significant improvement was observed for GLMall compared to a model containing only PET radiomics features or only clinical variables for any clinical endpoint. External validation of GLMall led to AUC values no higher than 0.55 for any clinical endpoint. In this study, robust independent FDG PET radiomics features did not have complementary value in predicting survival endpoints in lung cancer patients. Improving risk stratification and clinical decision making based on clinical variables and PET radiomics features has still been proven difficult in locally advanced lung cancer patients.
Collapse
Affiliation(s)
- Tom Konert
- Nuclear Medicine Department, Netherlands Cancer Institute, Amsterdam, The Netherlands
- Department of Radiation Oncology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Sarah Everitt
- Division of Radiation Oncology and Cancer Imaging, Peter MacCallum Cancer Centre, Melbourne, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Australia
| | - Matthew D. La Fontaine
- Department of Radiation Oncology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Jeroen B. van de Kamer
- Department of Radiation Oncology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Michael P. MacManus
- Division of Radiation Oncology and Cancer Imaging, Peter MacCallum Cancer Centre, Melbourne, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Australia
| | - Wouter V. Vogel
- Nuclear Medicine Department, Netherlands Cancer Institute, Amsterdam, The Netherlands
- Department of Radiation Oncology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Jason Callahan
- Division of Radiation Oncology and Cancer Imaging, Peter MacCallum Cancer Centre, Melbourne, Australia
| | - Jan-Jakob Sonke
- Department of Radiation Oncology, Netherlands Cancer Institute, Amsterdam, The Netherlands
- * E-mail:
| |
Collapse
|
75
|
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
|
76
|
Mayerhoefer ME, Materka A, Langs G, Häggström I, Szczypiński P, Gibbs P, Cook G. Introduction to Radiomics. J Nucl Med 2020; 61:488-495. [PMID: 32060219 DOI: 10.2967/jnumed.118.222893] [Citation(s) in RCA: 945] [Impact Index Per Article: 189.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Accepted: 01/28/2020] [Indexed: 12/11/2022] Open
Abstract
Radiomics is a rapidly evolving field of research concerned with the extraction of quantitative metrics-the so-called radiomic features-within medical images. Radiomic features capture tissue and lesion characteristics such as heterogeneity and shape and may, alone or in combination with demographic, histologic, genomic, or proteomic data, be used for clinical problem solving. The goal of this continuing education article is to provide an introduction to the field, covering the basic radiomics workflow: feature calculation and selection, dimensionality reduction, and data processing. Potential clinical applications in nuclear medicine that include PET radiomics-based prediction of treatment response and survival will be discussed. Current limitations of radiomics, such as sensitivity to acquisition parameter variations, and common pitfalls will also be covered.
Collapse
Affiliation(s)
- Marius E Mayerhoefer
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York .,Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, Vienna, Austria
| | - Andrzej Materka
- Institute of Electronics, Lodz University of Technology, Lodz, Poland
| | - Georg Langs
- Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, Vienna, Austria
| | - Ida Häggström
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Piotr Szczypiński
- Institute of Electronics, Lodz University of Technology, Lodz, Poland
| | - Peter Gibbs
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Gary Cook
- Cancer Imaging Department, School of Biomedical Engineering and Imaging Sciences, King's College London, London, United Kingdom; and.,King's College London and Guy's and St. Thomas' PET Centre, St. Thomas' Hospital, London, United Kingdom
| |
Collapse
|
77
|
Rubin DL, Ugur Akdogan M, Altindag C, Alkim E. ePAD: An Image Annotation and Analysis Platform for Quantitative Imaging. ACTA ACUST UNITED AC 2020; 5:170-183. [PMID: 30854455 PMCID: PMC6403025 DOI: 10.18383/j.tom.2018.00055] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Medical imaging is critical for assessing the response of patients to new cancer therapies. Quantitative lesion assessment on images is time-consuming, and adopting new promising quantitative imaging biomarkers of response in clinical trials is challenging. The electronic Physician Annotation Device (ePAD) is a freely available web-based zero-footprint software application for viewing, annotation, and quantitative analysis of radiology images designed to meet the challenges of quantitative evaluation of cancer lesions. For imaging researchers, ePAD calculates a variety of quantitative imaging biomarkers that they can analyze and compare in ePAD to identify potential candidates as surrogate endpoints in clinical trials. For clinicians, ePAD provides clinical decision support tools for evaluating cancer response through reports summarizing changes in tumor burden based on different imaging biomarkers. As a workflow management and study oversight tool, ePAD lets clinical trial project administrators create worklists for users and oversee the progress of annotations created by research groups. To support interoperability of image annotations, ePAD writes all image annotations and results of quantitative imaging analyses in standardized file formats, and it supports migration of annotations from various propriety formats. ePAD also provides a plugin architecture supporting MATLAB server-side modules in addition to client-side plugins, permitting the community to extend the ePAD platform in various ways for new cancer use cases. We present an overview of ePAD as a platform for medical image annotation and quantitative analysis. We also discuss use cases and collaborations with different groups in the Quantitative Imaging Network and future directions.
Collapse
Affiliation(s)
- Daniel L Rubin
- Department of Biomedical Data Science, Radiology, and Medicine (Biomedical Informatics Research), Stanford University, Stanford, CA
| | - Mete Ugur Akdogan
- Department of Biomedical Data Science, Radiology, and Medicine (Biomedical Informatics Research), Stanford University, Stanford, CA
| | - Cavit Altindag
- Department of Biomedical Data Science, Radiology, and Medicine (Biomedical Informatics Research), Stanford University, Stanford, CA
| | - Emel Alkim
- Department of Biomedical Data Science, Radiology, and Medicine (Biomedical Informatics Research), Stanford University, Stanford, CA
| |
Collapse
|
78
|
Jiang M, Sun D, Guo Y, Guo Y, Xiao J, Wang L, Yao X. Assessing PD-L1 Expression Level by Radiomic Features From PET/CT in Nonsmall Cell Lung Cancer Patients: An Initial Result. Acad Radiol 2020; 27:171-179. [PMID: 31147234 DOI: 10.1016/j.acra.2019.04.016] [Citation(s) in RCA: 79] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2019] [Revised: 04/12/2019] [Accepted: 04/13/2019] [Indexed: 12/24/2022]
Abstract
RATIONALE AND OBJECTIVES To explore the potential value of radiomic features-derived approach in assessing PD-L1 expression status in nonsmall cell lung cancer (NSCLC) patients. MATERIALS AND METHODS A cohort of 399 stage I-IV NSCLC patients were enrolled. Tumor segmentation was performed to select essential primary lesions of NSCLC cases after PET/CT images acquisition. Features were extracted, then filtered with automatic relevance determination and minimized with LASSO model based on its relevance of PD-L1 expression status. Finally, we built predictive models with features from the CT, the PET, and the PET/CT images, respectively, for differentiating different status of specific PD-L1 types. Five-fold cross validation was practiced to evaluate the signatures' accuracy, and the receiver operating characteristic as well as the corresponding area under the curve (AUC) was reckoned for each model. RESULTS With the total of 24 selected features which were significantly associated with PD-L1 expression levels, models based on CT-, PET-, PET/CT-derived features were built and compared. For PD-L1 (SP142) expression level over 1% prediction, models that comprised radiomic features from the CT, the PET, and the PET/CT images resulted in an AUC of 0.97, 0.61, and 0.97, respectively; models for over 50% prediction resulted with AUC of 0.80, 0.65, and 0.77. For PD-L1 (28-8) expression level prediction, predictive models of over 1% expression scored at 0.86, 0.62, and 0.85; and signatures of over 50% expression reached the score of AUCs at 0.91, 0.75, and 0.88, respectively. CONCLUSION The radiomic-based predictive approach, especially CT-derived predictive model, may anticipate PD-L1 expression status in NSCLC patients relatively accurate. It may be helpful in guiding immunotherapy in clinical practice and deserves further analysis.
