1
|
Xie Z, Zhang Q, Zhang R, Zhao Y, Zhang W, Song Y, Yu D, Lin J, Li X, Suo S, Zhou Y. Identification of D842V mutation in gastrointestinal stromal tumors based on CT radiomics: a multi-center study. Cancer Imaging 2024; 24:169. [PMID: 39707515 DOI: 10.1186/s40644-024-00815-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Accepted: 12/10/2024] [Indexed: 12/23/2024] Open
Abstract
BACKGROUND Gastrointestinal stromal tumors (GISTs) are the most common mesenchymal tumors of the gastrointestinal tract. Recent advent of tyrosine kinase inhibitors (TKIs) has significantly improved the prognosis of GIST patients. However, responses to TKI therapy can vary depending on the specific gene mutation. D842V, which is the most common mutation in platelet-derived growth factor receptor alpha exon 18, shows no response to imatinib and sunitinib. Radiomics features based on venous-phase contrast-enhanced computed tomography (CECT) have shown potential in non-invasive prediction of GIST genotypes. This study sought to determine whether radiomics features could help distinguish GISTs with D842V mutations. METHODS A total of 872 pathologically confirmed GIST patients with CECT data available from three independent centers were included and divided into the training cohort ( n = 487 ) and the external validation cohort ( n = 385 ). Clinical features including age, sex, tumor size and location were collected. Radiomics features on the largest axial image of venous-phase CECT were analyzed and a total of two radiomics features were selected after feature selection. Random forest models trained on non-radiomics features only (the non-radiomics model) and on both non-radiomics and radiomics features (the combined model) were compared. RESULTS The combined model showed better average precision (0.250 vs. 0.102, p = 0.039) and F1 score (0.253 vs. 0.155, p = 0.012) than the non-radiomics model. There was no significant difference in ROC-AUC (0.728 vs. 0.737, p = 0.836) and geometric mean (0.737 vs. 0.681, p = 0.352). CONCLUSIONS This study demonstrated the potential of radiomics features based on venous-phase CECT images to identify D842V mutation in GISTs. Our model may provide an alternative approach to guide TKI therapy for patients inaccessible to sequence variant testing, potentially improving treatment outcomes for GIST patients especially in resource-limited settings.
Collapse
Affiliation(s)
- Zhenhui Xie
- Department of Radiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Pujian Road 160, Pudong District, 200127, Shanghai, China
| | - Qingwei Zhang
- Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Institute of Digestive Disease, Pujian Road 160, Pudong District, 200127, Shanghai, China
| | - Ranying Zhang
- Department of Radiology, Zhongshan Hospital, Fudan University, and Shanghai Institute of Medical Imaging, 108 Fenglin Road, 200032, Shanghai, China
| | - Yuxuan Zhao
- Department of Radiology, Qilu Hospital of Shandong University, 107 Wenhuaxi Road, 250012, Jinan, Shandong, China
| | - Wang Zhang
- Department of Radiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Pujian Road 160, Pudong District, 200127, Shanghai, China
| | - Yang Song
- MR Research Collaboration Team, Siemens Healthineers Ltd., 399 Haiyang West Road, 200126, Shanghai, China
| | - Dexin Yu
- Department of Radiology, Qilu Hospital of Shandong University, 107 Wenhuaxi Road, 250012, Jinan, Shandong, China.
| | - Jiang Lin
- Department of Radiology, Zhongshan Hospital, Fudan University, and Shanghai Institute of Medical Imaging, 108 Fenglin Road, 200032, Shanghai, China.
| | - Xiaobo Li
- Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Institute of Digestive Disease, Pujian Road 160, Pudong District, 200127, Shanghai, China.
| | - Shiteng Suo
- Department of Radiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Pujian Road 160, Pudong District, 200127, Shanghai, China.
| | - Yan Zhou
- Department of Radiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Pujian Road 160, Pudong District, 200127, Shanghai, China.
| |
Collapse
|
2
|
Yin XN, Wang ZH, Zou L, Yang CW, Shen CY, Liu BK, Yin Y, Liu XJ, Zhang B. Computed tomography radiogenomics: A potential tool for prediction of molecular subtypes in gastric stromal tumor. World J Gastrointest Oncol 2024; 16:1296-1308. [PMID: 38660646 PMCID: PMC11037038 DOI: 10.4251/wjgo.v16.i4.1296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2023] [Revised: 01/23/2024] [Accepted: 02/25/2024] [Indexed: 04/10/2024] Open
Abstract
BACKGROUND Preoperative knowledge of mutational status of gastrointestinal stromal tumors (GISTs) is essential to guide the individualized precision therapy. AIM To develop a combined model that integrates clinical and contrast-enhanced computed tomography (CE-CT) features to predict gastric GISTs with specific genetic mutations, namely KIT exon 11 mutations or KIT exon 11 codons 557-558 deletions. METHODS A total of 231 GIST patients with definitive genetic phenotypes were divided into a training dataset and a validation dataset in a 7:3 ratio. The models were constructed using selected clinical features, conventional CT features, and radiomics features extracted from abdominal CE-CT images. Three models were developed: ModelCT sign, modelCT sign + rad, and model CTsign + rad + clinic. The diagnostic performance of these models was evaluated using receiver operating characteristic (ROC) curve analysis and the Delong test. RESULTS The ROC analyses revealed that in the training cohort, the area under the curve (AUC) values for modelCT sign, modelCT sign + rad, and modelCT sign + rad + clinic for predicting KIT exon 11 mutation were 0.743, 0.818, and 0.915, respectively. In the validation cohort, the AUC values for the same models were 0.670, 0.781, and 0.811, respectively. For predicting KIT exon 11 codons 557-558 deletions, the AUC values in the training cohort were 0.667, 0.842, and 0.720 for modelCT sign, modelCT sign + rad, and modelCT sign + rad + clinic, respectively. In the validation cohort, the AUC values for the same models were 0.610, 0.782, and 0.795, respectively. Based on the decision curve analysis, it was determined that the modelCT sign + rad + clinic had clinical significance and utility. CONCLUSION Our findings demonstrate that the combined modelCT sign + rad + clinic effectively distinguishes GISTs with KIT exon 11 mutation and KIT exon 11 codons 557-558 deletions. This combined model has the potential to be valuable in assessing the genotype of GISTs.
Collapse
Affiliation(s)
- Xiao-Nan Yin
- Gastric Cancer Research Center, West China Hospital, Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Zi-Hao Wang
- Gastric Cancer Research Center, West China Hospital, Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Li Zou
- Department of Paediatric Surgery, West China Hospital of Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Cai-Wei Yang
- Department of Radiology, West China Hospital, Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Chao-Yong Shen
- Gastric Cancer Research Center, West China Hospital, Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Bai-Ke Liu
- Gastric Cancer Research Center, West China Hospital, Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Yuan Yin
- Gastric Cancer Research Center, West China Hospital, Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Xi-Jiao Liu
- Department of Radiology, West China Hospital, Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Bo Zhang
- Department of Gastrointestinal Surgery, Sichuan University West China Hospital, Chengdu 610041, Sichuan Province, China
| |
Collapse
|
3
|
Barat M, Pellat A, Dohan A, Hoeffel C, Coriat R, Soyer P. CT and MRI of Gastrointestinal Stromal Tumors: New Trends and Perspectives. Can Assoc Radiol J 2024; 75:107-117. [PMID: 37386745 DOI: 10.1177/08465371231180510] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/01/2023] Open
Abstract
Gastrointestinal stromal tumors (GISTs) are defined as mesenchymal tumors of the gastrointestinal tract that express positivity for CD117, which is a c-KIT proto-oncogene antigen. Expression of the c-KIT protein, a tyrosine kinase growth factor receptor, allows the distinction between GISTs and other mesenchymal tumors such as leiomyoma, leiomyosarcoma, schwannoma and neurofibroma. GISTs can develop anywhere in the gastrointestinal tract, as well as in the mesentery and omentum. Over the years, the management of GISTs has improved due to a better knowledge of their behaviors and risk or recurrence, the identification of specific mutations and the use of targeted therapies. This has resulted in a better prognosis for patients with GISTs. In parallel, imaging of GISTs has been revolutionized by tremendous progress in the field of detection, characterization, survival prediction and monitoring during therapy. Recently, a particular attention has been given to radiomics for the characterization of GISTs using analysis of quantitative imaging features. In addition, radiomics has currently many applications that are developed in conjunction with artificial intelligence with the aim of better characterizing GISTs and providing a more precise assessment of tumor burden. This article sums up recent advances in computed tomography and magnetic resonance imaging of GISTs in the field of image/data acquisition, tumor detection, tumor characterization, treatment response evaluation, and preoperative planning.
Collapse
Affiliation(s)
- Maxime Barat
- Department of Radiology, Hopital Cochin, Paris, France
- Université Paris Cité, Faculté de Médecine, Paris, France
| | - Anna Pellat
- Université Paris Cité, Faculté de Médecine, Paris, France
- Department of Gastroenterology and Digestive Oncology, Hôpital Cochin, Paris, France
| | - Anthony Dohan
- Department of Radiology, Hopital Cochin, Paris, France
- Université Paris Cité, Faculté de Médecine, Paris, France
| | - Christine Hoeffel
- Reims Medical School, Department of Radiology, Hopital Robert Debré, CHU Reims, Université Champagne-Ardennes, Reims, France
| | - Romain Coriat
- Université Paris Cité, Faculté de Médecine, Paris, France
- Department of Gastroenterology and Digestive Oncology, Hôpital Cochin, Paris, France
| | - Philippe Soyer
- Department of Radiology, Hopital Cochin, Paris, France
- Université Paris Cité, Faculté de Médecine, Paris, France
| |
Collapse
|
4
|
Cappello G, Giannini V, Cannella R, Tabone E, Ambrosini I, Molea F, Damiani N, Landolfi I, Serra G, Porrello G, Gozzo C, Incorvaia L, Badalamenti G, Grignani G, Merlini A, D’Ambrosio L, Bartolotta TV, Regge D. A mutation-based radiomics signature predicts response to imatinib in Gastrointestinal Stromal Tumors (GIST). Eur J Radiol Open 2023; 11:100505. [PMID: 37484979 PMCID: PMC10362081 DOI: 10.1016/j.ejro.2023.100505] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/25/2023] Open
Abstract
Objectives To develop a mutation-based radiomics signature to predict response to imatinib in Gastrointestinal Stromal Tumors (GISTs). Methods Eighty-two patients with GIST were enrolled in this retrospective study, including 52 patients from one center that were used to develop the model, and 30 patients from a second center to validate it. Reference standard was the mutational status of tyrosine-protein kinase (KIT) and platelet-derived growth factor α (PDGFRA). Patients were dichotomized in imatinib sensitive (group 0 - mutation in KIT or PDGFRA, different from exon 18-D842V), and imatinib non-responsive (group 1 - PDGFRA exon 18-D842V mutation or absence of mutation in KIT/PDGFRA). Initially, 107 texture features were extracted from the tumor masks of baseline computed tomography scans. Different machine learning methods were then implemented to select the best combination of features for the development of the radiomics signature. Results The best performance was obtained with the 5 features selected by the ANOVA model and the Bayes classifier, using a threshold of 0.36. With this setting the radiomics signature had an accuracy and precision for sensitive patients of 82 % (95 % CI:60-95) and 90 % (95 % CI:73-97), respectively. Conversely, a precision of 80 % (95 % CI:34-97) was obtained in non-responsive patients using a threshold of 0.9. Indeed, with the latter setting 4 patients out of 5 were correctly predicted as non-responders. Conclusions The results are a first step towards using radiomics to improve the management of patients with GIST, especially when tumor tissue is unavailable for molecular analysis or when molecular profiling is inconclusive.
