1
|
Pecchi A, Bozzola C, Beretta C, Besutti G, Toss A, Cortesi L, Balboni E, Nocetti L, Ligabue G, Torricelli P. DCE-MRI Radiomic analysis in triple negative ductal invasive breast cancer. Comparison between BRCA and not BRCA mutated patients: Preliminary results. Magn Reson Imaging 2024; 113:110214. [PMID: 39047852 DOI: 10.1016/j.mri.2024.110214] [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: 05/13/2024] [Revised: 07/17/2024] [Accepted: 07/20/2024] [Indexed: 07/27/2024]
Abstract
OBJECTIVE The research aimed to determine whether and which radiomic features from breast dynamic contrast enhanced (DCE) MRI could predict the presence of BRCA1 mutation in patients with triple-negative breast cancer (TNBC). MATERIAL AND METHODS This retrospective study included consecutive patients histologically diagnosed with TNBC who underwent breast DCE-MRI in 2010-2021. Baseline DCE-MRIs were retrospectively reviewed; percentage maps of wash-in and wash-out were computed and breast lesions were manually segmented, drawing a 5 mm-Region of Interest (ROI) inside the tumor and another 5 mm-ROI inside the contralateral healthy gland. Features for each map and each ROI were extracted with Pyradiomics-3D Slicer and considered first separately (tumor and contralateral gland) and then together. In each analysis the more important features for BRCA1 status classification were selected with Maximum Relevance Minimum Redundancy algorithm and used to fit four classifiers. RESULTS The population included 67 patients and 86 lesions (21 in BRCA1-mutated, 65 in non BRCA-carriers). The best classifiers for BRCA mutation were Support Vector Classifier and Logistic Regression in models fitted with both gland and tumor features, reaching an Area Under ROC Curve (AUC) of 0.80 (SD 0.21) and of 0.79 (SD 0.20), respectively. Three features were higher in BRCA1-mutated compared to non BRCA-mutated: Total Energy and Correlation from gray level cooccurrence matrix, both measured in contralateral gland in wash-out maps, and Root Mean Squared, selected from the wash-out map of the tumor. CONCLUSIONS This study showed the feasibility of a radiomic study with breast DCE-MRI and the potential of radiomics in predicting BRCA1 mutational status.
Collapse
Affiliation(s)
- Annarita Pecchi
- Division of Radiology, Department of Medical and Surgical Sciences of Children and Adults, University of Modena and Reggio Emilia, 41224 Modena, Italy
| | - Chiara Bozzola
- Division of Radiology, Department of Medical and Surgical Sciences of Children and Adults, University of Modena and Reggio Emilia, 41224 Modena, Italy
| | - Cecilia Beretta
- Division of Radiology, Department of Medical and Surgical Sciences of Children and Adults, University of Modena and Reggio Emilia, 41224 Modena, Italy
| | - Giulia Besutti
- Division of Radiology, Department of Medical and Surgical Sciences of Children and Adults, University of Modena and Reggio Emilia, 41224 Modena, Italy; Radiology Unit, Department of Diagnostic Imaging and Laboratory Medicine, AUSL-IRCCS di Reggio Emilia, Via Risorgimento 80, 42123 Reggio Emilia, Italy.