Collapse
Affiliation(s)
- Mengmeng Jiang
- Shanghai Institute of Medical Imaging, No. 180 Fenglin Road, Xuhui District, Shanghai 200032, China; Department of Radiology, Zhongshan Hospital of Fudan University, Fudan University, No 180, Fenglin Road, Xuhui District, Shanghai 200032, China
| | - Dazhen Sun
- Shanghai Key Laboratory for Molecular Imaging, Shanghai University of Medicine and Health Sciences, and Department of Automation, Shanghai Jiao Tong University, 800 Dongchuan, RD. Minhang District, Shanghai, China
| | - Yinglong Guo
- Shanghai Institute of Medical Imaging, No. 180 Fenglin Road, Xuhui District, Shanghai 200032, China; Department of Radiology, Zhongshan Hospital of Fudan University, Fudan University, No 180, Fenglin Road, Xuhui District, Shanghai 200032, China
| | - Yixian Guo
- Shanghai Institute of Medical Imaging, No. 180 Fenglin Road, Xuhui District, Shanghai 200032, China; Department of Radiology, Zhongshan Hospital of Fudan University, Fudan University, No 180, Fenglin Road, Xuhui District, Shanghai 200032, China
| | - Jie Xiao
- Department of Nuclear Medicine, Zhongshan Hospital of Fudan University, Fudan University, Shanghai, China
| | - Lisheng Wang
- Shanghai Key Laboratory for Molecular Imaging, Shanghai University of Medicine and Health Sciences, and Department of Automation, Shanghai Jiao Tong University, 800 Dongchuan, RD. Minhang District, Shanghai, China.
| | - Xiuzhong Yao
- Shanghai Institute of Medical Imaging, No. 180 Fenglin Road, Xuhui District, Shanghai 200032, China; Department of Radiology, Zhongshan Hospital of Fudan University, Fudan University, No 180, Fenglin Road, Xuhui District, Shanghai 200032, China.
| |
Collapse
|
79
|
Aras G, Kanmaz ZD, Tuncay E, Çetinkaya E, Yentürk E, Kocatürk C, Öz B, Çermik TF, Purisa S. Relationship of radiometabolic biomarkers to KRAS mutation status and ALK rearrangements in cases of lung adenocarcinoma. TUMORI JOURNAL 2020; 105:501-508. [PMID: 31910789 DOI: 10.1177/0300891620902334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
PURPOSE Rapid diagnosis of genetic mutations is important for targeted therapies such as EGFR tyrosine kinase inhibitors. KRAS mutation and ALK rearrangement are also important in determining treatment. The purpose of our study was to evaluate the diagnostic value of 18F-FDG PET to predict KRAS mutation and ALK rearrangement in order to determine the frequency of these genetic markers in our lung adenocarcinoma cases and contribute to forthcoming meta-analysis studies. METHODS A total of 218 patients with lung adenocarcinoma (EGFR analyzed) who were seen at our clinic between 2012 and 2014 were included in the study. The results of the 18 F-FDG-PET scans for each patient were retrospectively recorded with the associated medical documents. ALK rearrangements were analyzed in 166 of the 218 patients, while 50 of the 218 patients were analyzed for KRAS mutational status. SPSS 15.0 for Windows was used for statistical analysis. RESULTS FDG avidity was higher in cases with KRAS mutations and ALK rearrangements than those without, but the difference was not significant. ALK rearrangements were more common in younger, female, and nonsmoking patients with lung adenocarcinoma. CONCLUSIONS The small numbers of KRAS mutations and ALK rearrangements are the limitation of this study for evaluation of diagnostic imaging. The frequency of these genetic alterations was as reported in the literature. We believe that our work will contribute to future meta-analysis.
Collapse
Affiliation(s)
- Gulfidan Aras
- Yedikule Chest Disease and Training Hospital, Istanbul - Turkey
| | | | - Esin Tuncay
- Yedikule Chest Disease and Training Hospital, Istanbul - Turkey
| | | | - Esin Yentürk
- Yedikule Chest Disease and Training Hospital, Istanbul - Turkey
| | | | - Büge Öz
- Cerrahpasa Medical Faculty, Pathology Department, Istanbul University, Istanbul - Turkey
| | - Tevfik Fikret Çermik
- Department of Nuclear Medicine, Istanbul Training and Research Hospital, Istanbul - Turkey
| | - Sevim Purisa
- Department of Statistics, Istanbul University, Istanbul - Turkey
| |
Collapse
|
80
|
Zhao W, Wu Y, Xu Y, Sun Y, Gao P, Tan M, Ma W, Li C, Jin L, Hua Y, Liu J, Li M. The Potential of Radiomics Nomogram in Non-invasively Prediction of Epidermal Growth Factor Receptor Mutation Status and Subtypes in Lung Adenocarcinoma. Front Oncol 2020; 9:1485. [PMID: 31993370 PMCID: PMC6962353 DOI: 10.3389/fonc.2019.01485] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Accepted: 12/10/2019] [Indexed: 12/13/2022] Open
Abstract
Purpose: Up to 50% of Asian patients with NSCLC have EGFR gene mutations, indicating that selecting eligible patients for EGFR-TKIs treatments is clinically important. The aim of the study is to develop and validate radiomics-based nomograms, integrating radiomics, CT features and clinical characteristics, to non-invasively predict EGFR mutation status and subtypes. Materials and Methods: We included 637 patients with lung adenocarcinomas, who performed the EGFR mutations analysis in the current study. The whole dataset was randomly split into a training dataset (n = 322) and validation dataset (n = 315). A sub-dataset of EGFR-mutant lesions (EGFR mutation in exon 19 and in exon 21) was used to explore the capability of radiomic features for predicting EGFR mutation subtypes. Four hundred seventy-five radiomic features were extracted and a radiomics sore (R-score) was constructed by using the least absolute shrinkage and selection operator (LASSO) regression in the training dataset. A radiomics-based nomogram, incorporating clinical characteristics, CT features and R-score was developed in the training dataset and evaluated in the validation dataset. Results: The constructed R-scores achieved promising performance on predicting EGFR mutation status and subtypes, with AUCs of 0.694 and 0.708 in two validation datasets, respectively. Moreover, the constructed radiomics-based nomograms excelled the R-scores, clinical, CT features alone in terms of predicting EGFR mutation status and subtypes, with AUCs of 0.734 and 0.757 in two validation datasets, respectively. Conclusions: Radiomics-based nomogram, incorporating clinical characteristics, CT features and radiomic features, can non-invasively and efficiently predict the EGFR mutation status and thus potentially fulfill the ultimate purpose of precision medicine. The methodology is a possible promising strategy to predict EGFR mutation subtypes, providing the support of clinical treatment scenario.