Collapse
Affiliation(s)
- Giovanni Cappello
- Department of Radiology, Candiolo Cancer Institute, FPO-IRCCS, Strada Provinciale 142 Km 3.95, Candiolo, Turin 10060, Italy
| | - Valentina Giannini
- Department of Radiology, Candiolo Cancer Institute, FPO-IRCCS, Strada Provinciale 142 Km 3.95, Candiolo, Turin 10060, Italy
- Department of Surgical Sciences, University of Turin, Turin 10124, Italy
| | - Roberto Cannella
- Section of Radiology - Department of Biomedicine, Neuroscience and Advanced Diagnostics (BiND), University of Palermo, Via del Vespro 129, Palermo 90127, Italy
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, Italy
| | - Emanuele Tabone
- Department of Radiology, Candiolo Cancer Institute, FPO-IRCCS, Strada Provinciale 142 Km 3.95, Candiolo, Turin 10060, Italy
| | - Ilaria Ambrosini
- Department of Translational Research, Academic Radiology, University of Pisa, Italy
- Previously at Division of Medical Oncology, Candiolo Cancer Institute, FPO-IRCCS, Strada Provinciale 142 Km 3.95, Candiolo, Turin 10060, Italy
| | - Francesca Molea
- Department of Radiology, Candiolo Cancer Institute, FPO-IRCCS, Strada Provinciale 142 Km 3.95, Candiolo, Turin 10060, Italy
- Department of Surgical Sciences, University of Turin, Turin 10124, Italy
| | - Nicolò Damiani
- Department of Radiology, Candiolo Cancer Institute, FPO-IRCCS, Strada Provinciale 142 Km 3.95, Candiolo, Turin 10060, Italy
- Department of Surgical Sciences, University of Turin, Turin 10124, Italy
| | - Ilenia Landolfi
- Department of Radiology, Candiolo Cancer Institute, FPO-IRCCS, Strada Provinciale 142 Km 3.95, Candiolo, Turin 10060, Italy
- Department of Surgical Sciences, University of Turin, Turin 10124, Italy
| | - Giovanni Serra
- Department of Surgical Sciences, University of Turin, Turin 10124, Italy
| | - Giorgia Porrello
- Section of Radiology - Department of Biomedicine, Neuroscience and Advanced Diagnostics (BiND), University of Palermo, Via del Vespro 129, Palermo 90127, Italy
| | - Cecilia Gozzo
- Department of Radiology, Humanitas, Istituto Clinico Catanese, Catania, Italy
| | - Lorena Incorvaia
- Department of Surgical, Oncological, and Oral Sciences, Section of Medical Oncology, University of Palermo, Palermo, Italy
| | - Giuseppe Badalamenti
- Department of Surgical, Oncological, and Oral Sciences, Section of Medical Oncology, University of Palermo, Palermo, Italy
| | - Giovanni Grignani
- Division of Medical Oncology, Candiolo Cancer Institute, FPO-IRCCS, Strada Provinciale 142 Km 3.95, Candiolo, Turin 10060, Italy
| | - Alessandra Merlini
- Division of Medical Oncology, Candiolo Cancer Institute, FPO-IRCCS, Strada Provinciale 142 Km 3.95, Candiolo, Turin 10060, Italy
- Department of Oncology, University of Turin, Regione Gonzole 10, Orbassano, Turin 10043, Italy
| | - Lorenzo D’Ambrosio
- Department of Oncology, University of Turin, Regione Gonzole 10, Orbassano, Turin 10043, Italy
- Medical Oncology, University Hospital S. Luigi Gonzaga, regione Gonzole 10, Orbassano, Turin 10043, Italy
- Previously at Division of Medical Oncology, Candiolo Cancer Institute, FPO-IRCCS, Strada Provinciale 142 Km 3.95, Candiolo, Turin 10060, Italy
| | - Tommaso Vincenzo Bartolotta
- Section of Radiology - Department of Biomedicine, Neuroscience and Advanced Diagnostics (BiND), University of Palermo, Via del Vespro 129, Palermo 90127, Italy
- Department of Radiology, Fondazione Istituto Giuseppe Giglio, Ct.da Pietrapollastra, Via Pisciotto, Cefalù, Palermo 90015, Italy
| | - Daniele Regge
- Department of Radiology, Candiolo Cancer Institute, FPO-IRCCS, Strada Provinciale 142 Km 3.95, Candiolo, Turin 10060, Italy
- Department of Surgical Sciences, University of Turin, Turin 10124, Italy
| |
Collapse
|
5
|
Zhang QW, Zhang RY, Yan ZB, Zhao YX, Wang XY, Jin JZ, Qiu QX, Chen JJ, Xie ZH, Lin J, Cao H, Zhou Y, Chen HM, Li XB. Personalized radiomics signature to screen for KIT-11 mutation genotypes among patients with gastrointestinal stromal tumors: a retrospective multicenter study. J Transl Med 2023; 21:726. [PMID: 37845765 PMCID: PMC10577986 DOI: 10.1186/s12967-023-04520-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Accepted: 09/11/2023] [Indexed: 10/18/2023] Open
Abstract
OBJECTIVES Gastrointestinal stromal tumors (GISTs) carrying different KIT exon 11 (KIT-11) mutations exhibit varying prognoses and responses to Imatinib. Herein, we aimed to determine whether computed tomography (CT) radiomics can accurately stratify KIT-11 mutation genotypes to benefit Imatinib therapy and GISTs monitoring. METHODS Overall, 1143 GISTs from 3 independent centers were separated into a training cohort (TC) or validation cohort (VC). In addition, the KIT-11 mutation genotype was classified into 4 categories: no KIT-11 mutation (K11-NM), point mutations or duplications (K11-PM/D), KIT-11 557/558 deletions (K11-557/558D), and KIT-11 deletion without codons 557/558 involvement (K11-D). Subsequently, radiomic signatures (RS) were generated based on the arterial phase of contrast CT, which were then developed as KIT-11 mutation predictors using 1408 quantitative image features and LASSO regression analysis, with further evaluation of its predictive capability. RESULTS The TC AUCs for K11-NM, K11-PM/D, K11-557/558D, and K11-D ranged from 0.848 (95% CI 0.812-0.884), 0.759 (95% CI 0.722-0.797), 0.956 (95% CI 0.938-0.974), and 0.876 (95% CI 0.844-0.908), whereas the VC AUCs ranged from 0.723 (95% CI 0.660-0.786), 0.688 (95% CI 0.643-0.732), 0.870 (95% CI 0.824-0.918), and 0.830 (95% CI 0.780-0.878). Macro-weighted AUCs for the KIT-11 mutant genotype ranged from 0.838 (95% CI 0.820-0.855) in the TC to 0.758 (95% CI 0.758-0.784) in VC. TC had an overall accuracy of 0.694 (95%CI 0.660-0.729) for RS-based predictions of the KIT-11 mutant genotype, whereas VC had an accuracy of 0.637 (95%CI 0.595-0.679). CONCLUSIONS CT radiomics signature exhibited good predictive performance in estimating the KIT-11 mutation genotype, especially in prediction of K11-557/558D genotype. RS-based classification of K11-NM, K11-557/558D, and K11-D patients may be an indication for choice of Imatinib therapy.
Collapse
Affiliation(s)
- Qing-Wei Zhang
- Division of Gastroenterology and Hepatology, Shanghai Institute of Digestive Disease, NHC Key Laboratory of Digestive Diseases, State Key Laboratory for Oncogenes and Related Genes, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Ran-Ying Zhang
- Department of Radiology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Zhi-Bo Yan
- Department of General Surgery, Qilu Hospital of Shandong University, Jinan, China
| | - Yu-Xuan Zhao
- Department of Radiology, Qilu Hospital of Shandong University, Jinan, China
| | - Xin-Yuan Wang
- Division of Gastroenterology and Hepatology, Shanghai Institute of Digestive Disease, NHC Key Laboratory of Digestive Diseases, State Key Laboratory for Oncogenes and Related Genes, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Jing-Zheng Jin
- Division of Gastroenterology and Hepatology, Shanghai Institute of Digestive Disease, NHC Key Laboratory of Digestive Diseases, State Key Laboratory for Oncogenes and Related Genes, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Qi-Xuan Qiu
- Department of Radiology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Jie-Jun Chen
- Department of Radiology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Zhen-Hui Xie
- Department of Radiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, No. 160, Pujian Rd., Shanghai, 200127, China
| | - Jiang Lin
- Department of Radiology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
| | - Hui Cao
- Department of Gastrointestinal Surgery, Renji Hospital, Shanghai Jiao Tong University School of Medicine, No. 160 Pujian Road, Shanghai, 200127, China.
| | - Yan Zhou
- Department of Radiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, No. 160, Pujian Rd., Shanghai, 200127, China.
| | - Hui-Min Chen
- Division of Gastroenterology and Hepatology, Shanghai Institute of Digestive Disease, NHC Key Laboratory of Digestive Diseases, State Key Laboratory for Oncogenes and Related Genes, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
| | - Xiao-Bo Li
- Division of Gastroenterology and Hepatology, Shanghai Institute of Digestive Disease, NHC Key Laboratory of Digestive Diseases, State Key Laboratory for Oncogenes and Related Genes, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
| |
Collapse
|
6
|
Guo C, Zhou H, Chen X, Feng Z. Computed tomography texture-based models for predicting KIT exon 11 mutation of gastrointestinal stromal tumors. Heliyon 2023; 9:e20983. [PMID: 37876490 PMCID: PMC10590931 DOI: 10.1016/j.heliyon.2023.e20983] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 10/11/2023] [Accepted: 10/12/2023] [Indexed: 10/26/2023] Open
Abstract
Background KIT exon 11 mutation in gastrointestinal stromal tumors (GISTs) is associated with treatment strategies. However, few studies have shown the role of imaging-based texture analysis in KIT exon 11 mutation in GISTs. In this study, we aimed to show the association between computed tomography (CT)-based texture features and KIT exon 11 mutation. Methods Ninety-five GISTs confirmed by surgery and identified with mutational genotype of KIT were included in this study. By amplifying the samples using over-sampling technique, a total of 183 region of interest (ROI) segments were extracted from 63 patients as training cohort. The 63 new ROI segments were extracted from the 63 patients as internal validation cohort. Thirty-two patients who underwent KIT exon 11 mutation test during 2021-2023 was selected as external validation cohort. The textural parameters were evaluated both in training cohort and validation cohort. Least absolute shrinkage and selection operator (LASSO) algorithms and logistic regression analysis were used to select the discriminant features. Results Three of textural features were obtained using LASSO analysis. Logistic regression analysis showed that patients' age, tumor location and radiomics features were significantly associated with KIT exon 11 mutation (p < 0.05). A nomogram was developed based on the associated factors. The area under the curve (AUC) of clinical features, radiomics features and their combination in training cohort was 0.687 (95 % CI: 0.604-0.771), 0.829 (95 % CI: 0.768-0.890) and 0.874 (95 % CI: 0.822-0.926), respectively. The AUC of radiomics features in internal validation cohort and external cohort was 0.880 (95 % CI: 0.796-0.964) and 0.827 (95%CI: 0.667-0.987), respectively. Conclusion The CT texture-based model can be used to predict KIT exon 11 mutation in GISTs.
Collapse
Affiliation(s)
- Chuangen Guo
- Department of Radiology, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Road, Hangzhou, 310003, China
| | - Hao Zhou
- Department of Radiology, The Affiliated Hospital of Nanjing University of Chinese Medicine, 155 hanzhong Road, Nanjing, 210029, China
| | - Xiao Chen
- Department of Radiology, The Affiliated Hospital of Nanjing University of Chinese Medicine, 155 hanzhong Road, Nanjing, 210029, China
| | - Zhan Feng
- Department of Radiology, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Road, Hangzhou, 310003, China
| |
Collapse
|
7
|
Galluzzo A, Boccioli S, Danti G, De Muzio F, Gabelloni M, Fusco R, Borgheresi A, Granata V, Giovagnoni A, Gandolfo N, Miele V. Radiomics in gastrointestinal stromal tumours: an up-to-date review. Jpn J Radiol 2023; 41:1051-1061. [PMID: 37171755 DOI: 10.1007/s11604-023-01441-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Accepted: 04/29/2023] [Indexed: 05/13/2023]
Abstract
Gastrointestinal stromal tumours are rare mesenchymal neoplasms originating from the Cajal cells and represent the most common sarcomas in the gastroenteric tract. Symptoms may be absent or non-specific, ranging from fatigue and weight loss to acute abdomen. Nowadays endoscopy, echoendoscopy, contrast-enhanced computed tomography, magnetic resonance imaging and positron emission tomography are the main methods for diagnosis. Because of their rarity, these neoplasms may not be included immediately in the differential diagnosis of a solitary abdominal mass. Radiomics is an emerging technique that can extract medical imaging information, not visible to the human eye, transforming it into quantitative data. The purpose of this review is to demonstrate how radiomics can improve the already known imaging techniques by providing useful tools for the diagnosis, treatment, and prognosis of these tumours.