| | - Angela Toss
- Division of Oncology, Department of Medical and Surgical Sciences of Children and Adults, University of Modena and Reggio Emilia, 41224 Modena, Italy
| | - Laura Cortesi
- Division of Oncology, Department of Medical and Surgical Sciences of Children and Adults, University of Modena and Reggio Emilia, 41224 Modena, Italy
| | - Erica Balboni
- Medical Physics Unit, University Hospital of Modena, 41124 Modena, Italy
| | - Luca Nocetti
- Medical Physics Unit, University Hospital of Modena, 41124 Modena, Italy
| | - Guido Ligabue
- Division of Radiology, Department of Medical and Surgical Sciences of Children and Adults, University of Modena and Reggio Emilia, 41224 Modena, Italy
| | - Pietro Torricelli
- Division of Radiology, Department of Medical and Surgical Sciences of Children and Adults, University of Modena and Reggio Emilia, 41224 Modena, Italy
| |
Collapse
|
2
|
Zhang J, Wang Q, Guo TH, Gao W, Yu YM, Wang RF, Yu HL, Chen JJ, Sun LL, Zhang BY, Wang HJ. Computed tomography-based radiomic model for the prediction of neoadjuvant immunochemotherapy response in patients with advanced gastric cancer. World J Gastrointest Oncol 2024; 16:4115-4128. [DOI: 10.4251/wjgo.v16.i10.4115] [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: 05/16/2024] [Revised: 08/18/2024] [Accepted: 08/28/2024] [Indexed: 09/26/2024] Open
Abstract
BACKGROUND Neoadjuvant immunochemotherapy (nICT) has emerged as a popular treatment approach for advanced gastric cancer (AGC) in clinical practice worldwide. However, the response of AGC patients to nICT displays significant heterogeneity, and no existing radiomic model utilizes baseline computed tomography to predict treatment outcomes.
AIM To establish a radiomic model to predict the response of AGC patients to nICT.
METHODS Patients with AGC who received nICT (n = 60) were randomly assigned to a training cohort (n = 42) or a test cohort (n = 18). Various machine learning models were developed using selected radiomic features and clinical risk factors to predict the response of AGC patients to nICT. An individual radiomic nomogram was established based on the chosen radiomic signature and clinical signature. The performance of all the models was assessed through receiver operating characteristic curve analysis, decision curve analysis (DCA) and the Hosmer-Lemeshow goodness-of-fit test.
RESULTS The radiomic nomogram could accurately predict the response of AGC patients to nICT. In the test cohort, the area under curve was 0.893, with a 95% confidence interval of 0.803-0.991. DCA indicated that the clinical application of the radiomic nomogram yielded greater net benefit than alternative models.
CONCLUSION A nomogram combining a radiomic signature and a clinical signature was designed to predict the efficacy of nICT in patients with AGC. This tool can assist clinicians in treatment-related decision-making.
Collapse
Affiliation(s)
- Jun Zhang
- Department of Radiation Oncology, Affiliated Hospital of Qingdao University, Qingdao 266000, Shandong Province, China
| | - Qi Wang
- Department of Radiation Oncology, Affiliated Hospital of Qingdao University, Qingdao 266000, Shandong Province, China
| | - Tian-Hui Guo
- Department of Radiation Oncology, Affiliated Hospital of Qingdao University, Qingdao 266000, Shandong Province, China
| | - Wen Gao
- Department of Radiation Oncology, Affiliated Hospital of Qingdao University, Qingdao 266000, Shandong Province, China
| | - Yi-Miao Yu
- Department of Radiation Oncology, Affiliated Hospital of Qingdao University, Qingdao 266000, Shandong Province, China
| | - Rui-Feng Wang
- Department of Radiation Oncology, Affiliated Hospital of Qingdao University, Qingdao 266000, Shandong Province, China
| | - Hua-Long Yu
- Department of Radiology, Affiliated Hospital of Qingdao University, Qingdao 266000, Shandong Province, China
| | - Jing-Jing Chen
- Department of Radiology, Affiliated Hospital of Qingdao University, Qingdao 266000, Shandong Province, China
| | - Ling-Ling Sun
- Department of Pathology, Affiliated Hospital of Qingdao University, Qingdao 266000, Shandong Province, China
| | - Bi-Yuan Zhang
- Department of Radiation Oncology, Affiliated Hospital of Qingdao University, Qingdao 266000, Shandong Province, China
| | - Hai-Ji Wang
- Department of Radiation Oncology, Affiliated Hospital of Qingdao University, Qingdao 266000, Shandong Province, China
| |
Collapse
|
3
|