Collapse
Affiliation(s)
- Wei Zhao
- Department of Radiology, Second Xiangya Hospital, Central South University, Changsha, China.,Department of Radiology, Huadong Hospital Affiliated to Fudan University, Shanghai, China
| | - Yuzhi Wu
- Department of Radiology, Second Xiangya Hospital, Central South University, Changsha, China
| | - Ya'nan Xu
- School of Biomedical Engineering, Capital Medical University, Beijing, China
| | - Yingli Sun
- Department of Radiology, Huadong Hospital Affiliated to Fudan University, Shanghai, China
| | - Pan Gao
- Department of Radiology, Huadong Hospital Affiliated to Fudan University, Shanghai, China
| | - Mingyu Tan
- Department of Radiology, Huadong Hospital Affiliated to Fudan University, Shanghai, China
| | - Weiling Ma
- Department of Radiology, Huadong Hospital Affiliated to Fudan University, Shanghai, China
| | - Cheng Li
- Department of Radiology, Huadong Hospital Affiliated to Fudan University, Shanghai, China
| | - Liang Jin
- Department of Radiology, Huadong Hospital Affiliated to Fudan University, Shanghai, China
| | - Yanqing Hua
- Department of Radiology, Huadong Hospital Affiliated to Fudan University, Shanghai, China
| | - Jun Liu
- Department of Radiology, Second Xiangya Hospital, Central South University, Changsha, China
| | - Ming Li
- Department of Radiology, Huadong Hospital Affiliated to Fudan University, Shanghai, China.,Diagnosis and Treatment Center of Small Lung Nodules of Huadong Hospital, Shanghai, China.,Institute of Functional and Molecular Medical Imaging, Fudan University, Shanghai, China
| |
Collapse
|
81
|
Correlation between combining 18F–FDG PET/CT metabolic parameters and other clinical features and ALK or ROS1 fusion in patients with non-small-cell lung cancer. Eur J Nucl Med Mol Imaging 2020; 47:1183-1197. [DOI: 10.1007/s00259-019-04652-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Accepted: 12/09/2019] [Indexed: 01/03/2023]
|
82
|
Assessing EGFR gene mutation status in non-small cell lung cancer with imaging features from PET/CT. Nucl Med Commun 2019; 40:842-849. [PMID: 31290849 DOI: 10.1097/mnm.0000000000001043] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
OBJECTIVE The aim of this study was to investigate whether quantitative and qualitative features extracted from PET/computed tomography (CT) can be used as imaging biomarkers for evaluating epidermal growth factor receptor (EGFR) mutation status in non-small cell lung cancer patients. METHODS Eighty patients with stage II and III non-small cell lung cancer from January 2017 to December 2017 were included in this study. All patients underwent PET/CT examination before operation. Patients with 30 EGFR positive and 50 EGFR negative were confirmed by pathological verification and gene detection. Least absolute shrinkage and selection operator was used for analysis and selection of imaging features. Support vector machine was used to classify EGFR positive/negative using the selected features. Ten-fold cross validation was used to estimate the accuracy. RESULTS A total of 512 quantitative features (radiomic features) were extracted from PET/CT (256 for PET and 256 for CT), and 12 qualitative features (semantic features) were extracted from CT. A total of 35 features were finally retained after least absolute shrinkage and selection operator (31 quantitative features and 4 qualitative features). The 35 selected features were significantly associated with EGFR mutation status. A predictive model was built using PET/CT data. Its performance was revealed as 0.953 using the area under the receiver operating characteristic curve. CONCLUSION A predictive model using PET/CT images might be used to detect EGFR mutation status in non-small cell lung cancer patients.
Collapse
|
83
|
Cook GJR, Goh V. What can artificial intelligence teach us about the molecular mechanisms underlying disease? Eur J Nucl Med Mol Imaging 2019; 46:2715-2721. [PMID: 31190176 PMCID: PMC6879441 DOI: 10.1007/s00259-019-04370-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Accepted: 05/23/2019] [Indexed: 12/24/2022]
Abstract
While molecular imaging with positron emission tomography or single-photon emission computed tomography already reports on tumour molecular mechanisms on a macroscopic scale, there is increasing evidence that there are multiple additional features within medical images that can further improve tumour characterization, treatment prediction and prognostication. Early reports have already revealed the power of radiomics to personalize and improve patient management and outcomes. What remains unclear is how these additional metrics relate to underlying molecular mechanisms of disease. Furthermore, the ability to deal with increasingly large amounts of data from medical images and beyond in a rapid, reproducible and transparent manner is essential for future clinical practice. Here, artificial intelligence (AI) may have an impact. AI encompasses a broad range of 'intelligent' functions performed by computers, including language processing, knowledge representation, problem solving and planning. While rule-based algorithms, e.g. computer-aided diagnosis, have been in use for medical imaging since the 1990s, the resurgent interest in AI is related to improvements in computing power and advances in machine learning (ML). In this review we consider why molecular and cellular processes are of interest and which processes have already been exposed to AI and ML methods as reported in the literature. Non-small-cell lung cancer is used as an exemplar and the focus of this review as the most common tumour type in which AI and ML approaches have been tested and to illustrate some of the concepts.
Collapse
Affiliation(s)
- Gary J R Cook
- Cancer Imaging Department, School of Biomedical Engineering and Imaging Sciences, King's College London, London, SE1 7EH, UK.
- King's College London & Guy's and St Thomas' PET Centre, St Thomas' Hospital, London, UK.
| | - Vicky Goh
- Cancer Imaging Department, School of Biomedical Engineering and Imaging Sciences, King's College London, London, SE1 7EH, UK
- Radiology Department, Guy's and St Thomas' Hospitals NHS Trust, London, UK
| |
Collapse
|
84
|
Cheze Le Rest C, Hustinx R. Are radiomics ready for clinical prime-time in PET/CT imaging? 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; 63:347-354. [DOI: 10.23736/s1824-4785.19.03210-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
85
|
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
|
86
|
Liang M, Cai Z, Zhang H, Huang C, Meng Y, Zhao L, Li D, Ma X, Zhao X. Machine Learning-based Analysis of Rectal Cancer MRI Radiomics for Prediction of Metachronous Liver Metastasis. Acad Radiol 2019; 26:1495-1504. [PMID: 30711405 DOI: 10.1016/j.acra.2018.12.019] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2018] [Revised: 12/17/2018] [Accepted: 12/21/2018] [Indexed: 12/12/2022]
Abstract
RATIONALE AND OBJECTIVES To use machine learning-based magnetic resonance imaging radiomics to predict metachronous liver metastases (MLM) in patients with rectal cancer. MATERIALS AND METHODS This study retrospectively analyzed 108 patients with rectal cancer (54 in MLM group and 54 in nonmetastases group). Feature selection were performed in the radiomic feature sets extracted from images of T2-weighted image (T2WI) and venous phase (VP) sequence respectively, and the combining feature set with 2058 radiomic features incorporating two sequences with the least absolute shrinkage and selection operator method. Five-fold cross-validation and two machine learning algorithms (support vector machine [SVM]; logistic regression [LR]) were utilized for predictive model constructing. The diagnostic performance of the models was evaluated by receiver operating characteristic curves with indicators of accuracy, sensitivity, specificity and area under the curve, and compared by DeLong test. RESULTS Five, 8, and 22 optimal features were selected from 1029 T2WI, 1029 VP, and 2058 combining features, respectively. Four-group models were constructed using the five T2WI features (ModelT2), the 8 VP features (ModelVP), the combined 13 optimal features (Modelcombined), and the 22 optimal features selected from 2058 features (Modeloptimal). In ModelVP, the LR was superior to the SVM algorithm (P = 0.0303). The Modeloptimal using LR algorithm showed the best prediction performance (P = 0.0019-0.0081) with accuracy, sensitivity, specificity, and area under the curve of 0.80, 0.83, 0.76, and 0.87, respectively. CONCLUSION Radiomics models based on baseline rectal magnetic resonance imaging has high potential for MLM prediction, especially the Modeloptimal using LR algorithm. Moreover, except for ModelVP, the LR was not superior to the SVM algorithm for model construction.