Collapse
Affiliation(s)
- Antonio Galluzzo
- Department of Radiology, Careggi University Hospital, Largo Brambilla 3, 50134, Florence, Italy
| | - Sofia Boccioli
- Department of Radiology, Careggi University Hospital, Largo Brambilla 3, 50134, Florence, Italy
| | - Ginevra Danti
- Department of Radiology, Careggi University Hospital, Largo Brambilla 3, 50134, Florence, Italy.
| | - Federica De Muzio
- Department of Medicine and Health Sciences V. Tiberio, University of Molise, 86100, Campobasso, Italy
| | - Michela Gabelloni
- Department of Translational Research, Diagnostic and Interventional Radiology, University of Pisa, Pisa, Italy
| | - Roberta Fusco
- Medical Oncology Division, Igea SpA, 80013, Naples, Italy
| | - Alessandra Borgheresi
- Department of Clinical, Special and Dental Sciences, University Politecnica Delle Marche, Via Conca 71, 60126, Ancona, Italy
- Department of Radiology, University Hospital "Azienda Ospedaliera Universitaria Delle Marche", Via Conca 71, 60126, Ancona, Italy
| | - Vincenza Granata
- Department of Radiology, "Istituto Nazionale Tumori IRCCS Fondazione, Pascale-IRCCS di Napoli", 80131, Naples, Italy
| | - Andrea Giovagnoni
- Department of Clinical, Special and Dental Sciences, University Politecnica Delle Marche, Via Conca 71, 60126, Ancona, Italy
- Department of Radiology, University Hospital "Azienda Ospedaliera Universitaria Delle Marche", Via Conca 71, 60126, Ancona, Italy
| | - Nicoletta Gandolfo
- Diagnostic Imaging Department, Villa Scassi Hospital-ASL 3, Corso Scassi 1, 16149, Genoa, Italy
- Italian Society of Medical and Interventional Radiology (SIRM), SIRM Foundation, Via Della Signora 2, 20122, Milan, Italy
| | - Vittorio Miele
- Department of Radiology, Careggi University Hospital, Largo Brambilla 3, 50134, Florence, Italy
| |
Collapse
|
8
|
Zhang Y, Yue X, Zhang P, Zhang Y, Wu L, Diao N, Ma G, Lu Y, Ma L, Tao K, Li Q, Han P. Clinical-radiomics-based treatment decision support for KIT Exon 11 deletion in gastrointestinal stromal tumors: a multi-institutional retrospective study. Front Oncol 2023; 13:1193010. [PMID: 37645430 PMCID: PMC10461453 DOI: 10.3389/fonc.2023.1193010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Accepted: 07/26/2023] [Indexed: 08/31/2023] Open
Abstract
Objective gastrointestinal stromal tumors (GISTs) with KIT exon 11 deletions have more malignant clinical outcomes. A radiomics model was constructed for the preoperative prediction of KIT exon 11 deletion in GISTs. Methods Overall, 126 patients with GISTs who underwent preoperative enhanced CT were included. GISTs were manually segmented using ITK-SNAP in the arterial phase (AP) and portal venous phase (PVP) images of enhanced CT. Features were extracted using Anaconda (version 4.2.0) with PyRadiomics. Radiomics models were constructed by LASSO. The clinical-radiomics model (combined model) was constructed by combining the clinical model with the best diagnostic effective radiomics model. ROC curves were used to compare the diagnostic effectiveness of radiomics model, clinical model, and combined model. Diagnostic effectiveness among radiomics model, clinical model and combine model were analyzed in external cohort (n=57). Statistics were carried out using R 3.6.1. Results The Radscore showed favorable diagnostic efficacy. Among all radiomics models, the AP-PVP radiomics model exhibited excellent performance in the training cohort, with an AUC of 0.787 (95% CI: 0.687-0.866), which was verified in the test cohort (AUC=0.775, 95% CI: 0.608-0.895). Clinical features were also analyzed. Among the radiomics, clinical and combined models, the combined model showed favorable diagnostic efficacy in the training (AUC=0.863) and test cohorts (AUC=0.851). The combined model yielded the largest AUC of 0.829 (95% CI, 0.621-0.950) for the external validation of the combined model. GIST patients could be divided into high or low risk subgroups of recurrence and mortality by the Radscore. Conclusion The radiomics models based on enhanced CT for predicting KIT exon 11 deletion mutations have good diagnostic performance.
Collapse
Affiliation(s)
- Yu Zhang
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, China
| | - Xiaofei Yue
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, China
| | - Peng Zhang
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yuying Zhang
- Department of Radiology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Linxia Wu
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, China
| | - Nan Diao
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, China
| | - Guina Ma
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, China
| | - Yuting Lu
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, China
| | - Ling Ma
- He Kang Corporate Management (SH) Co. Ltd., Shanghai, China
| | - Kaixiong Tao
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qian Li
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, China
| | - Ping Han
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, China
| |
Collapse
|
9
|
Rengo M, Onori A, Caruso D, Bellini D, Carbonetti F, De Santis D, Vicini S, Zerunian M, Iannicelli E, Carbone I, Laghi A. Development and Validation of Artificial-Intelligence-Based Radiomics Model Using Computed Tomography Features for Preoperative Risk Stratification of Gastrointestinal Stromal Tumors. J Pers Med 2023; 13:jpm13050717. [PMID: 37240887 DOI: 10.3390/jpm13050717] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 04/18/2023] [Accepted: 04/19/2023] [Indexed: 05/28/2023] Open
Abstract
BACKGROUND preoperative risk assessment of gastrointestinal stromal tumors (GISTS) is required for optimal and personalized treatment planning. Radiomics features are promising tools to predict risk assessment. The purpose of this study is to develop and validate an artificial intelligence classification algorithm, based on CT features, to define GIST's prognosis as determined by the Miettinen classification. METHODS patients with histological diagnosis of GIST and CT studies were retrospectively enrolled. Eight morphologic and 30 texture CT features were extracted from each tumor and combined to obtain three models (morphologic, texture and combined). Data were analyzed using a machine learning classification (WEKA). For each classification process, sensitivity, specificity, accuracy and area under the curve were evaluated. Inter- and intra-reader agreement were also calculated. RESULTS 52 patients were evaluated. In the validation population, highest performances were obtained by the combined model (SE 85.7%, SP 90.9%, ACC 88.8%, and AUC 0.954) followed by the morphologic (SE 66.6%, SP 81.8%, ACC 76.4%, and AUC 0.742) and texture (SE 50%, SP 72.7%, ACC 64.7%, and AUC 0.613) models. Reproducibility was high of all manual evaluations. CONCLUSIONS the AI-based radiomics model using a CT feature demonstrates good predictive performance for preoperative risk stratification of GISTs.
Collapse
Affiliation(s)
- Marco Rengo
- Department of Medical-Surgical Sciences and Biotechnologies, Academic Diagnostic Imaging Division, I.C.O.T. Hospital, University of Rome Sapienza, Via F. Faggiana 1668, 04100 Latina, Italy
| | - Alessandro Onori
- Department of Radiological, Oncological and Pathological Sciences, Academic Diagnostic Imaging Division, I.C.O.T. Hospital, University of Rome Sapienza, Via F. Faggiana 1668, 04100 Latina, Italy
| | - Damiano Caruso
- Department of Surgical and Medical Sciences and Translational Medicine, Radiology Unit, Sant'Andrea University Hospital, University of Rome Sapienza, Via di Grottarossa 1035, 00189 Rome, Italy
| | - Davide Bellini
- Department of Medical-Surgical Sciences and Biotechnologies, Academic Diagnostic Imaging Division, I.C.O.T. Hospital, University of Rome Sapienza, Via F. Faggiana 1668, 04100 Latina, Italy
| | - Francesco Carbonetti
- Radiology Unit, Sant'Eugenio Hospital, Piazzale dell'Umanesimo 10, 00144 Rome, Italy
| | - Domenico De Santis
- Department of Surgical and Medical Sciences and Translational Medicine, Radiology Unit, Sant'Andrea University Hospital, University of Rome Sapienza, Via di Grottarossa 1035, 00189 Rome, Italy
| | - Simone Vicini
- Department of Medical-Surgical Sciences and Biotechnologies, Academic Diagnostic Imaging Division, I.C.O.T. Hospital, University of Rome Sapienza, Via F. Faggiana 1668, 04100 Latina, Italy
| | - Marta Zerunian
- Department of Surgical and Medical Sciences and Translational Medicine, Radiology Unit, Sant'Andrea University Hospital, University of Rome Sapienza, Via di Grottarossa 1035, 00189 Rome, Italy
| | - Elsa Iannicelli
- Department of Surgical and Medical Sciences and Translational Medicine, Radiology Unit, Sant'Andrea University Hospital, University of Rome Sapienza, Via di Grottarossa 1035, 00189 Rome, Italy
| | - Iacopo Carbone
- Department of Radiological, Oncological and Pathological Sciences, Academic Diagnostic Imaging Division, I.C.O.T. Hospital, University of Rome Sapienza, Via F. Faggiana 1668, 04100 Latina, Italy
| | - Andrea Laghi
- Department of Surgical and Medical Sciences and Translational Medicine, Radiology Unit, Sant'Andrea University Hospital, University of Rome Sapienza, Via di Grottarossa 1035, 00189 Rome, Italy
| |
Collapse
|
10
|
Wei Y, Lu Z, Ren Y. Predictive Value of a Radiomics Nomogram Model Based on Contrast-Enhanced Computed Tomography for KIT Exon 9 Gene Mutation in Gastrointestinal Stromal Tumors. Technol Cancer Res Treat 2023; 22:15330338231181260. [PMID: 37296525 PMCID: PMC10272646 DOI: 10.1177/15330338231181260] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2023] [Revised: 04/28/2023] [Accepted: 05/24/2023] [Indexed: 06/12/2023] Open
Abstract
OBJECTIVES To establish and validate a radiomics nomogram model for preoperative prediction of KIT exon 9 mutation status in patients with gastrointestinal stromal tumors (GISTs). MATERIALS AND METHODS Eighty-seven patients with pathologically confirmed GISTs were retrospectively enrolled in this study. Imaging and clinicopathological data were collected and randomly assigned to the training set (n = 60) and test set (n = 27) at a ratio of 7:3. Based on contrast-enhanced CT (CE-CT) arterial and venous phase images, the region of interest (ROI) of the tumors were manually drawn layer by layer, and the radiomics features were extracted. The intra-class correlation coefficient (ICC) was used to test the consistency between observers. Least absolute shrinkage and selection operator regression (LASSO) were used to further screen the features. The nomogram of integrated radiomics score (Rad-Score) and clinical risk factors (extra-gastric location and distant metastasis) was drawn on the basis of multivariate logistic regression. The area under the receiver operating characteristic (AUC) curve and decision curve analysis were used to evaluate the predictive efficiency of the nomogram, and the clinical benefits that the decision curve evaluation model may bring to patients. RESULTS The selected radiomics features (arterial phase and venous phase features) were significantly correlated with the KIT exon 9 mutation status of GISTs. The AUC, sensitivity, specificity, and accuracy in the radiomics model were 0.863, 85.7%, 80.4%, and 85.0% for the training group (95% confidence interval [CI]: 0.750-0.938), and 0.883, 88.9%, 83.3%, and 81.5% for the test group (95% CI: 0.701-0.974), respectively. The AUC, sensitivity, specificity, and accuracy in the nomogram model were 0.902 (95% confidence interval [CI]: 0.798-0.964), 85.7%, 86.9%, and 91.7% for the training group, and 0.907 (95% CI: 0.732-0.984), 77.8%, 94.4%, and 88.9% for the test group, respectively. The decision curve showed the clinical application value of the radiomic nomogram. CONCLUSION The radiomics nomogram model based on CE-CT can effectively predict the KIT exon 9 mutation status of GISTs and may be used for selective gene analysis in the future, which is of great significance for the accurate treatment of GISTs.
Collapse
Affiliation(s)
- Yuze Wei
- Department of Radiology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Zaiming Lu
- Department of Radiology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Ying Ren
- Department of Radiology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| |
Collapse
|
11
|
Weeda YA, Kalisvaart GM, van Velden FHP, Gelderblom H, van der Molen AJ, Bovee JVMG, van der Hage JA, Grootjans W, de Geus-Oei LF. Early Prediction and Monitoring of Treatment Response in Gastrointestinal Stromal Tumors by Means of Imaging: A Systematic Review. Diagnostics (Basel) 2022; 12:2722. [PMID: 36359564 PMCID: PMC9689665 DOI: 10.3390/diagnostics12112722] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 11/03/2022] [Accepted: 11/04/2022] [Indexed: 05/11/2025] Open
Abstract
Gastrointestinal stromal tumors (GISTs) are rare mesenchymal neoplasms. Tyrosine kinase inhibitor (TKI) therapy is currently part of routine clinical practice for unresectable and metastatic disease. It is important to assess the efficacy of TKI treatment at an early stage to optimize therapy strategies and eliminate futile ineffective treatment, side effects and unnecessary costs. This systematic review provides an overview of the imaging features obtained from contrast-enhanced (CE)-CT and 2-deoxy-2-[18F]fluoro-D-glucose ([18F]FDG) PET/CT to predict and monitor TKI treatment response in GIST patients. PubMed, Web of Science, the Cochrane Library and Embase were systematically screened. Articles were considered eligible if quantitative outcome measures (area under the curve (AUC), correlations, sensitivity, specificity, accuracy) were used to evaluate the efficacy of imaging features for predicting and monitoring treatment response to various TKI treatments. The methodological quality of all articles was assessed using the Quality Assessment of Diagnostic Accuracy Studies, v2 (QUADAS-2) tool and modified versions of the Radiomics Quality Score (RQS). A total of 90 articles were included, of which 66 articles used baseline [18F]FDG-PET and CE-CT imaging features for response prediction. Generally, the presence of heterogeneous enhancement on baseline CE-CT imaging was considered predictive for high-risk GISTs, related to underlying neovascularization and necrosis of the tumor. The remaining articles discussed therapy monitoring. Clinically established imaging features, including changes in tumor size and density, were considered unfavorable monitoring criteria, leading to under- and overestimation of response. Furthermore, changes in glucose metabolism, as reflected by [18F]FDG-PET imaging features, preceded changes in tumor size and were more strongly correlated with tumor response. Although CE-CT and [18F]FDG-PET can aid in the prediction and monitoring in GIST patients, further research on cost-effectiveness is recommended.
Collapse
Affiliation(s)
- Ylva. A. Weeda
- Department of Radiology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - Gijsbert M. Kalisvaart
- Department of Radiology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | | | - Hans Gelderblom
- Department of Medical Oncology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - Aart. J. van der Molen
- Department of Radiology, Section of Abdominal Radiology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - Judith V. M. G. Bovee
- Department of Pathology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - Jos A. van der Hage
- Department of Surgical Oncology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - Willem Grootjans
- Department of Radiology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - Lioe-Fee de Geus-Oei
- Department of Radiology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
- Biomedical Photonic Imaging Group, University of Twente, 7522 NB Enschede, The Netherlands
- Department of Radiation Science & Technology, Technical University of Delft, 2629 JB Delft, The Netherlands
| |
Collapse
|
12
|
Inoue A, Ota S, Yamasaki M, Batsaikhan B, Furukawa A, Watanabe Y. Gastrointestinal stromal tumors: a comprehensive radiological review. Jpn J Radiol 2022; 40:1105-1120. [PMID: 35809209 DOI: 10.1007/s11604-022-01305-x] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Accepted: 06/08/2022] [Indexed: 11/29/2022]
Abstract
Gastrointestinal stromal tumors (GISTs) originating from the interstitial cells of Cajal in the muscularis propria are the most common mesenchymal tumor of the gastrointestinal tract. Multiple modalities, including computed tomography (CT), magnetic resonance imaging (MRI), fluorodeoxyglucose positron emission tomography, ultrasonography, digital subtraction angiography, and endoscopy, have been performed to evaluate GISTs. CT is most frequently used for diagnosis, staging, surveillance, and response monitoring during molecularly targeted therapy in clinical practice. The diagnosis of GISTs is sometimes challenging because of the diverse imaging findings, such as anatomical location (esophagus, stomach, duodenum, small bowel, colorectum, appendix, and peritoneum), growth pattern, and enhancement pattern as well as the presence of necrosis, calcification, ulceration, early venous return, and metastasis. Imaging findings of GISTs treated with antineoplastic agents are quite different from those of other neoplasms (e.g. adenocarcinomas) because only subtle changes in size are seen even in responsive lesions. Furthermore, the recurrence pattern of GISTs is different from that of other neoplasms. This review discusses the advantages and disadvantages of each imaging modality, describes imaging findings obtained before and after treatment, presents a few cases of complicated GISTs, and discusses recent investigations performed using CT and MRI to predict histological risk grade, gene mutations, and patient outcomes.
Collapse
Affiliation(s)
- Akitoshi Inoue
- Department of Radiology, Shiga University of Medical Science, Seta, Tsukinowa-cho, Otsu, Shiga, 520-2192, Japan. .,Department of Radiology, Mayo Clinic, Rochester, MN, USA.
| | - Shinichi Ota
- Department of Radiology, Nagahama Red Cross Hospital, Shiga, Japan
| | - Michio Yamasaki
- Department of Radiology, Kohka Public Hospital, Shiga, Japan
| | - Bolorkhand Batsaikhan
- Graduate School of Human Health Sciences, Department of Radiological Science, Tokyo Metropolitan University, Tokyo, Japan
| | - Akira Furukawa
- Graduate School of Human Health Sciences, Department of Radiological Science, Tokyo Metropolitan University, Tokyo, Japan
| | - Yoshiyuki Watanabe
- Department of Radiology, Shiga University of Medical Science, Seta, Tsukinowa-cho, Otsu, Shiga, 520-2192, Japan
| |
Collapse
|
13
|
Wu K, Wu P, Yang K, Li Z, Kong S, Yu L, Zhang E, Liu H, Guo Q, Wu S. A comprehensive texture feature analysis framework of renal cell carcinoma: pathological, prognostic, and genomic evaluation based on CT images. Eur Radiol 2022; 32:2255-2265. [PMID: 34800150 DOI: 10.1007/s00330-021-08353-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 09/16/2021] [Accepted: 09/22/2021] [Indexed: 02/06/2023]
Abstract
OBJECTIVES We tried to realize accurate pathological classification, assessment of prognosis, and genomic molecular typing of renal cell carcinoma by CT texture feature analysis. To determine whether CT texture features can perform accurate pathological classification and evaluation of prognosis and genomic characteristics in renal cell carcinoma. METHODS Patients with renal cell carcinoma from five open-source cohorts were analyzed retrospectively in this study. These data were randomly split to train and test machine learning algorithms to segment the lesion, predict the histological subtype, tumor stage, and pathological grade. Dice coefficient and performance metrics such as accuracy and AUC were calculated to evaluate the segmentation and classification model. Quantitative decomposition of the predictive model was conducted to explore the contribution of each feature. Besides, survival analysis and the statistical correlation between CT texture features, pathological, and genomic signatures were investigated. RESULTS A total of 569 enhanced CT images of 443 patients (mean age 59.4, 278 males) were included in the analysis. In the segmentation task, the mean dice coefficient was 0.96 for the kidney and 0.88 for the cancer region. For classification of histologic subtype, tumor stage, and pathological grade, the model was on a par with radiologists and the AUC was 0.83 [Formula: see text] 0.1, 0.80 [Formula: see text] 0.1, and 0.77 [Formula: see text] 0.1 at 95% confidence intervals, respectively. Moreover, specific quantitative CT features related to clinical prognosis were identified. A strong statistical correlation (R2 = 0.83) between the feature crosses and genomic characteristics was shown. The structural equation modeling confirmed significant associations between CT features, pathological (β = - 0.75), and molecular subtype (β = - 0.30). CONCLUSIONS The framework illustrates high performance in the pathological classification of renal cell carcinoma. Prognosis and genomic characteristics can be inferred by quantitative image analysis. KEY POINTS • The analytical framework exhibits high-performance pathological classification of renal cell carcinoma and is on a par with human radiologists. • Quantitative decomposition of the predictive model shows that specific texture features contribute to histologic subtype and tumor stage classification. • Structural equation modeling shows the associations of genomic characteristics to CT texture features. Overall survival and molecular characteristics can be inferred by quantitative CT texture analysis in renal cell carcinoma.
Collapse
Affiliation(s)
- Kai Wu
- Department of Urology, The Third Affiliated Hospital of Shenzhen University (Luohu Hospital Group), Shenzhen, 518001, China
- Shenzhen Following Precision Medical Research Institute, Luohu Hospital Group, Shenzhen, 518001, China
| | - Peng Wu
- Department of Urology, The Third Affiliated Hospital of Shenzhen University (Luohu Hospital Group), Shenzhen, 518001, China
- Shenzhen Following Precision Medical Research Institute, Luohu Hospital Group, Shenzhen, 518001, China
| | - Kai Yang
- Department of Urology, South China Hospital, Health Science Center, Shenzhen University, Shenzhen, 518116, China
| | - Zhe Li
- Department of Urology, The Third Affiliated Hospital of Shenzhen University (Luohu Hospital Group), Shenzhen, 518001, China
- Shenzhen Following Precision Medical Research Institute, Luohu Hospital Group, Shenzhen, 518001, China
| | - Sijia Kong
- Department of Urology, The Third Affiliated Hospital of Shenzhen University (Luohu Hospital Group), Shenzhen, 518001, China
- Shenzhen Following Precision Medical Research Institute, Luohu Hospital Group, Shenzhen, 518001, China
| | - Lu Yu
- Department of Urology, The Third Affiliated Hospital of Shenzhen University (Luohu Hospital Group), Shenzhen, 518001, China
- Shenzhen Following Precision Medical Research Institute, Luohu Hospital Group, Shenzhen, 518001, China
| | - Enpu Zhang
- Department of Urology, The Third Affiliated Hospital of Shenzhen University (Luohu Hospital Group), Shenzhen, 518001, China
- Shenzhen Following Precision Medical Research Institute, Luohu Hospital Group, Shenzhen, 518001, China
| | - Hanlin Liu
- Department of Urology, The Third Affiliated Hospital of Shenzhen University (Luohu Hospital Group), Shenzhen, 518001, China
- Shenzhen Following Precision Medical Research Institute, Luohu Hospital Group, Shenzhen, 518001, China
| | - Qing Guo
- Department of Urology, The Third Affiliated Hospital of Shenzhen University (Luohu Hospital Group), Shenzhen, 518001, China
- Shenzhen Following Precision Medical Research Institute, Luohu Hospital Group, Shenzhen, 518001, China
| | - Song Wu
- Department of Urology, The Third Affiliated Hospital of Shenzhen University (Luohu Hospital Group), Shenzhen, 518001, China
- Department of Urology, South China Hospital, Health Science Center, Shenzhen University, Shenzhen, 518116, China
- Teaching Center of Shenzhen Luohu Hospital, Shantou University Medical College, Shantou, 515041, China
| |
Collapse
|
14
|
Comparison of Computed Tomography Features of Gastric and Small Bowel Gastrointestinal Stromal Tumors With Different Risk Grades. J Comput Assist Tomogr 2022; 46:175-182. [PMID: 35297574 DOI: 10.1097/rct.0000000000001262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
OBJECTIVE This study aimed to compare the computed tomography (CT) features of gastric and small bowel gastrointestinal stromal tumors (GISTs) and further identify the predictors for risk stratification of them, respectively. METHODS According to the modified National Institutes of Health criteria, patients were classified into low-malignant potential group and high-malignant potential group. Two experienced radiologists reviewed the CT features including the difference of CT values between arterial phase and portal venous phase (PVPMAP) by consensus. The CT features of gastric and small bowel GISTs were compared, and the association of CT features with risk grades was analyzed, respectively. Determinant CT features were used to construct corresponding models. RESULTS Univariate analysis showed that small bowel GISTs tended to present with irregular contour, mixed growth pattern, ill-defined margin, severe necrosis, ulceration, tumor vessels, heterogeneous enhancement, larger size, and marked enhancement compared with gastric GISTs. According to multivariate analysis, tumor size (P < 0.001; odds ratio [OR], 3.279), necrosis (P = 0.008; OR, 2.104) and PVPMAP (P = 0.045; OR, 0.958) were the independent influencing factors for risk stratification of gastric GISTs. In terms of small bowel GISTs, the independent predictors were tumor size (P < 0.001; OR, 3.797) and ulceration (P = 0.031; OR, 4.027). Receiver operating characteristic curve indicated that the CT models for risk stratification of gastric and small bowel GISTs both achieved the best predictive performance. CONCLUSIONS Computed tomography features of gastric and small bowel GISTs are different. Furthermore, the qualitative and quantitative CT features of GISTs may be favorable for preoperative risk stratification.
Collapse
|
15
|
Prediction of the Ki-67 expression level and prognosis of gastrointestinal stromal tumors based on CT radiomics nomogram. Int J Comput Assist Radiol Surg 2022; 17:1167-1175. [PMID: 35195831 DOI: 10.1007/s11548-022-02575-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Accepted: 01/31/2022] [Indexed: 11/05/2022]
Abstract
PURPOSE To build and validate a radiomics nomogram integrated with the radiomics signature and subjective CT characteristics to predict the Ki-67 expression level of gastrointestinal stromal tumors (GISTs). Moreover, the purpose was to compare the performance of pathological Ki-67 expression level with predicted Ki-67 expression level in estimating the prognosis of GISTs patients. METHODS According to pathological results, patients were classified into high-Ki-67 labeling index group (Ki-67 LI ≥ 5%) and low-Ki-67 LI group (Ki-67 LI < 5%). Radiomics features extracted from contrast-enhanced CT(CECT) images were selected and classified to build a radiomics signature. A combined model was built by incorporating radiomics signature and determinant subjective CT characteristics using multivariate logistic regression analysis. The diagnostic performance of the radiomics signature, subjective CT model and combined model were explored by receiver operating characteristic (ROC) curve analysis and Delong test. The model with best diagnostic performance was then set up for the prediction nomogram. Recurrence-free survival (RFS) rates were compared utilizing Kaplan-Meier curve. RESULTS The generated combined model yielded the best diagnostic performance with area under the curve (AUC) values of 0.738 [95% confidence interval (CI): 0.669-0.807] and 0.772 (95% CI 0.683-0.860) in the training set and testing set respectively. The nomogram based on the combined model demonstrated good calibration in the training set and testing set (both P > 0.05). Patients of high-Ki-67 LI group predicted by our nomogram had a poorer RFS than patients of low-Ki-67 LI group (P < 0.0001). CONCLUSION This radiomics nomogram based on CECT had a satisfactory performance in predicting both the Ki-67 expression level and prognosis noninvasively in patients with GISTs, which may serve as an effective imaging tool that can assist in guiding personalized clinical treatment.
Collapse
|
16
|
Starmans MPA, Timbergen MJM, Vos M, Renckens M, Grünhagen DJ, van Leenders GJLH, Dwarkasing RS, Willemssen FEJA, Niessen WJ, Verhoef C, Sleijfer S, Visser JJ, Klein S. Differential Diagnosis and Molecular Stratification of Gastrointestinal Stromal Tumors on CT Images Using a Radiomics Approach. J Digit Imaging 2022; 35:127-136. [PMID: 35088185 PMCID: PMC8921463 DOI: 10.1007/s10278-022-00590-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Revised: 01/05/2022] [Accepted: 01/14/2022] [Indexed: 12/21/2022] Open
Abstract
Treatment planning of gastrointestinal stromal tumors (GISTs) includes distinguishing GISTs from other intra-abdominal tumors and GISTs’ molecular analysis. The aim of this study was to evaluate radiomics for distinguishing GISTs from other intra-abdominal tumors, and in GISTs, predict the c-KIT, PDGFRA, BRAF mutational status, and mitotic index (MI). Patients diagnosed at the Erasmus MC between 2004 and 2017, with GIST or non-GIST intra-abdominal tumors and a contrast-enhanced venous-phase CT, were retrospectively included. Tumors were segmented, from which 564 image features were extracted. Prediction models were constructed using a combination of machine learning approaches. The evaluation was performed in a 100 × random-split cross-validation. Model performance was compared to that of three radiologists. One hundred twenty-five GISTs and 122 non-GISTs were included. The GIST vs. non-GIST radiomics model had a mean area under the curve (AUC) of 0.77. Three radiologists had an AUC of 0.69, 0.76, and 0.84, respectively. The radiomics model had an AUC of 0.52 for c-KIT, 0.56 for c-KIT exon 11, and 0.52 for the MI. The numbers of PDGFRA, BRAF, and other c-KIT mutations were too low for analysis. Our radiomics model was able to distinguish GISTs from non-GISTs with a performance similar to three radiologists, but less observer dependent. Therefore, it may aid in the early diagnosis of GIST, facilitating rapid referral to specialized treatment centers. As the model was not able to predict any genetic or molecular features, it cannot aid in treatment planning yet.