Collapse
Affiliation(s)
- Meng Liang
- Department of Radiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, People's Republic of China
| | - Zhengting Cai
- Huiying Medical Technology Co., Ltd., HaiDian District, Beijing City, 100192, People's Republic of China
| | - Hongmei Zhang
- Department of Radiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, People's Republic of China
| | - Chencui Huang
- Huiying Medical Technology Co., Ltd., HaiDian District, Beijing City, 100192, People's Republic of China
| | - Yankai Meng
- Department of Radiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, People's Republic of China; Department of Radiology, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu Province, People's Republic of China
| | - Li Zhao
- Department of Radiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, People's Republic of China
| | - Dengfeng Li
- Department of Radiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, People's Republic of China
| | - Xiaohong Ma
- Department of Radiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, People's Republic of China.
| | - Xinming Zhao
- Department of Radiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, People's Republic of China.
| |
Collapse
|
87
|
Li X, Yin G, Zhang Y, Dai D, Liu J, Chen P, Zhu L, Ma W, Xu W. Predictive Power of a Radiomic Signature Based on 18F-FDG PET/CT Images for EGFR Mutational Status in NSCLC. Front Oncol 2019; 9:1062. [PMID: 31681597 PMCID: PMC6803612 DOI: 10.3389/fonc.2019.01062] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Accepted: 09/30/2019] [Indexed: 12/13/2022] Open
Abstract
Radiomics has become an area of interest for tumor characterization in 18F-Fluorodeoxyglucose positron emission tomography/computed tomography (18F-FDG PET/CT) imaging. The aim of the present study was to demonstrate how imaging phenotypes was connected to somatic mutations through an integrated analysis of 115 non-small cell lung cancer (NSCLC) patients with somatic mutation testings and engineered computed PET/CT image analytics. A total of 38 radiomic features quantifying tumor morphological, grayscale statistic, and texture features were extracted from the segmented entire-tumor region of interest (ROI) of the primary PET/CT images. The ensembles for boosting machine learning scheme were employed for classification, and the least absolute shrink age and selection operator (LASSO) method was used to select the most predictive radiomic features for the classifiers. A radiomic signature based on both PET and CT radiomic features outperformed individual radiomic features, the PET or CT radiomic signature, and the conventional PET parameters including the maximum standardized uptake value (SUVmax), SUVmean, SUVpeak, metabolic tumor volume (MTV), and total lesion glycolysis (TLG), in discriminating between mutant-type of epidermal growth factor receptor (EGFR) and wild-type of EGFR- cases with an AUC of 0.805, an accuracy of 80.798%, a sensitivity of 0.826 and a specificity of 0.783. Consistently, a combined radiomic signature with clinical factors exhibited a further improved performance in EGFR mutation differentiation in NSCLC. In conclusion, tumor imaging phenotypes that are driven by somatic mutations may be predicted by radiomics based on PET/CT images.
Collapse
Affiliation(s)
- Xiaofeng Li
- Department of Molecular Imaging and Nuclear Medicine, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China.,National Clinical Research Center for Cancer, Tianjin, China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin, China.,Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Guotao Yin
- Department of Molecular Imaging and Nuclear Medicine, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China.,National Clinical Research Center for Cancer, Tianjin, China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin, China.,Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Yufan Zhang
- Department of Molecular Imaging and Nuclear Medicine, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China.,National Clinical Research Center for Cancer, Tianjin, China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin, China.,Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Dong Dai
- Department of Molecular Imaging and Nuclear Medicine, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China.,National Clinical Research Center for Cancer, Tianjin, China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin, China.,Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Jianjing Liu
- Department of Molecular Imaging and Nuclear Medicine, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China.,National Clinical Research Center for Cancer, Tianjin, China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin, China.,Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Peihe Chen
- Department of Molecular Imaging and Nuclear Medicine, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China.,National Clinical Research Center for Cancer, Tianjin, China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin, China.,Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Lei Zhu
- Department of Molecular Imaging and Nuclear Medicine, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China.,National Clinical Research Center for Cancer, Tianjin, China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin, China.,Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Wenjuan Ma
- National Clinical Research Center for Cancer, Tianjin, China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin, China.,Tianjin's Clinical Research Center for Cancer, Tianjin, China.,Department of Breast Imaging, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Wengui Xu
- Department of Molecular Imaging and Nuclear Medicine, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China.,National Clinical Research Center for Cancer, Tianjin, China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin, China.,Tianjin's Clinical Research Center for Cancer, Tianjin, China
| |
Collapse
|
88
|
Kong Z, Li J, Liu Z, Liu Z, Zhao D, Cheng X, Li L, Lin Y, Wang Y, Tian J, Ma W. Radiomics signature based on FDG-PET predicts proliferative activity in primary glioma. Clin Radiol 2019; 74:815.e15-815.e23. [DOI: 10.1016/j.crad.2019.06.019] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Accepted: 06/26/2019] [Indexed: 01/04/2023]
|
89
|
Saadani H, van der Hiel B, Aalbersberg EA, Zavrakidis I, Haanen JBAG, Hoekstra OS, Boellaard R, Stokkel MPM. Metabolic Biomarker-Based BRAFV600 Mutation Association and Prediction in Melanoma. J Nucl Med 2019; 60:1545-1552. [PMID: 31481581 DOI: 10.2967/jnumed.119.228312] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Accepted: 08/05/2019] [Indexed: 12/24/2022] Open
Abstract
The aim of this study was to associate and predict B-rapidly accelerated fibrosarcoma valine 600 (BRAFV600) mutation status with both conventional and radiomics 18F-FDG PET/CT features, while exploring several methods of feature selection in melanoma radiomics. Methods: Seventy unresectable stage III-IV melanoma patients who underwent a baseline 18F-FDG PET/CT scan were identified. Patients were assigned to the BRAFV600 group or BRAF wild-type group according to mutational status. 18F-FDG uptake quantification was performed by semiautomatic lesion delineation. Four hundred eighty radiomics features and 4 conventional PET features (SUVmax, SUVmean, SUVpeak, and total lesion glycolysis) were extracted per lesion. Six different methods of feature selection were implemented, and 10-fold cross-validated predictive models were built for each. Model performances were evaluated with areas under the curve (AUCs) for the receiver operating characteristic curves. Results: Thirty-five BRAFV600 mutated patients (100 lesions) and 35 BRAF wild-type patients (79 lesions) were analyzed. AUCs predicting the BRAFV600 mutation varied from 0.54 to 0.62 and were susceptible to feature selection method. The best AUCs were achieved by feature selection based on literature, a penalized binary logistic regression model, and random forest model. No significant difference was found between the BRAFV600 and BRAF wild-type group in conventional PET features or predictive value. Conclusion: BRAFV600 mutation status is not associated with, nor can it be predicted with, conventional PET features, whereas radiomics features were of low predictive value (AUC = 0.62). We showed feature selection methods to influence predictive model performance, describing and evaluating 6 unique methods. Detecting BRAFV600 status in melanoma based on 18F-FDG PET/CT alone does not yet provide clinically relevant knowledge.