Collapse
Affiliation(s)
- Martijn P A Starmans
- Department of Radiology and Nuclear Medicine, Erasmus Medical Center, Rotterdam, The Netherlands.
- Department of Medical Informatics, Erasmus Medical Center, Rotterdam, The Netherlands.
| | - Milea J M Timbergen
- Department of Surgical Oncology, Erasmus MC Cancer Institute, Erasmus Medical Center, Rotterdam, The Netherlands
- Department of Medical Oncology, Erasmus MC Cancer Institute, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Melissa Vos
- Department of Surgical Oncology, Erasmus MC Cancer Institute, Erasmus Medical Center, Rotterdam, The Netherlands
- Department of Medical Oncology, Erasmus MC Cancer Institute, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Michel Renckens
- Department of Radiology and Nuclear Medicine, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Dirk J Grünhagen
- Department of Surgical Oncology, Erasmus MC Cancer Institute, Erasmus Medical Center, Rotterdam, The Netherlands
| | | | - Roy S Dwarkasing
- Department of Radiology and Nuclear Medicine, Erasmus Medical Center, Rotterdam, The Netherlands
| | | | - Wiro J Niessen
- Department of Radiology and Nuclear Medicine, Erasmus Medical Center, Rotterdam, The Netherlands
- Department of Medical Informatics, Erasmus Medical Center, Rotterdam, The Netherlands
- Faculty of Applied Sciences, Delft University of Technology, Delft, The Netherlands
| | - Cornelis Verhoef
- Department of Surgical Oncology, Erasmus MC Cancer Institute, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Stefan Sleijfer
- Department of Medical Oncology, Erasmus MC Cancer Institute, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Jacob J Visser
- Department of Radiology and Nuclear Medicine, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Stefan Klein
- Department of Radiology and Nuclear Medicine, Erasmus Medical Center, Rotterdam, The Netherlands
- Department of Medical Informatics, Erasmus Medical Center, Rotterdam, The Netherlands
| |
Collapse
|
17
|
Park HJ, Qin L, Bakouny Z, Krajewski KM, Van Allen EM, Choueiri TK, Shinagare AB. OUP accepted manuscript. Oncologist 2022; 27:389-397. [PMID: 35348767 PMCID: PMC9074990 DOI: 10.1093/oncolo/oyac034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2021] [Accepted: 01/07/2022] [Indexed: 11/15/2022] Open
Abstract
Background Materials and Methods Results Conclusion
Collapse
Affiliation(s)
- Hyo Jung Park
- Department of Radiology and Research Institute of Radiology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Republic of Korea
| | - Lei Qin
- Department of Imaging, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
- Department of Radiology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Ziad Bakouny
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Katherine M Krajewski
- Department of Imaging, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
- Department of Radiology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Eliezer M Van Allen
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Toni K Choueiri
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Atul B Shinagare
- Department of Imaging, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
- Department of Radiology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
- Corresponding author: Atul B. Shinagare, Department of Radiology, Brigham and Womens Hospital, Harvard Medical School, 75 Francis Street, Boston, MA 02115, USA. Tel.: +1 6176322988; Fax: +1 6175828574;
| |
Collapse
|
18
|
Shao M, Niu Z, He L, Fang Z, He J, Xie Z, Cheng G, Wang J. Building Radiomics Models Based on Triple-Phase CT Images Combining Clinical Features for Discriminating the Risk Rating in Gastrointestinal Stromal Tumors. Front Oncol 2021; 11:737302. [PMID: 34950578 PMCID: PMC8689687 DOI: 10.3389/fonc.2021.737302] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Accepted: 11/15/2021] [Indexed: 12/24/2022] Open
Abstract
We aimed to build radiomics models based on triple-phase CT images combining clinical features to predict the risk rating of gastrointestinal stromal tumors (GISTs). A total of 231 patients with pathologically diagnosed GISTs from July 2012 to July 2020 were categorized into a training data set (82 patients with high risk, 80 patients with low risk) and a validation data set (35 patients with high risk, 34 patients with low risk) with a ratio of 7:3. Four diagnostic models were constructed by assessing 20 clinical characteristics and 18 radiomic features that were extracted from a lesion mask based on triple-phase CT images. The receiver operating characteristic (ROC) curves were applied to calculate the diagnostic performance of these models, and ROC curves of these models were compared using Delong test in different data sets. The results of ROC analyses showed that areas under ROC curves (AUC) of model 4 [Clinic + CT value of unenhanced (CTU) + CT value of arterial phase (CTA) + value of venous phase (CTV)], model 1 (Clinic + CTU), model 2 (Clinic + CTA), and model 3 (Clinic + CTV) were 0.925, 0.894, 0.909, and 0.914 in the training set and 0.897, 0.866, 0,892, and 0.892 in the validation set, respectively. Model 4, model 1, model 2, and model 3 yielded an accuracy of 88.3%, 85.8%, 86.4%, and 84.6%, a sensitivity of 85.4%, 84.2%, 76.8%, and 78.0%, and a specificity of 91.2%, 87.5%, 96.2%, and 91.2% in the training set and an accuracy of 88.4%, 84.1%, 82.6%, and 82.6%, a sensitivity of 88.6%, 77.1%, 74.3%, and 85.7%, and a specificity of 88.2%, 91.2%, 91.2%, and 79.4% in the validation set, respectively. There was a significant difference between model 4 and model 1 in discriminating the risk rating in gastrointestinal stromal tumors in the training data set (Delong test, p < 0.05). The radiomic models based on clinical features and triple-phase CT images manifested excellent accuracy for the discrimination of risk rating of GISTs.
Collapse
Affiliation(s)
- Meihua Shao
- Department of Radiology, Tongde Hospital of Zhejiang Province, Hangzhou, China
| | - Zhongfeng Niu
- Department of Radiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Linyang He
- Hangzhou Jianpei Technology Company, Hangzhou, China
| | - Zhaoxing Fang
- Hangzhou Jianpei Technology Company, Hangzhou, China
| | - Jie He
- Department of Radiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Zongyu Xie
- Department of Radiology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, China
| | - Guohua Cheng
- Hangzhou Jianpei Technology Company, Hangzhou, China
| | - Jian Wang
- Department of Radiology, Tongde Hospital of Zhejiang Province, Hangzhou, China
| |
Collapse
|
19
|
Liu X, Yin Y, Wang X, Yang C, Wan S, Yin X, Wu T, Chen H, Xu Z, Li X, Song B, Zhang B. Gastrointestinal stromal tumors: associations between contrast-enhanced CT images and KIT exon 11 gene mutation. ANNALS OF TRANSLATIONAL MEDICINE 2021; 9:1496. [PMID: 34805358 PMCID: PMC8573436 DOI: 10.21037/atm-21-3811] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Accepted: 09/02/2021] [Indexed: 02/05/2023]
Abstract
Background Mutation screening for gastrointestinal stromal tumor (GIST) is crucial and the c kit gene (KIT) exon 11 mutation is the most common type. This study aimed to explore the associations between GIST with KIT exon 11 mutation and contrast-enhanced computed tomography (CT) images. Methods Pathologically proven GISTs with definitive genotype testing results in our hospital were retrospectively included. Abdominal contrast-enhanced CT images were analyzed. Conventional CT image features and radiomic features were recorded and extracted to build the following models: model [CT], model [radiomic + clinic] and model [CT + radiomic + clinic]. The diagnostic performances of GISTs with KIT exon 11 mutation and KIT exon 11 deletion involving codons 557–558 were evaluated. Results In total, 327 GISTs (255 with KIT exon 11 mutation, and 73 with KIT exon 11 mutation deletion involving codons 557–558) were included. Significant CT features were found for GISTs with KIT exon 11 mutation. The area under curves (AUCs) of the models for KIT exon 11 mutation were 0.7158, 0.7530, and 0.8375 in the training cohort, and 0.6777, 0.7349, and 0.8105 in validation cohort, respectively. The AUCs of the models for KIT exon 11 mutation deletion involving codons 557–558 were 0.7155, 8621, and 0.8691 in the training cohort, and 0.7099, 0.8355, and 0.8488 in the validation cohort, respectively. The model [CT + radiomic + clinic] demonstrated the highest AUCs for prediction of KIT exon 11 mutation and those with deletion involving codons 557–558 (P<0.05), respectively. The model [radiomic + clinic] showed higher diagnostic performance than model [CT] significantly. Conclusions Our results demonstrated the associations between GIST with KIT exon 11 mutation and contrast-enhanced CT images. We found combing conventional image analysis and texture analysis is a useful tool to distinguish GIST with KIT exon 11 mutation. CT radiogenomics exhibited good application potential in predict the KIT exon 11 mutation of GIST.
Collapse
Affiliation(s)
- Xijiao Liu
- Department of Radiology, West China Hospital, Sichuan University, Chengdu, China
| | - Yuan Yin
- Department of Gastrointestinal Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Xiaozhou Wang
- Department of Radiology, Meishan People's Hospital, Meishan, China
| | - Caiwei Yang
- Department of Radiology, West China Hospital, Sichuan University, Chengdu, China
| | - Shang Wan
- Department of Radiology, West China Hospital, Sichuan University, Chengdu, China
| | - Xiaonan Yin
- Department of Gastrointestinal Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Tingfan Wu
- Clinical Education Team, GE Healthcare China, Shanghai, China
| | - Huijiao Chen
- Department of Pathology, West China Hospital, Sichuan University, Chengdu, China
| | - Zhongming Xu
- Department of Radiology, Meishan People's Hospital, Meishan, China
| | - Xin Li
- Global Research, GE Healthcare China, Shanghai, China
| | - Bin Song
- Department of Radiology, West China Hospital, Sichuan University, Chengdu, China
| | - Bo Zhang
- Department of Gastrointestinal Surgery, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
20
|
Liang CW, Fang PW, Huang HY, Lo CM. Deep Convolutional Neural Networks Detect Tumor Genotype from Pathological Tissue Images in Gastrointestinal Stromal Tumors. Cancers (Basel) 2021; 13:5787. [PMID: 34830948 PMCID: PMC8616403 DOI: 10.3390/cancers13225787] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Revised: 11/07/2021] [Accepted: 11/16/2021] [Indexed: 11/17/2022] Open
Abstract
Gastrointestinal stromal tumors (GIST) are common mesenchymal tumors, and their effective treatment depends upon the mutational subtype of the KIT/PDGFRA genes. We established deep convolutional neural network (DCNN) models to rapidly predict drug-sensitive mutation subtypes from images of pathological tissue. A total of 5153 pathological images of 365 different GISTs from three different laboratories were collected and divided into training and validation sets. A transfer learning mechanism based on DCNN was used with four different network architectures, to identify cases with drug-sensitive mutations. The accuracy ranged from 87% to 75%. Cross-institutional inconsistency, however, was observed. Using gray-scale images resulted in a 7% drop in accuracy (accuracy 80%, sensitivity 87%, specificity 73%). Using images containing only nuclei (accuracy 81%, sensitivity 87%, specificity 73%) or cytoplasm (accuracy 79%, sensitivity 88%, specificity 67%) produced 6% and 8% drops in accuracy rate, respectively, suggesting buffering effects across subcellular components in DCNN interpretation. The proposed DCNN model successfully inferred cases with drug-sensitive mutations with high accuracy. The contribution of image color and subcellular components was also revealed. These results will help to generate a cheaper and quicker screening method for tumor gene testing.
Collapse
Affiliation(s)
- Cher-Wei Liang
- Department of Pathology, Fu Jen Catholic University Hospital, Fu Jen Catholic University, New Taipei City 243, Taiwan; (C.-W.L.); (P.-W.F.)
- School of Medicine, College of Medicine, Fu Jen Catholic University, New Taipei City 242, Taiwan
- Graduate Institute of Pathology, College of Medicine, National Taiwan University, Taipei 100, Taiwan
| | - Pei-Wei Fang
- Department of Pathology, Fu Jen Catholic University Hospital, Fu Jen Catholic University, New Taipei City 243, Taiwan; (C.-W.L.); (P.-W.F.)
| | - Hsuan-Ying Huang
- Department of Anatomic Pathology, Chang Gung Memorial Hospital, College of Medicine, Chang Gung University, Kaohsiung 833, Taiwan;
| | - Chung-Ming Lo
- Graduate Institute of Library, Information and Archival Studies, National Chengchi University, Taipei 116, Taiwan
| |
Collapse
|
21
|
Chen Y, Wei K, Liu D, Xiang J, Wang G, Meng X, Peng J. A Machine Learning Model for Predicting a Major Response to Neoadjuvant Chemotherapy in Advanced Gastric Cancer. Front Oncol 2021; 11:675458. [PMID: 34141620 PMCID: PMC8204104 DOI: 10.3389/fonc.2021.675458] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Accepted: 05/11/2021] [Indexed: 01/08/2023] Open
Abstract
Aims To develop and validate a model for predicting major pathological response to neoadjuvant chemotherapy (NAC) in advanced gastric cancer (AGC) based on a machine learning algorithm. Method A total of 221 patients who underwent NAC and radical gastrectomy between February 2013 and September 2020 were enrolled in this study. A total of 144 patients were assigned to the training cohort for model building, and 77 patients were assigned to the validation cohort. A major pathological response was defined as primary tumor regressing to ypT0 or T1. Radiomic features extracted from venous-phase computed tomography (CT) images were selected by machine learning algorithms to calculate a radscore. Together with other clinical variables selected by univariate analysis, the radscores were included in a binary logistic regression analysis to construct an integrated prediction model. The data obtained for the validation cohort were used to test the predictive accuracy of the model. Result A total of 27.6% (61/221) patients achieved a major pathological response. Five features of 572 radiomic features were selected to calculate the radscores. The final established model incorporates adenocarcinoma differentiation and radscores. The model showed satisfactory predictive accuracy with a C-index of 0.763 and good fitting between the validation data and the model in the calibration curve. Conclusion A prediction model incorporating adenocarcinoma differentiation and radscores was developed and validated. The model helps stratify patients according to their potential sensitivity to NAC and could serve as an individualized treatment strategy-making tool for AGC patients.
Collapse
Affiliation(s)
- Yonghe Chen
- Department of Gastrointestinal Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.,Guangdong Institute of Gastroenterology, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangzhou, China
| | - Kaikai Wei
- Department of Radiology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.,Guangdong Institute of Gastroenterology, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangzhou, China
| | - Dan Liu
- Department of Laboratory Science, The Second Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jun Xiang
- Department of Gastrointestinal Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.,Guangdong Institute of Gastroenterology, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangzhou, China
| | - Gang Wang
- School of Public Health, Sun Yat-sen University, Shenzhen, China
| | - Xiaochun Meng
- Department of Radiology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.,Guangdong Institute of Gastroenterology, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangzhou, China
| | - Junsheng Peng
- Department of Gastrointestinal Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.,Guangdong Institute of Gastroenterology, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangzhou, China
| |
Collapse
|
22
|
Ganeshan B, Miles K, Afaq A, Punwani S, Rodriguez M, Wan S, Walls D, Hoy L, Khan S, Endozo R, Shortman R, Hoath J, Bhargava A, Hanson M, Francis D, Arulampalam T, Dindyal S, Chen SH, Ng T, Groves A. Texture Analysis of Fractional Water Content Images Acquired during PET/MRI: Initial Evidence for an Association with Total Lesion Glycolysis, Survival and Gene Mutation Profile in Primary Colorectal Cancer. Cancers (Basel) 2021; 13:2715. [PMID: 34072712 PMCID: PMC8199380 DOI: 10.3390/cancers13112715] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 05/19/2021] [Accepted: 05/21/2021] [Indexed: 01/07/2023] Open
Abstract
To assess the capability of fractional water content (FWC) texture analysis (TA) to generate biologically relevant information from routine PET/MRI acquisitions for colorectal cancer (CRC) patients. Thirty consecutive primary CRC patients (mean age 63.9, range 42-83 years) prospectively underwent FDG-PET/MRI. FWC tumor parametric images generated from Dixon MR sequences underwent TA using commercially available research software (TexRAD). Data analysis comprised (1) identification of functional imaging correlates for texture features (TF) with low inter-observer variability (intraclass correlation coefficient: ICC > 0.75), (2) evaluation of prognostic performance for FWC-TF, and (3) correlation of prognostic imaging signatures with gene mutation (GM) profile. Of 32 FWC-TF with ICC > 0.75, 18 correlated with total lesion glycolysis (TLG, highest: rs = -0.547, p = 0.002). Using optimized cut-off values, five MR FWC-TF identified a good prognostic group with zero mortality (lowest: p = 0.017). For the most statistically significant prognostic marker, favorable prognosis was significantly associated with a higher number of GM per patient (medians: 7 vs. 1.5, p = 0.009). FWC-TA derived from routine PET/MRI Dixon acquisitions shows good inter-operator agreement, generates biological relevant information related to TLG, GM count, and provides prognostic information that can unlock new clinical applications for CRC patients.
Collapse
Affiliation(s)
- Balaji Ganeshan
- Research Department of Imaging, Division of Medicine, University College London (UCL), London WC1E 6BT, UK; (K.M.); (S.P.); (D.W.); (L.H.); (J.H.); (S.-H.C.); (A.G.)
| | - Kenneth Miles
- Research Department of Imaging, Division of Medicine, University College London (UCL), London WC1E 6BT, UK; (K.M.); (S.P.); (D.W.); (L.H.); (J.H.); (S.-H.C.); (A.G.)
| | - Asim Afaq
- Imaging Division, Surgery and Cancer Board, University College London Hospitals (UCLH) NHS Foundation Trust, University College Hospital (UCH), London NW1 2BU, UK; (A.A.); (M.R.); (S.W.); (S.K.); (R.E.); (R.S.); (S.D.)
- Department of Radiology, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
| | - Shonit Punwani
- Research Department of Imaging, Division of Medicine, University College London (UCL), London WC1E 6BT, UK; (K.M.); (S.P.); (D.W.); (L.H.); (J.H.); (S.-H.C.); (A.G.)
| | - Manuel Rodriguez
- Imaging Division, Surgery and Cancer Board, University College London Hospitals (UCLH) NHS Foundation Trust, University College Hospital (UCH), London NW1 2BU, UK; (A.A.); (M.R.); (S.W.); (S.K.); (R.E.); (R.S.); (S.D.)
| | - Simon Wan
- Imaging Division, Surgery and Cancer Board, University College London Hospitals (UCLH) NHS Foundation Trust, University College Hospital (UCH), London NW1 2BU, UK; (A.A.); (M.R.); (S.W.); (S.K.); (R.E.); (R.S.); (S.D.)
| | - Darren Walls
- Research Department of Imaging, Division of Medicine, University College London (UCL), London WC1E 6BT, UK; (K.M.); (S.P.); (D.W.); (L.H.); (J.H.); (S.-H.C.); (A.G.)
| | - Luke Hoy
- Research Department of Imaging, Division of Medicine, University College London (UCL), London WC1E 6BT, UK; (K.M.); (S.P.); (D.W.); (L.H.); (J.H.); (S.-H.C.); (A.G.)
| | - Saif Khan
- Imaging Division, Surgery and Cancer Board, University College London Hospitals (UCLH) NHS Foundation Trust, University College Hospital (UCH), London NW1 2BU, UK; (A.A.); (M.R.); (S.W.); (S.K.); (R.E.); (R.S.); (S.D.)
| | - Raymond Endozo
- Imaging Division, Surgery and Cancer Board, University College London Hospitals (UCLH) NHS Foundation Trust, University College Hospital (UCH), London NW1 2BU, UK; (A.A.); (M.R.); (S.W.); (S.K.); (R.E.); (R.S.); (S.D.)
| | - Robert Shortman
- Imaging Division, Surgery and Cancer Board, University College London Hospitals (UCLH) NHS Foundation Trust, University College Hospital (UCH), London NW1 2BU, UK; (A.A.); (M.R.); (S.W.); (S.K.); (R.E.); (R.S.); (S.D.)
| | - John Hoath
- Research Department of Imaging, Division of Medicine, University College London (UCL), London WC1E 6BT, UK; (K.M.); (S.P.); (D.W.); (L.H.); (J.H.); (S.-H.C.); (A.G.)
| | - Aman Bhargava
- Institute of Health Barts and London Medical School, Queen Mary University of London (QMUL), London E1 2AD, UK;
| | - Matthew Hanson
- Division of Cancer and Clinical Support, Barking, Havering and Redbridge University Hospitals NHS Trust, Queens and King George Hospitals, Essex IG3 8YB, UK;
| | - Daren Francis
- Department of Colorectal Surgery, Royal Free London NHS Foundation Trust, Barnet and Chase Farm Hospitals, London NW3 2QG, UK;
| | - Tan Arulampalam
- Department of Surgery, East Suffolk and North Essex NHS Foundation Trust, Colchester General Hospital, Colchester CO4 5JL, UK;
| | - Sanjay Dindyal
- Imaging Division, Surgery and Cancer Board, University College London Hospitals (UCLH) NHS Foundation Trust, University College Hospital (UCH), London NW1 2BU, UK; (A.A.); (M.R.); (S.W.); (S.K.); (R.E.); (R.S.); (S.D.)
| | - Shih-Hsin Chen
- Research Department of Imaging, Division of Medicine, University College London (UCL), London WC1E 6BT, UK; (K.M.); (S.P.); (D.W.); (L.H.); (J.H.); (S.-H.C.); (A.G.)
- Department of Nuclear Medicine, Keelung Chang Gung Memorial Hospital, Keelung 204, Taiwan
| | - Tony Ng
- School of Cancer & Pharmaceutical Sciences, King’s College London (KCL), London WC2R 2LS, UK;
| | - Ashley Groves
- Research Department of Imaging, Division of Medicine, University College London (UCL), London WC1E 6BT, UK; (K.M.); (S.P.); (D.W.); (L.H.); (J.H.); (S.-H.C.); (A.G.)
| |
Collapse
|
23
|
TCGA-TCIA-Based CT Radiomics Study for Noninvasively Predicting Epstein-Barr Virus Status in Gastric Cancer. AJR Am J Roentgenol 2021; 217:124-134. [PMID: 33955777 DOI: 10.2214/ajr.20.23534] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
OBJECTIVE. The purpose of this study was to investigate the value of TCGA-TCIA (The Cancer Genome Atlas and The Cancer Imaging Archive)-based CT radiomics for noninvasive prediction of Epstein-Barr virus (EBV) status in gastric cancer (GC). MATERIALS AND METHODS. A total of 133 patients with pathologically confirmed GC (94 in the training cohort and 39 in the validation cohort) who were identified from the TCGA-TCIA public data repository and two hospitals were retrospectively enrolled in the study. Two-dimensional and 3D radiomics features were extracted to construct corresponding radiomics signatures. Then, 2D and 3D nomograms were built by combining radiomics signatures and clinical information on the basis of multivariable analysis. Their performance and clinical practicability were determined, validated, and compared with respect to discrimination, calibration, reclassification, and time spent on tumor segmentation. RESULTS. Both 2D and 3D nomograms were robust and showed good calibration. The AUCs of the 2D and 3D nomograms showed no significant difference in the training cohort (0.919 vs 0.945, respectively; p = .41) or validation cohort (0.939 vs 0.955, respectively; p = .71). The net reclassification index showed that the 3D nomogram revealed no significant improvement in risk reclassification when compared with the 2D nomogram in the training cohort (net reclassification index, 0.68%; p = .14) and the validation cohort (net reclassification index, 6.06%; p = .08). Of note, the time spent on 3D segmentation (median, 907 seconds) was higher than that spent on 2D segmentation (median, 129 seconds). CONCLUSION. The 2D and 3D radiomics nomograms might have the potential to be used as effective tools for prediction of EBV in GC. When time spent on segmentation is considered, the 2D nomogram is more highly recommended for clinical application.
Collapse
|
24
|
Liu Z, Wu K, Wu B, Tang X, Yuan H, Pang H, Huang Y, Zhu X, Luo H, Qi Y. Imaging genomics for accurate diagnosis and treatment of tumors: A cutting edge overview. Biomed Pharmacother 2020; 135:111173. [PMID: 33383370 DOI: 10.1016/j.biopha.2020.111173] [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: 09/26/2020] [Revised: 11/30/2020] [Accepted: 12/14/2020] [Indexed: 02/07/2023] Open
Abstract
Imaging genomics refers to the establishment of the connection between invasive gene expression features and non-invasive imaging features. Tumor imaging genomics can not only understand the macroscopic phenotype of tumor, but also can deeply analyze the cellular and molecular characteristics of tumor tissue. In recent years, tumor imaging genomics has been a key in the field of medicine. The incidence of cancer in China has increased significantly, which is the main reason of disease death of urban residents. With the rapid development of imaging medicine, depending on imaging genomics, many experts have made remarkable achievements in tumor screening and diagnosis, prognosis evaluation, new treatment targets and understanding of tumor biological mechanism. This review analyzes the relationship between tumor radiology and gene expression, which provides a favorable direction for clinical staging, prognosis evaluation and accurate treatment of tumors.