Collapse
Affiliation(s)
- Hanna Saadani
- Department of Nuclear Medicine, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Bernies van der Hiel
- Department of Nuclear Medicine, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Else A Aalbersberg
- Department of Nuclear Medicine, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Ioannis Zavrakidis
- Department of Epidemiology and Biostatistics, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - John B A G Haanen
- Department of Medical Oncology, Netherlands Cancer Institute, Amsterdam, The Netherlands; and
| | - Otto S Hoekstra
- Department of Radiology and Nuclear Medicine, Amsterdam UMC, Vrije Universiteit Amsterdam, Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - Ronald Boellaard
- Department of Radiology and Nuclear Medicine, Amsterdam UMC, Vrije Universiteit Amsterdam, Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - Marcel P M Stokkel
- Department of Nuclear Medicine, Netherlands Cancer Institute, Amsterdam, The Netherlands
| |
Collapse
|
90
|
Li S, Ding C, Zhang H, Song J, Wu L. Radiomics for the prediction of EGFR mutation subtypes in non-small cell lung cancer. Med Phys 2019; 46:4545-4552. [PMID: 31376283 DOI: 10.1002/mp.13747] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Revised: 07/08/2019] [Accepted: 07/22/2019] [Indexed: 12/15/2022] Open
Abstract
PURPOSE This retrospective study was designed to investigate the ability of radiomics to predict the mutation status of epidermal growth factor receptor (EGFR) subtypes (19Del and L858R) in patients with non-small cell lung cancer (NSCLC). METHODS In total, 312 patients with NSCLC were included, and 580 radiomic features were extracted from the computed tomography images of each patient. In the training set, univariate analysis was performed on the clinical and radiomic features; logistic regression models were established using a 5-fold cross validation strategy for the prediction of EGFR subtypes 19Del and L858R. Subsequently, the predictive ability of the joint models was evaluated using the test set. RESULTS The results revealed that the radiomic features specific for EGFR 19Del and L858R were Gabor's MTRVariance, Gabor's PTREntropy, and sphericity. Additionally, the respective areas under the receiver operating characteristic curves of the EGFR 19Del and L858R joint models were 0.7925 and 0.7750 for the test set. CONCLUSIONS Our study demonstrated the potential for radiomics to predict EGFR 19Del and L858R. Epidermal growth factor receptor 19Del and L858R exhibited distinct imaging phenotypes, which may help to guide the selection of more accurate and personalized treatment programs for patients with NSCLC.
Collapse
Affiliation(s)
- Shu Li
- School of Medical Informatics, China Medical University, Shenyang, Liaoning, 110122, China
| | - Changwei Ding
- Department of Radiology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, 110004, China
| | - Hao Zhang
- School of Medical Informatics, China Medical University, Shenyang, Liaoning, 110122, China
| | - Jiangdian Song
- School of Medical Informatics, China Medical University, Shenyang, Liaoning, 110122, China
| | - Lei Wu
- College of Information Engineering, Liaoning University of Traditional Chinese Medicine, Shenyang, Liaoning, 110847, China
| |
Collapse
|
91
|
Wang X, Kong C, Xu W, Yang S, Shi D, Zhang J, Du M, Wang S, Bai Y, Zhang T, Chen Z, Ma Z, Wang J, Dong G, Sun M, Yin R, Chen F. Decoding tumor mutation burden and driver mutations in early stage lung adenocarcinoma using CT-based radiomics signature. Thorac Cancer 2019; 10:1904-1912. [PMID: 31414580 PMCID: PMC6775017 DOI: 10.1111/1759-7714.13163] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2019] [Revised: 07/23/2019] [Accepted: 07/23/2019] [Indexed: 12/17/2022] Open
Abstract
Background Tumor mutation burden (TMB) is an important determinant and biomarker for response of targeted therapy and prognosis in patients with lung cancer. The present study aimed to determine whether radiomics signature could non‐invasively predict the TMB status and driver mutations in patients with resectable early stage lung adenocarcinoma (LUAD). Methods A total of 61pulmonary nodules (PNs) from 51 patients post‐operatively diagnosed LUAD were enrolled for analysis. Two datasets were divided according to two‐thirds of patients from different commercial Comprehensive Genomic Profiling (CGP) panels: a training cohort including 41 PNs and a testing cohort including rest 20PNs. We sequenced all tumor specimens and paired blood cells using next generation sequencing (NGS), so as to detect TMB status and somatic mutations. We collected 718 quantitative 3D radiomics features extracted from segmented volumes of each PNs and 78 clinical and pathological features retrieved from medical records as well. Support vector machine methods were performed to establish the predictive model. Results We established an efficient fusion‐positive tumor prediction model that predicts TMB status and EGFR/TP53 mutations of early stage LUAD. The radiomics signature yielded a median AUC value of 0.606, 0.604, and 0.586 respectively. Combining radiomics with the clinical information can further improve the prediction performance, which the median AUC values are 0.671 for TMB, 0.697 and 0.656 for EGFR/TP53 respectively. Conclusion It is feasible and effective to facilitate TMB and somatic driver mutations prediction by using the radiomics signature and NGS data in early stage LUAD.
Collapse
Affiliation(s)
- Xiaoxiao Wang
- Department of Biostatistics, School of Public Health, Nanjing Medical University, Nanjing, China.,Department of GCP Research Center, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of TCM, Nanjing, China
| | - Cheng Kong
- Department of Radiotherapy, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing Medical University Affiliated Cancer Hospital, Nanjing, China
| | - Weizhang Xu
- Department of Thoracic Surgery, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing Medical University Affiliated Cancer Hospital, Nanjing, China.,Department of Oncology, Jiangsu Key Laboratory of Molecular and Translational Cancer Research, Nanjing, China.,Department of The Fourth Clinical College, Nanjing Medical University, Nanjing, China
| | - Sheng Yang
- Department of Biostatistics, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Dan Shi
- Department of Radiology, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of TCM, Nanjing, China
| | - Jingyuan Zhang
- Department of Pathology, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing Medical University Affiliated Cancer Hospital, Nanjing, China
| | - Mulong Du
- Department of Biostatistics, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Siwei Wang
- Department of Thoracic Surgery, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing Medical University Affiliated Cancer Hospital, Nanjing, China.,Department of Oncology, Jiangsu Key Laboratory of Molecular and Translational Cancer Research, Nanjing, China.,Department of The Fourth Clinical College, Nanjing Medical University, Nanjing, China
| | - Yongkang Bai
- Department of Thoracic Surgery, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing Medical University Affiliated Cancer Hospital, Nanjing, China.,Department of Oncology, Jiangsu Key Laboratory of Molecular and Translational Cancer Research, Nanjing, China.,Department of The Fourth Clinical College, Nanjing Medical University, Nanjing, China
| | - Te Zhang
- Department of Thoracic Surgery, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing Medical University Affiliated Cancer Hospital, Nanjing, China.,Department of Oncology, Jiangsu Key Laboratory of Molecular and Translational Cancer Research, Nanjing, China.,Department of The Fourth Clinical College, Nanjing Medical University, Nanjing, China
| | - Zeng Chen
- Department of Information, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing Medical University Affiliated Cancer Hospital, Nanjing, China
| | - Zhifei Ma
- Department of Thoracic Surgery, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing Medical University Affiliated Cancer Hospital, Nanjing, China.,Department of Oncology, Jiangsu Key Laboratory of Molecular and Translational Cancer Research, Nanjing, China
| | - Jie Wang
- Department of Oncology, Jiangsu Key Laboratory of Molecular and Translational Cancer Research, Nanjing, China.,Department of Scientific Research, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing Medical University Affiliated Cancer Hospital, Nanjing, China
| | - Gaochao Dong
- Department of Oncology, Jiangsu Key Laboratory of Molecular and Translational Cancer Research, Nanjing, China.,Department of Scientific Research, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing Medical University Affiliated Cancer Hospital, Nanjing, China
| | - Mengting Sun
- Department of Oncology, Jiangsu Key Laboratory of Molecular and Translational Cancer Research, Nanjing, China.,Department of Scientific Research, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing Medical University Affiliated Cancer Hospital, Nanjing, China
| | - Rong Yin
- Department of Thoracic Surgery, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing Medical University Affiliated Cancer Hospital, Nanjing, China.,Department of Oncology, Jiangsu Key Laboratory of Molecular and Translational Cancer Research, Nanjing, China.,Department of Scientific Research, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing Medical University Affiliated Cancer Hospital, Nanjing, China
| | - Feng Chen
- Department of Biostatistics, School of Public Health, Nanjing Medical University, Nanjing, China.,Department of Oncology, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Nanjing Medical University, Nanjing, China.,Department of Oncology, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, China.,Department of Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China
| |
Collapse
|
92
|
Chen SW, Shen WC, Chen WTL, Hsieh TC, Yen KY, Chang JG, Kao CH. Metabolic Imaging Phenotype Using Radiomics of [ 18F]FDG PET/CT Associated with Genetic Alterations of Colorectal Cancer. Mol Imaging Biol 2019; 21:183-190. [PMID: 29948642 DOI: 10.1007/s11307-018-1225-8] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
PURPOSE To understand the association between genetic mutations and radiomics of 2-deoxy-2-[18F]fluoro-D-glucose ([18F]FDG) positron emission tomography (PET)/x-ray computed tomography (CT) in patients with colorectal cancer (CRC). PROCEDURES This study included 74 CRC patients who had undergone preoperative [18F]FDG PET/CT. A total of 65 PET/CT-related features including intensity, volume-based, histogram, and textural features were calculated. High-resolution melting methods were used for genetic mutation analysis. RESULTS Genetic mutants were found in 21 KRAS tumors (28 %), 31 TP53 tumors (42 %), and 17 APC tumors (23 %). Tumors with a mutated KRAS had an increased value at the 25th percentile of maximal standardized uptake value (SUVmax) within their metabolic tumor volume (MTV) (P < .0001; odds ratio [OR] 1.99; 95 % confidence interval [CI] 1.37-2.90) and their contrast from the gray-level cooccurrence matrix (P = .005; OR 1.52; 95 % CI 1.14-2.04). A mutated TP53 was associated with an increased value of short-run low gray-level emphasis derived from the gray-level run length matrix (P = .001; OR 243006.0; 95 % CI 59.2-996,872,313). APC mutants exhibited lower low gray-level zone emphasis derived from the gray-level zone length matrix (P = .006; OR < .0001; 95 % CI 0.000-0.22). CONCLUSION PET/CT-derived radiomics can provide supplemental information to determine KRAS, TP53, and APC genetic alterations in CRC.