Collapse
Affiliation(s)
- Zhen Liu
- Southern Marine Science and Engineering Guangdong Laboratory (Zhanjiang), Zhanjiang, China; Guangdong Key Laboratory for Research and Development of Natural Drugs, The Marine Biomedical Research Institute, Guangdong Medical University, Zhanjiang, China; The Key Lab of Zhanjiang for R&D Marine Microbial Resources in the Beibu Gulf Rim, Guangdong Medical University, Zhanjiang, China; The Marine Biomedical Research Institute of Guangdong Zhanjiang, Zhanjiang, China
| | - Kefeng Wu
- Southern Marine Science and Engineering Guangdong Laboratory (Zhanjiang), Zhanjiang, China
| | - Binhua Wu
- Southern Marine Science and Engineering Guangdong Laboratory (Zhanjiang), Zhanjiang, China; Guangdong Key Laboratory for Research and Development of Natural Drugs, The Marine Biomedical Research Institute, Guangdong Medical University, Zhanjiang, China; The Key Lab of Zhanjiang for R&D Marine Microbial Resources in the Beibu Gulf Rim, Guangdong Medical University, Zhanjiang, China; The Marine Biomedical Research Institute of Guangdong Zhanjiang, Zhanjiang, China
| | - Xiaoning Tang
- Southern Marine Science and Engineering Guangdong Laboratory (Zhanjiang), Zhanjiang, China; Guangdong Key Laboratory for Research and Development of Natural Drugs, The Marine Biomedical Research Institute, Guangdong Medical University, Zhanjiang, China
| | - Huiqing Yuan
- Southern Marine Science and Engineering Guangdong Laboratory (Zhanjiang), Zhanjiang, China; Guangdong Key Laboratory for Research and Development of Natural Drugs, The Marine Biomedical Research Institute, Guangdong Medical University, Zhanjiang, China; The Key Lab of Zhanjiang for R&D Marine Microbial Resources in the Beibu Gulf Rim, Guangdong Medical University, Zhanjiang, China; The Marine Biomedical Research Institute of Guangdong Zhanjiang, Zhanjiang, China
| | - Hao Pang
- Southern Marine Science and Engineering Guangdong Laboratory (Zhanjiang), Zhanjiang, China
| | - Yongmei Huang
- Southern Marine Science and Engineering Guangdong Laboratory (Zhanjiang), Zhanjiang, China; Guangdong Key Laboratory for Research and Development of Natural Drugs, The Marine Biomedical Research Institute, Guangdong Medical University, Zhanjiang, China; The Key Lab of Zhanjiang for R&D Marine Microbial Resources in the Beibu Gulf Rim, Guangdong Medical University, Zhanjiang, China; The Marine Biomedical Research Institute of Guangdong Zhanjiang, Zhanjiang, China
| | - Xiao Zhu
- Southern Marine Science and Engineering Guangdong Laboratory (Zhanjiang), Zhanjiang, China; Guangdong Key Laboratory for Research and Development of Natural Drugs, The Marine Biomedical Research Institute, Guangdong Medical University, Zhanjiang, China; The Key Lab of Zhanjiang for R&D Marine Microbial Resources in the Beibu Gulf Rim, Guangdong Medical University, Zhanjiang, China; The Marine Biomedical Research Institute of Guangdong Zhanjiang, Zhanjiang, China.
| | - Hui Luo
- Southern Marine Science and Engineering Guangdong Laboratory (Zhanjiang), Zhanjiang, China; Guangdong Key Laboratory for Research and Development of Natural Drugs, The Marine Biomedical Research Institute, Guangdong Medical University, Zhanjiang, China; The Key Lab of Zhanjiang for R&D Marine Microbial Resources in the Beibu Gulf Rim, Guangdong Medical University, Zhanjiang, China; The Marine Biomedical Research Institute of Guangdong Zhanjiang, Zhanjiang, China.
| | - Yi Qi
- Southern Marine Science and Engineering Guangdong Laboratory (Zhanjiang), Zhanjiang, China; Guangdong Key Laboratory for Research and Development of Natural Drugs, The Marine Biomedical Research Institute, Guangdong Medical University, Zhanjiang, China; The Key Lab of Zhanjiang for R&D Marine Microbial Resources in the Beibu Gulf Rim, Guangdong Medical University, Zhanjiang, China; The Marine Biomedical Research Institute of Guangdong Zhanjiang, Zhanjiang, China.
| |
Collapse
|
25
|
Yang CW, Liu XJ, Liu SY, Wan S, Ye Z, Song B. Current and Potential Applications of Artificial Intelligence in Gastrointestinal Stromal Tumor Imaging. CONTRAST MEDIA & MOLECULAR IMAGING 2020; 2020:6058159. [PMID: 33304203 PMCID: PMC7714601 DOI: 10.1155/2020/6058159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 10/18/2020] [Accepted: 10/31/2020] [Indexed: 02/05/2023]
Abstract
The most common mesenchymal tumors are gastrointestinal stromal tumors (GISTs), which have malignant potential and can occur anywhere along the gastrointestinal system. Imaging methods are important and indispensable of GISTs in diagnosis, risk staging, therapy, and follow-up. The recommended imaging method for staging and follow-up is computed tomography (CT) according to current guidelines. Artificial intelligence (AI) applies and elaborates theses, procedures, modes, and utilization systems for simulating, enlarging, and stretching the intellectual capacity of humans. Recently, researchers have done a few studies to explore AI applications in GIST imaging. This article reviews the present AI studies in GISTs imaging, including preoperative diagnosis, risk stratification and prediction of prognosis, gene mutation, and targeted therapy response.
Collapse
Affiliation(s)
- Cai-Wei Yang
- Department of Radiology, West China Hospital, Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Xi-Jiao Liu
- Department of Radiology, West China Hospital, Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Si-Yun Liu
- GE Healthcare (China), Beijing 100176, China
| | - Shang Wan
- Department of Radiology, West China Hospital, Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Zheng Ye
- Department of Radiology, West China Hospital, Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Bin Song
- Department of Radiology, West China Hospital, Sichuan University, Chengdu 610041, Sichuan Province, China
| |
Collapse
|
26
|
Cannella R, La Grutta L, Midiri M, Bartolotta TV. New advances in radiomics of gastrointestinal stromal tumors. World J Gastroenterol 2020; 26:4729-4738. [PMID: 32921953 PMCID: PMC7459199 DOI: 10.3748/wjg.v26.i32.4729] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 06/16/2020] [Accepted: 08/01/2020] [Indexed: 02/06/2023] Open
Abstract
Gastrointestinal stromal tumors (GISTs) are uncommon neoplasms of the gastrointestinal tract with peculiar clinical, genetic, and imaging characteristics. Preoperative knowledge of risk stratification and mutational status is crucial to guide the appropriate patients’ treatment. Predicting the clinical behavior and biological aggressiveness of GISTs based on conventional computed tomography (CT) and magnetic resonance imaging (MRI) evaluation is challenging, unless the lesions have already metastasized at the time of diagnosis. Radiomics is emerging as a promising tool for the quantification of lesion heterogeneity on radiological images, extracting additional data that cannot be assessed by visual analysis. Radiomics applications have been explored for the differential diagnosis of GISTs from other gastrointestinal neoplasms, risk stratification and prediction of prognosis after surgical resection, and evaluation of mutational status in GISTs. The published researches on GISTs radiomics have obtained excellent performance of derived radiomics models on CT and MRI. However, lack of standardization and differences in study methodology challenge the application of radiomics in clinical practice. The purpose of this review is to describe the new advances of radiomics applied to CT and MRI for the evaluation of gastrointestinal stromal tumors, discuss the potential clinical applications that may impact patients’ management, report limitations of current radiomics studies, and future directions.
Collapse
Affiliation(s)
- Roberto Cannella
- Section of Radiology - BiND, University Hospital “Paolo Giaccone”, Palermo 90127, Italy
| | - Ludovico La Grutta
- Section of Radiology - BiND, University Hospital “Paolo Giaccone”, Palermo 90127, Italy
| | - Massimo Midiri
- Section of Radiology - BiND, University Hospital “Paolo Giaccone”, Palermo 90127, Italy
| | - Tommaso Vincenzo Bartolotta
- Section of Radiology - BiND, University Hospital “Paolo Giaccone”, Palermo 90127, Italy
- Department of Radiology, Fondazione Istituto Giuseppe Giglio, Ct.da Pietrapollastra, Cefalù (Palermo) 90015, Italy
| |
Collapse
|
27
|
Yang G, Gong A, Nie P, Yan L, Miao W, Zhao Y, Wu J, Cui J, Jia Y, Wang Z. Contrast-Enhanced CT Texture Analysis for Distinguishing Fat-Poor Renal Angiomyolipoma From Chromophobe Renal Cell Carcinoma. Mol Imaging 2020; 18:1536012119883161. [PMID: 31625454 PMCID: PMC6801892 DOI: 10.1177/1536012119883161] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Objective: To evaluate the value of 2-dimensional (2D) and 3-dimensional (3D) computed tomography
texture analysis (CTTA) models in distinguishing fat-poor angiomyolipoma (fpAML) from
chromophobe renal cell carcinoma (chRCC). Methods: We retrospectively enrolled 32 fpAMLs and 24 chRCCs. Texture features were extracted
from 2D and 3D regions of interest in triphasic CT images. The 2D and 3D CTTA models
were constructed with the least absolute shrinkage and selection operator algorithm and
texture scores were calculated. The diagnostic performance of the 2D and 3D CTTA models
was evaluated with respect to calibration, discrimination, and clinical usefulness. Results: Of the 177 and 183 texture features extracted from 2D and 3D regions of interest,
respectively, 5 2D features and 8 3D features were selected to build 2D and 3D CTTA
models. The 2D CTTA model (area under the curve [AUC], 0.811; 95% confidence interval
[CI], 0.695-0.927) and the 3D CTTA model (AUC, 0.915; 95% CI, 0.838-0.993) showed good
discrimination and calibration (P > .05). There was no significant
difference in AUC between the 2 models (P = .093). Decision curve
analysis showed the 3D model outperformed the 2D model in terms of clinical
usefulness. Conclusions: The CTTA models based on contrast-enhanced CT images had a high value in
differentiating fpAML from chRCC.
Collapse
Affiliation(s)
- Guangjie Yang
- PET-CT Center, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Aidi Gong
- PET-CT Center, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Pei Nie
- Radiology Department, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Lei Yan
- PET-CT Center, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Wenjie Miao
- PET-CT Center, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Yujun Zhao
- PET-CT Center, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Jie Wu
- Pathology Department, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Jingjing Cui
- Huiying Medical Technology Co, Ltd, Beijing, China
| | - Yan Jia
- Huiying Medical Technology Co, Ltd, Beijing, China
| | - Zhenguang Wang
- PET-CT Center, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| |
Collapse
|
28
|
Computed tomography-based radiomics model for discriminating the risk stratification of gastrointestinal stromal tumors. Radiol Med 2020; 125:465-473. [PMID: 32048155 DOI: 10.1007/s11547-020-01138-6] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2019] [Accepted: 01/16/2020] [Indexed: 12/22/2022]
Abstract
PURPOSE The pathological risk degree of gastrointestinal stromal tumors (GISTs) has become an issue of great concern. Computed tomography (CT) is beneficial for showing adjacent tissues in detail and determining metastasis or recurrence of GISTs, but its function is still limited. Radiomics has recently shown a great potential in aiding clinical decision-making. The purpose of our study is to develop and validate CT-based radiomics models for GIST risk stratification. METHODS Three hundred and sixty-six patients clinically suspected of primary GISTs from January 2013 to February 2018 were retrospectively enrolled, among which data from 140 patients were eventually analyzed after exclusion. Data from patient CT images were partitioned based on the National Institutes of Health Consensus Classification, including tumor segmentation, radiomics feature extraction and selection. A radiomics model was then proposed and validated. RESULTS The radiomics signature demonstrated discriminative performance for advanced and nonadvanced GISTs with an area under the curve (AUC) of 0.935 [95% confidence interval (CI) 0.870-1.000] and an accuracy of 90.2% for validation cohort. The radiomics signature demonstrated favorable performance for the risk stratification of GISTs with an AUC of 0.809 (95% CI 0.777-0.841) and an accuracy of 67.5% for the validation cohort. Radiomics analysis could capture features of the four risk categories of GISTs. Meanwhile, this CT-based radiomics signature showed good diagnostic accuracy to distinguish between nonadvanced and advanced GISTs, as well as the four risk stratifications of GISTs. CONCLUSION Our findings highlight the potential of a quantitative radiomics analysis as a complementary tool to achieve an accurate diagnosis for GISTs.