Collapse
Affiliation(s)
- Shang-Wen Chen
- Department of Radiation Oncology, China Medical University Hospital, Taichung, Taiwan.,Graduate Institute of Clinical Medical Science, School of Medicine, College of Medicine, China Medical University, No. 2, Yuh-Der Road, Taichung, 404, Taiwan.,Department of Radiology, School of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Wei-Chih Shen
- Department of Computer Science and Information Engineering, Asia University, Taichung, Taiwan
| | - William Tzu-Liang Chen
- Graduate Institute of Clinical Medical Science, School of Medicine, College of Medicine, China Medical University, No. 2, Yuh-Der Road, Taichung, 404, Taiwan.,Department of Surgery, China Medical University Hospital, Taichung, Taiwan
| | - Te-Chun Hsieh
- Department of Nuclear Medicine and PET Center, China Medical University Hospital, Taichung, Taiwan.,Department of Biomedical Imaging and Radiological Science, China Medical University, Taichung, Taiwan
| | - Kuo-Yang Yen
- Department of Nuclear Medicine and PET Center, China Medical University Hospital, Taichung, Taiwan.,Department of Biomedical Imaging and Radiological Science, China Medical University, Taichung, Taiwan
| | - Jan-Gowth Chang
- Graduate Institute of Clinical Medical Science, School of Medicine, College of Medicine, China Medical University, No. 2, Yuh-Der Road, Taichung, 404, Taiwan.,Department of Laboratory Medicine, Chine Medical University Hospital, Taichung, Taiwan
| | - Chia-Hung Kao
- Graduate Institute of Clinical Medical Science, School of Medicine, College of Medicine, China Medical University, No. 2, Yuh-Der Road, Taichung, 404, Taiwan. .,Department of Nuclear Medicine and PET Center, China Medical University Hospital, Taichung, Taiwan. .,Department of Bioinformatics and Medical Engineering, Asia University, Taichung, Taiwan.
| |
Collapse
|
93
|
Kong Z, Jiang C, Zhu R, Feng S, Wang Y, Li J, Chen W, Liu P, Zhao D, Ma W, Wang Y, Cheng X. 18F-FDG-PET-based radiomics features to distinguish primary central nervous system lymphoma from glioblastoma. NEUROIMAGE-CLINICAL 2019; 23:101912. [PMID: 31491820 PMCID: PMC6702330 DOI: 10.1016/j.nicl.2019.101912] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/07/2019] [Revised: 06/22/2019] [Accepted: 06/25/2019] [Indexed: 12/28/2022]
Abstract
The differential diagnosis of primary central nervous system lymphoma from glioblastoma multiforme (GBM) is essential due to the difference in treatment strategies. This study retrospectively reviewed 77 patients (24 with lymphoma and 53 with GBM) to identify the stable and distinguishable characteristics of lymphoma and GBM in 18F-fluorodeocxyglucose (FDG) positron emission tomography (PET) images using a radiomics approach. Three groups of maps, namely, a standardized uptake value (SUV) map, an SUV map calibrated with the normal contralateral cortex (ncc) activity (SUV/ncc map), and an SUV map calibrated with the normal brain mean (nbm) activity (SUV/nbm map), were generated, and a total of 107 radiomics features were extracted from each SUV map. The margins of the ROI were adjusted to assess the stability of the features, and the area under the curve (AUC) of the receiver operating characteristic curve of each feature was compared with the SUVmax to evaluate the distinguishability of the features. Nighty-five radiomics features from the SUV map were significantly different between lymphoma and GBM, 46 features were numeric stable after marginal adjustment, and 31 features displayed better performance than SUVmax. Features extracted from the SUV map demonstrated higher AUCs than features from the further calibrated maps. Tumors with solid metabolic patterns were also separately evaluated and revealed similar results. Thirteen radiomics features that were stable and distinguishable than SUVmax in every circumstance were selected to distinguish lymphoma from glioblastoma, and they suggested that lymphoma has a higher SUV in most interval segments and is more mathematically heterogeneous than GBM. This study suggested that 18F-FDG-PET-based radiomics is a reliable noninvasive method to distinguish lymphoma and GBM. 18F-FDG-PET is a reliable technique to differentiate primary CNS lymphoma from GBM. Radiomics identify the stable and distinguishable characteristics of lymphoma and GBM. 18F-FDG-PET-based radiomics provide assistance to the preoperative diagnosis.