Collapse
|
29
|
Fu J, Fang MJ, Dong D, Li J, Sun YS, Tian J, Tang L. Heterogeneity of metastatic gastrointestinal stromal tumor on texture analysis: DWI texture as potential biomarker of overall survival. Eur J Radiol 2020; 125:108825. [PMID: 32035324 DOI: 10.1016/j.ejrad.2020.108825] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2019] [Revised: 12/23/2019] [Accepted: 01/08/2020] [Indexed: 12/18/2022]
Abstract
PURPOSE To determine if texture features of diffusion weighted imaging (DWI) on MRI of metastatic gastrointestinal stromal tumor (mGIST) have correlation with overall survival (OS). METHOD Fifty-one GIST patients with metastatic lesions who received imatinib targeted therapy were included. Texture features of the largest metastatic lesion were analyzed using inhouse software. Three types of texture features were assessed: fractal features, gray-level co-occurrence matrix (GLCM) features, and gray-level run-length matrix (GLRLM) features. The features were extracted from the regions of interest (ROIs) on T2-weighted imaging (T2WI), DWI and apparent diffusion coefficient (ADC) maps. Histogram analysis was performed on ADC maps. Patients were followed up until death. Kaplan-Meier analysis was performed to determine the correlation of texture features with OS. The curves of the high- and low-risk groups were compared using log-rank test. The prognostic efficacy of the predictors was assessed by calculating the concordance probability. RESULTS The median survival time was 43.5 months (range, 3.97-120.90 m). Four DWI and three ADC texture features showed significant correlation with OS on univariate analysis (p < 0.05). DWI_L_GLCM_maximum_probability [hazard ratio (HR): 2.062 (1.357-3.131)], ADC_H_GLRLM_mean [HR: 2.174 (1.457-3.244)], and ADC_O_GLCM_cluster_shade [HR: 1.882 (1.324-2.674)] were identified as representative prognostic indicators. The optimum threshold levels for these three features were 1.19×100, 1.71×10 and 2.19×0.1, respectively. Neither histogram analysis values nor fractal features revealed significant correlation with survival status (p > 0.05). CONCLUSIONS Texture features of the mGIST on DWI exhibited correlation with overall survival. High-grade heterogeneity was associated with poor prognosis.
Collapse
Affiliation(s)
- Jia Fu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Radiology Department, Peking University Cancer Hospital & Institute, Beijing, 100142, China; Department of Radiology, Civil Aviation General Hospital, No. 1 Chaoyang Road, Chaoyang District, Beijing, 100123, China
| | - Meng-Jie Fang
- CAS Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, No.95 East Zhongguancun Road, Beijing, 100190, China; University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Di Dong
- CAS Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, No.95 East Zhongguancun Road, Beijing, 100190, China; University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Jian Li
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Departments of Gastroenterology, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Ying-Shi Sun
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Radiology Department, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Jie Tian
- CAS Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, No.95 East Zhongguancun Road, Beijing, 100190, China; University of Chinese Academy of Sciences, Beijing, 100049, China.
| | - Lei Tang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Radiology Department, Peking University Cancer Hospital & Institute, Beijing, 100142, China.
| |
Collapse
|
30
|
Wu G, Xie R, Li Y, Hou B, Morelli JN, Li X. Histogram analysis with computed tomography angiography for discriminating soft tissue sarcoma from benign soft tissue tumor. Medicine (Baltimore) 2020; 99:e18742. [PMID: 31914093 PMCID: PMC6959892 DOI: 10.1097/md.0000000000018742] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
To investigate the feasibility of histogram analysis with computed tomography angiography (CTA) in distinguishing between soft tissue sarcomas and benign soft tissue tumors. Fourty nine patients (23 men, mean age = 44.3 years, age range = 25-64) with pathologically-confirmed soft tissue sarcoma (n = 24) or benign soft tissue tumors (n = 25) in the lower extremities undergoing CTA for tumor evaluation were retrospectively analyzed. Two radiologists separately performed histogram analyses of CT density with CTA images by drawing a region of interest (ROI). The 10th (P10), 25th (P25), 50th (P50), 75th (P75), 90th percentiles (P90), mean, and standard deviations (SD) of measured tumor density were obtained along with measurements of the absolute value of kurtosis (AVK), absolute value of skewness (AVS), and inhomogeneity for each tumor. Intra-class correlation coefficients (ICC) were calculated to determine inter- and intra-reader variability in parameter measurements. The Mann-Whitney U test was used to compare histogram parameters between soft tissue sarcomas and benign soft tissue tumors. Receiver operator characteristic (ROC) curves were constructed to evaluate the accuracy of tumor discrimination. ICC was greater than 0.7 for AVS, AVK, and inhomogeneity, and >0.9 for mean, SD, and all percentile measures. There was no significant difference in P10, P25, P50, P75, P90, mean, or SD between soft tissue sarcomas and benign tumors (P > .05). AVS, AVK, and inhomogeneity were significantly higher in soft tissue sarcomas (P < .05). Areas under the curve (AUC) were 0.81, 0.83, and 0.84 for AVS, AVK, and inhomogeneity respectively. AUC were below 0.6 for mean, SD, and all percentiles.Skewness, kurtosis, and inhomogeneity measurements derived from histogram analysis from CTA distinguish between soft tissue sarcomas and benign soft tissue tumors.
Collapse
Affiliation(s)
- Gang Wu
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ruyi Xie
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yitong Li
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Bowen Hou
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | | | - Xiaoming Li
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
31
|
Chen C, Guo X, Wang J, Guo W, Ma X, Xu J. The Diagnostic Value of Radiomics-Based Machine Learning in Predicting the Grade of Meningiomas Using Conventional Magnetic Resonance Imaging: A Preliminary Study. Front Oncol 2019; 9:1338. [PMID: 31867272 PMCID: PMC6908490 DOI: 10.3389/fonc.2019.01338] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Accepted: 11/15/2019] [Indexed: 02/05/2023] Open
Abstract
Objective: The purpose of the current study is to investigate whether texture analysis-based machine learning algorithms could help devise a non-invasive imaging biomarker for accurate classification of meningiomas using machine learning algorithms. Method: The study cohort was established from the hospital database by reviewing the medical records. Patients were selected if they underwent meningioma resection in the neurosurgery department between January 2015 and December 2018. A total number of 40 texture parameters were extracted from pretreatment postcontrast T1-weighted (T1C) images based on six matrixes. Three feature selection methods were adopted, namely, distance correlation, least absolute shrinkage and selection operator (LASSO), and gradient boosting decision tree (GBDT). Multiclass classification methods of linear discriminant analysis (LDA) and support vector machine (SVM) algorithms were employed to establish the classification models. The diagnostic performances of models were evaluated with confusion matrix based on which the areas under the curve, accuracy, and Kappa value of models were calculated. Result: Confusion matrix showed that the LDA-based models represented better diagnostic performances than SVM-based models. The highest accuracy among LDA-based models was 75.6%, shown in the combination of Lasso + LDA. The optimal models for SVM-based models was Lasso+SVM, with accuracy of 59.0% in the testing group. One of the SVM-based models, GBDT+SVM, was overfitting, suggesting that this model was not suitable for application. Conclusion: Machine learning algorithms with texture features extracted from T1C images could potentially serve as the assistant imaging biomarkers for presurgically grading meningiomas.
Collapse
Affiliation(s)
- Chaoyue Chen
- Department of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, China.,State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, China.,Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, China
| | - Xinyi Guo
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, China.,West China School of Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Jian Wang
- School of Computer Science, Nanjing University of Science and Technology, Nanjing, China
| | - Wen Guo
- Department of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, China.,State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, China.,Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, China.,West China School of Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Xuelei Ma
- Department of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, China.,State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, China
| | - Jianguo Xu
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
32
|
Alessandrino F, Gujrathi R, Nassar AH, Alzaghal A, Ravi A, McGregor B, Sonpavde G, Shinagare AB. Predictive Role of Computed Tomography Texture Analysis in Patients with Metastatic Urothelial Cancer Treated with Programmed Death-1 and Programmed Death-ligand 1 Inhibitors. Eur Urol Oncol 2019; 3:680-686. [PMID: 31412003 DOI: 10.1016/j.euo.2019.02.002] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Revised: 01/31/2019] [Accepted: 02/14/2019] [Indexed: 12/31/2022]
Abstract
BACKGROUND Reliable biomarkers to predict the response of metastatic urothelial cancer (mUC) to programmed death-1 and programmed death-ligand 1 (PD-1/PD-L1) inhibitors are being investigated. Texture analysis represents tumor heterogeneity and may serve as a predictor of response in mUC. OBJECTIVE To assess the predictive ability of computed tomography (CT) texture analysis for progression-free survival (PFS) in patients with mUC treated with PD-1/PD-L1 inhibitors. DESIGN, SETTING, AND PARTICIPANTS Forty-two postplatinum patients with mUC treated with PD-1/PD-L1 inhibitors from 2013 to 2018, including those with measurable disease per RECIST 1.1 who had contrast-enhanced baseline or first follow-up CT within 3mo after starting treatment, were included. PFS was calculated based on serial follow-up CT scans. Eleven patients with follow-up of <12mo without progression were excluded. Texture features of measurable lesions on baseline and first follow-up CT were extracted using commercially available software (TexRAD; Feedback Plc, Cambridge, UK) using different spatial scaling factors (0, 2-6). OUTCOME MEASUREMENTS AND STATISTICAL ANALYSIS Stepwise logistic regression analysis was conducted to identify patients with PFS <12mo, and performance was assessed using receiver operator characteristic curves. RESULTS AND LIMITATIONS Of 31 included patients, 18 had PFS <12mo. Twenty-five baseline CT and 29 first follow-up CT scans met the inclusion criteria. In patients with PFS <12mo, entropy and mean were higher on first follow-up CT (p=0.02 and p=0.005, respectively). A predictive model including mean and entropy on first follow-up CT yielded 95% sensitivity, 80% specificity, 90% positive predictive value, 89% negative predictive value, and 90% accuracy (area under the curve=0.963) to identify patients with PFS <12mo. Limitations include retrospective nature and small sample size. CONCLUSIONS CT texture analysis can help predict early progression with high accuracy soon after starting PD-1/PD-L1 inhibitors. Studies investigating the correlation of texture analysis with survival endpoints may help validate texture analysis as a biomarker of PD-1/PD-L1 inhibitors' treatment response. PATIENT SUMMARY Computed tomography texture analysis can help predict durability of response in patients with metastatic urothelial cancer early during treatment with programmed death-1 and programmed death-ligand 1 (PD-1/PD-L1) inhibitors.
Collapse
Affiliation(s)
- Francesco Alessandrino
- Department of Imaging, Dana Farber Cancer Institute, Harvard Medical School, Boston, MA, USA; Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Rahul Gujrathi
- Department of Imaging, Dana Farber Cancer Institute, Harvard Medical School, Boston, MA, USA; Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Amin H Nassar
- Lank Center for Genitourinary Oncology, Dana Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Arwa Alzaghal
- Department of Imaging, Dana Farber Cancer Institute, Harvard Medical School, Boston, MA, USA; Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Arvind Ravi
- Lank Center for Genitourinary Oncology, Dana Farber Cancer Institute, Harvard Medical School, Boston, MA, USA; Broad Institute, Cambridge, MA, USA
| | - Bradley McGregor
- Lank Center for Genitourinary Oncology, Dana Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Guru Sonpavde
- Lank Center for Genitourinary Oncology, Dana Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Atul B Shinagare
- Department of Imaging, Dana Farber Cancer Institute, Harvard Medical School, Boston, MA, USA; Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
33
|
Sun ZQ, Hu SD, Li J, Wang T, Duan SF, Wang J. Radiomics study for differentiating gastric cancer from gastric stromal tumor based on contrast-enhanced CT images. JOURNAL OF X-RAY SCIENCE AND TECHNOLOGY 2019; 27:1021-1031. [PMID: 31640109 DOI: 10.3233/xst-190574] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
PURPOSE To test the feasibility of differentiate gastric cancer from gastric stromal tumor using a radiomics study based on contrast-enhanced CT images. MATERIALS AND METHODS The contrast-enhanced CT image data of 60 patients with gastric cancer and 40 patients with gastric stromal tumor confirmed by postoperative pathology were retrospectively analyzed. First, CT images were read by two senior radiologists to acquire subjective CT signs model, including perigastric fatty infiltration, perigastric enlarged lymph nodes, the enhancement and growth modes of gastric tumors. Second, the manual segmentation of gastric tumors from the CT images was performed by the two radiologists to extract radiomics features via ITK-SNAP software, and to construct radiomics signature model. Finally, a diagnostic model integrated with subjective CT signs and radiomics signatures was constructed. The diagnostic efficacy of three models in differentiating gastric cancer from gastric stromal tumor was compared by using receiver operating characteristic curves (ROC). RESULTS There are statistically significant differences between the gastric cancer and gastric stromal tumor in the perigastric enlarged lymph nodes, growth mode and radiomics signature (p < 0.05). The area under ROC curve (AUC), sensitivity and accuracy of subjective CT signs model were the lowest among the three models. While the combined model yields the highest AUC value (0.903), specificity (93.33%) and accuracy (86.00%) among the three models (p = 0.03). CONCLUSION The diagnostic model integrating subjective CT signs and radiomics signature can improve the diagnostic accuracy of gastric tumors.
Collapse
Affiliation(s)
- Zong-Qiong Sun
- Department of Radiology, Affiliated Hospital of Jiangnan University, The Fourth People's Hospital of Wuxi City, Jiangsu Province, China
| | - Shu-Dong Hu
- Department of Radiology, Affiliated Hospital of Jiangnan University, The Fourth People's Hospital of Wuxi City, Jiangsu Province, China
| | - Jie Li
- Department of Intervention Affiliated Hospital of Jiangnan University, The Fourth People's Hospital of Wuxi City, Jiangsu Province, China
| | - Teng Wang
- Department of Oncology, Affiliated Hospital of Jiangnan University, The Fourth People's Hospital of Wuxi City, Jiangsu Province, China
| | - Shao-Feng Duan
- General Electric Company (GE) Healthcare China, Pudong New Town, Shanghai, China
| | - Jun Wang
- Shanghai Institute for Advanced Communication and Data Science, School of Communication and Information Engineering, Shanghai University, Shanghai, China
| |
Collapse
|