Collapse
Affiliation(s)
- Ziren Kong
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, No.1 Shuaifuyuan Wangfujing Dongcheng District, Beijing, China
| | - Chendan Jiang
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, No.1 Shuaifuyuan Wangfujing Dongcheng District, Beijing, China
| | - Ruizhe Zhu
- Department of Nuclear Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, No.1 Shuaifuyuan Wangfujing Dongcheng District, Beijing, China
| | - Shi Feng
- Department of Nuclear Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, No.1 Shuaifuyuan Wangfujing Dongcheng District, Beijing, China
| | - Yaning Wang
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, No.1 Shuaifuyuan Wangfujing Dongcheng District, Beijing, China
| | - Jiatong Li
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, No.1 Shuaifuyuan Wangfujing Dongcheng District, Beijing, China; School of Medicine, Tsinghua University, Haidian District, Beijing, China
| | - Wenlin Chen
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, No.1 Shuaifuyuan Wangfujing Dongcheng District, Beijing, China
| | - Penghao Liu
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, No.1 Shuaifuyuan Wangfujing Dongcheng District, Beijing, China
| | - Dachun Zhao
- Department of Pathology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, No.1 Shuaifuyuan Wangfujing Dongcheng District, Beijing, China
| | - Wenbin Ma
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, No.1 Shuaifuyuan Wangfujing Dongcheng District, Beijing, China
| | - Yu Wang
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, No.1 Shuaifuyuan Wangfujing Dongcheng District, Beijing, China
| | - Xin Cheng
- Department of Nuclear Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, No.1 Shuaifuyuan Wangfujing Dongcheng District, Beijing, China.
| |
Collapse
|
94
|
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
|
95
|
Xiong J, Li X, Lu L, Lawrence SH, Fu X, Zhao J, Zhao B. Implementation strategy of a CNN model affects the performance of CT assessment of EGFR mutation status in lung cancer patients. IEEE ACCESS : PRACTICAL INNOVATIONS, OPEN SOLUTIONS 2019; 7:64583-64591. [PMID: 32953368 PMCID: PMC7500487 DOI: 10.1109/access.2019.2916557] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
OBJECTIVE To compare CNN models implemented using different strategies in the CT assessment of EGFR mutation status in patients with lung adenocarcinoma. METHODS 1,010 consecutive lung adenocarcinoma patients with known EGFR mutation status were randomly divided into a training set (n=810) and a testing set (n=200). CNN models were constructed based on ResNet-101 architecture but implemented using different strategies: dimension filters (2D/3D), input sizes (small/middle/large and their fusion), slicing methods (transverse plane only and arbitrary multi-view planes), and training approaches (from scratch and fine-tuning a pre-trained CNN). The performance of the CNN models was compared using AUC. RESULTS The fusion approach yielded consistently better performance than other input sizes, although the effect often did not reach statistical significance. Multi-view slicing was significantly superior to the transverse method when fine-tuning a pre-trained 2D CNN but not a CNN trained from scratch. The 3D CNN was significantly better than the 2D transverse plane method but only marginally better than the multi-view slicing method when trained from scratch. The highest performance (AUC=0.838) was achieved for the fine-tuned 2D CNN model when built using the fusion input size and multi-view slicing method. CONCLUSION The assessment of EGFR mutation status in patients is more accurate when CNN models use more spatial information and are fine-tuned by transfer learning. Our finding about implementation strategy of a CNN model could be a guidance to other medical 3D images applications. Compared with other published studies which used medical images to identify EGFR mutation status, our CNN model achieved the best performance in a biggest patient cohort.
Collapse
Affiliation(s)
- Junfeng Xiong
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, 200240 China
- Department of Radiology, Columbia University Medical Center, NY 10032 USA
| | - Xiaoyang Li
- Department of Radiation Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, 200030 China
| | - Lin Lu
- Department of Radiology, Columbia University Medical Center, NY 10032 USA
| | | | - Xiaolong Fu
- Department of Radiation Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, 200030 China
| | - Jun Zhao
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, 200240 China
| | - Binsheng Zhao
- Department of Radiology, Columbia University Medical Center, NY 10032 USA
| |
Collapse
|
96
|
Zhao W, Yang J, Ni B, Bi D, Sun Y, Xu M, Zhu X, Li C, Jin L, Gao P, Wang P, Hua Y, Li M. Toward automatic prediction of EGFR mutation status in pulmonary adenocarcinoma with 3D deep learning. Cancer Med 2019; 8:3532-3543. [PMID: 31074592 PMCID: PMC6601587 DOI: 10.1002/cam4.2233] [Citation(s) in RCA: 72] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Revised: 04/24/2019] [Accepted: 04/25/2019] [Indexed: 12/24/2022] Open
Abstract
To develop a deep learning system based on 3D convolutional neural networks (CNNs), and to automatically predict EGFR‐mutant pulmonary adenocarcinoma in CT images. A dataset of 579 nodules with EGFR mutation status labels of mutant (Mut) or wild‐type (WT) was retrospectively analyzed. A deep learning system, namely 3D DenseNets, was developed to process 3D patches of nodules from CT data, and learn strong representations with supervised end‐to‐end training. The 3D DenseNets were trained with a training subset of 348 nodules and tuned with a development subset of 116 nodules. A strong data augmentation technique, mixup, was used for better generalization. We evaluated our model on a holdout subset of 115 nodules. An independent public dataset of 37 nodules from the cancer imaging archive (TCIA) was also used to test the generalization of our method. Conventional radiomics analysis was also performed for comparison. Our method achieved promising performance on predicting EGFR mutation status, with AUCs of 75.8% and 75.0% for our holdout test set and public test set, respectively. Moreover, strong relations were found between deep learning feature and conventional radiomics, while deep learning worked through an enhanced radiomics manner, that is, deep learned radiomics (DLR), in terms of robustness, compactness and expressiveness. The proposed deep learning system predicts EGFR‐mutant of lung adenocarcinomas in CT images noninvasively and automatically, indicating its potential to help clinical decision‐making by identifying eligible patients of pulmonary adenocarcinoma for EGFR‐targeted therapy.
Collapse
Affiliation(s)
- Wei Zhao
- Department of Radiology, Huadong Hospital Affiliated to Fudan University, Shanghai, China.,Diagnosis and Treatment Center of Small Lung Nodules, Huadong Hospital, Shanghai, China
| | - Jiancheng Yang
- Department of Electronic Engineering, Shanghai Jiao Tong University, Shanghai, China.,SJTU-UCLA Joint Center for Machine Perception and Inference, Shanghai Jiao Tong University, Shanghai, China.,Diannei Technology, Shanghai, China
| | - Bingbing Ni
- Department of Electronic Engineering, Shanghai Jiao Tong University, Shanghai, China.,SJTU-UCLA Joint Center for Machine Perception and Inference, Shanghai Jiao Tong University, Shanghai, China
| | - Dexi Bi
- Department of Pathology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yingli Sun
- Department of Radiology, Huadong Hospital Affiliated to Fudan University, Shanghai, China
| | | | - Xiaoxia Zhu
- Department of Radiology, Huadong Hospital Affiliated to Fudan University, Shanghai, China
| | - Cheng Li
- Department of Radiology, Huadong Hospital Affiliated to Fudan University, Shanghai, China
| | - Liang Jin
- Department of Radiology, Huadong Hospital Affiliated to Fudan University, Shanghai, China
| | - Pan Gao
- Department of Radiology, Huadong Hospital Affiliated to Fudan University, Shanghai, China
| | - Peijun Wang
- Department of Radiology, School of Medicine, Tongji Hospital, Tongji University, Shanghai, China
| | - Yanqing Hua
- Department of Radiology, Huadong Hospital Affiliated to Fudan University, Shanghai, China
| | - Ming Li
- Department of Radiology, Huadong Hospital Affiliated to Fudan University, Shanghai, China.,Diagnosis and Treatment Center of Small Lung Nodules, Huadong Hospital, Shanghai, China.,Institute of Functional and Molecular Medical Imaging, Fudan University, Shanghai, China
| |
Collapse
|
97
|
Harmon S, Seder CW, Chen S, Traynor A, Jeraj R, Blasberg JD. Quantitative FDG PET/CT may help risk-stratify early-stage non-small cell lung cancer patients at risk for recurrence following anatomic resection. J Thorac Dis 2019; 11:1106-1116. [PMID: 31179052 PMCID: PMC6531752 DOI: 10.21037/jtd.2019.04.46] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Accepted: 04/03/2019] [Indexed: 01/05/2023]
Abstract
BACKGROUND Preoperative identification of non-small cell lung cancer (NSCLC) patients at risk for disease recurrence has proven unreliable. The extraction of quantitative metrics from imaging based on tumor intensity and texture may enhanced disease characterization. This study evaluated tumor-specific 18F-fluorodeoxyglucose (18F-FDG) positron emission tomography/computerized tomography (PET/CT) uptake patterns and their association with disease recurrence in early-stage NSCLC. METHODS Sixty-four stage I/II NSCLC patients who underwent anatomic resection between 2001 and 2014 were examined. Pathologically or radiographic confirmed disease recurrence within 5 years of resection comprised the study group. Quantitative imaging metrics were extracted within the primary tumor volume. Squamous cell carcinoma (SCC) (N=27) and adenocarcinoma (AC) (N=41) patients were compared using a Wilcoxon signed-rank test. Associations between imaging and clinical variables with 5-year disease-free survival (DFS) and overall survival (OS) were evaluated by Cox proportional-hazards regression. RESULTS Clinical and pathologic characteristics were similar between recurrence (N=34) and patients achieving 5-year DFS (N=30). Standardized uptake value (SUV)max and SUVmean varied significantly by histology, with SCC demonstrating higher uptake intensity and heterogeneity patterns. Entropy-grey-level co-occurrence matrix (GLCM) was a significant univariate predictor of DFS (HR =0.72, P=0.04) and OS (HR =0.65, P=0.007) independent of histology. Texture features showed higher predictive ability for DFS in SCC than AC. Pathologic node status and staging classification were the strongest clinical predictors of DFS, independent of histology. CONCLUSIONS Several imaging metrics correlate with increased risk for disease recurrence in early-stage NSCLC. The predictive ability of imaging was strongest when patients are stratified by histology. The incorporation of 18F-FDG PET/CT texture features with preoperative risk factors and tumor characteristics may improve identification of high-risk patients.
Collapse
Affiliation(s)
- Stephanie Harmon
- Department of Medical Physics, University of Wisconsin, Madison, WI, USA
| | - Christopher W. Seder
- Department of Thoracic and Cardiovascular Surgery, Rush University Medical Center, Chicago, IL, USA
| | - Song Chen
- Department of Medical Physics, University of Wisconsin, Madison, WI, USA
- Department of Nuclear Medicine, The 1st Hospital of China Medical University, Shenyang 110016, China
| | - Anne Traynor
- Department of Medical Physics, University of Wisconsin, Madison, WI, USA
| | - Robert Jeraj
- Department of Medical Physics, University of Wisconsin, Madison, WI, USA
| | | |
Collapse
|
98
|
Tu W, Sun G, Fan L, Wang Y, Xia Y, Guan Y, Li Q, Zhang D, Liu S, Li Z. Radiomics signature: A potential and incremental predictor for EGFR mutation status in NSCLC patients, comparison with CT morphology. Lung Cancer 2019; 132:28-35. [PMID: 31097090 DOI: 10.1016/j.lungcan.2019.03.025] [Citation(s) in RCA: 106] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2018] [Revised: 03/04/2019] [Accepted: 03/25/2019] [Indexed: 02/06/2023]
Abstract
OBJECTIVES To compare the predictive performance of radiomics signature and CT morphological features for epidermal growth factor receptor (EGFR) mutation status; then further to develop and compare the different predictive models for EGFR mutation in non-small cell lung cancer (NSCLC) patients. MATERIALS AND METHODS This retrospective study involved 404 patients with NSCLC (243 cases in the training cohort and 161 cases in the validation cohort). Radiomics features were extracted from preoperative non-contrast CT images of the entire tumor. Correlations between the EGFR mutation status and candidate predictors were assessed using Mann-Whitney U test or Chi-square test. Unsupervised consensus clustering was used to analyze the representativeness and reduce the redundancy of radiomics features. Multivariable logistic regression analysis was performed to build radiomics signature and develop predictive models of EGFR mutation. ROC curve analysis and Delong test were used to compare the predictive performance among individual features and models. RESULTS Of the 234 radiomics features, 93 radiomics features with high repeatability and high predictive significance were selected. The radiomics signature, which was built with one histogram and two textural features, showed the best predictive performance (AUC = 0.762 and 0.775 in the training and validation cohort) in comparison with all the clinical characteristics and conventional CT morphological features to differentiate EGFR mutation status (P < 0.05). The integrated model was developed with maximum diameter, location, sex and radiomics signature. In the training and validation cohort, the integrated model showed the most optimal predictive performance (AUC = 0.798, 0.818 in the training and validation cohort) compared with the clinical models. CONCLUSION The radiomics signature showed better performance for predicting EGFR mutant than all the clinical and morphological features. Moreover, the integrated model built with radiomics signature, clinical and morphological features outperformed the clinical models, which is helpful for physicians to determine the targeted therapy.
Collapse
Affiliation(s)
- Wenting Tu
- Department of Radiology, Changzheng Hospital, Second Military Medical University, No. 415 Fengyang Road, Shanghai 200003, China
| | - Guangyuan Sun
- Department of Thoracic and Cardiovascular Surgery, Changzheng Hospital, Second Military Medical University, No. 415 Fengyang Road, Shanghai 200003, China
| | - Li Fan
- Department of Radiology, Changzheng Hospital, Second Military Medical University, No. 415 Fengyang Road, Shanghai 200003, China.
| | - Yun Wang
- Department of Radiology, Changzheng Hospital, Second Military Medical University, No. 415 Fengyang Road, Shanghai 200003, China
| | - Yi Xia
- Department of Radiology, Changzheng Hospital, Second Military Medical University, No. 415 Fengyang Road, Shanghai 200003, China
| | - Yu Guan
- Department of Radiology, Changzheng Hospital, Second Military Medical University, No. 415 Fengyang Road, Shanghai 200003, China
| | - Qiong Li
- Department of Radiology, Changzheng Hospital, Second Military Medical University, No. 415 Fengyang Road, Shanghai 200003, China
| | - Di Zhang
- Department of Radiology, Changzheng Hospital, Second Military Medical University, No. 415 Fengyang Road, Shanghai 200003, China
| | - Shiyuan Liu
- Department of Radiology, Changzheng Hospital, Second Military Medical University, No. 415 Fengyang Road, Shanghai 200003, China
| | - Zhaobin Li
- Department of Radiation Oncology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, No. 600 Yishan Road, Shanghai 200233, China.
| |
Collapse
|
99
|
|
100
|
Perspectives in Radiomics for Personalized Medicine and Theranostics. Nucl Med Mol Imaging 2019; 53:164-166. [PMID: 31231435 DOI: 10.1007/s13139-019-00578-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2019] [Revised: 01/11/2019] [Accepted: 01/13/2019] [Indexed: 01/09/2023] Open
Abstract
Radiomics handles imaging biomarker from high-throughput feature extraction through complex pattern recognition that is difficult for human to process. Recent medical paradigms are rapidly changing to personalized medicine, including molecular targeted therapy, immunotherapy, and theranostics, and the importance of biomarkers for these is growing day by day. Even though biopsy continues to gold standard for tumor assessment in personalized medicine, imaging is expected to complement biopsy because it allows whole tumor evaluation, whole body evaluation, and non-invasive and repetitive evaluation. Radiomics is known as a useful method to get imaging biomarkers related to intratumor heterogeneity in molecular targeted therapy as well as one-size-fits-all therapy. It is also expected to be useful in new paradigms such as immunotherapy and somatostatin receptor (SSTR) or prostate-specific membrane antigen (PSMA)-targeted theranostics. Radiomics research should move to multimodality (CT, MR, PET, etc.), multicenter, and prospective studies from current single modality, single institution, and retrospective studies. Image-quality harmonization, intertumor heterogeneity, and integrative analysis of information from different scales are thought to be important keywords in future radiomics research. It is clear that radiomics will play an important role in personalized medicine.
Collapse
|