Chen W, Zhang W, Chen X, Dong W, Cai Y, Cheng J, Jin J. Computed tomography-based radiomics nomogram for predicting therapeutic response to neoadjuvant chemotherapy in locally advanced gastric cancer : A scale for treatment predicting. Clin Transl Oncol 2024; 26:1944-1955. [PMID: 38467894 DOI: 10.1007/s12094-024-03417-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Accepted: 02/20/2024] [Indexed: 03/13/2024]
Abstract
BACKGROUND AND OBJECTIVE Neoadjuvant chemotherapy results in various responses when used to treat locally advanced gastric cancer, we aimed to develop and validate a predictive model of the response to neoadjuvant chemotherapy in patients with gastric cancer. METHODS A total of 128 patients with locally advanced gastric cancer who underwent pre-treatment computed tomography (CT) scanning followed by neoadjuvant chemoradiotherapy were included (training cohort: n = 64; validation cohort: n = 64). We built a radiomics score combined with laboratory parameters to create a nomogram for predicting the efficacy of neoadjuvant chemotherapy and calculating scores for risk factors. RESULTS The radiomics score system demonstrated good stability and prediction performance for the response to neoadjuvant chemotherapy, with the area under the curve of the training and validation cohorts being 0.8 and 0.64, respectively. The radiomics score proved to be an independent risk factor affecting the efficacy of neoadjuvant chemotherapy. In addition to the radiomics score, four other risk factors were included in the nomogram, namely the platelet-to-lymphocyte ratio, total bilirubin, ALT/AST, and CA199. The model had a C-index of 0.8. CONCLUSIONS Our results indicated that radiomics features could be potential biomarkers for the early prediction of the response to neoadjuvant treatment.
Collapse
Affiliation(s)
- Wenjing Chen
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Weiteng Zhang
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Xietao Chen
- School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Weisong Dong
- Department of Pathology, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yiqi Cai
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Jun Cheng
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China.
| | - Jinji Jin
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China.
| |
Collapse
|
4
|
Zhang W, Wang S, Dong Q, Chen W, Wang P, Zhu G, Chen X, Cai Y. Predictive nomogram for lymph node metastasis and survival in gastric cancer using contrast-enhanced computed tomography-based radiomics: a retrospective study. PeerJ 2024; 12:e17111. [PMID: 38525272 PMCID: PMC10960528 DOI: 10.7717/peerj.17111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Accepted: 02/23/2024] [Indexed: 03/26/2024] Open
Abstract
Background Lymph node involvement significantly impacts the survival of gastric cancer patients and is a crucial factor in determining the appropriate treatment. This study aimed to evaluate the potential of enhanced computed tomography (CT)-based radiomics in predicting lymph node metastasis (LNM) and survival in patients with gastric cancer before surgery. Methods Retrospective analysis of clinical data from 192 patients diagnosed with gastric carcinoma was conducted. The patients were randomly divided into a training cohort (n = 128) and a validation cohort (n = 64). Radiomic features of CT images were extracted using the Pyradiomics software platform, and distinctive features were further selected using a Lasso Cox regression model. Features significantly associated with LNM were identified through univariate and multivariate analyses and combined with radiomic scores to create a nomogram model for predicting lymph node involvement before surgery. The predictive performance of radiomics features, CT-reported lymph node status, and the nomogram model for LNM were compared in the training and validation cohorts by plotting receiver operating characteristic (ROC) curves. High-risk and low-risk groups were identified in both cohorts based on the cut-off value of 0.582 within the radiomics evaluation scheme, and survival rates were compared. Results Seven radiomic features were identified and selected, and patients were stratified into high-risk and low-risk groups using a 0.582 cut-off radiomics score. Univariate and multivariate analyses revealed that radiomics features, diabetes mellitus, Nutrition Risk Screening (NRS) 2002 score, and CT-reported lymph node status were significant predictors of LNM in patients with gastric cancer. A predictive nomogram model was developed by combining these predictors with the radiomics score, which accurately predicted LNM in gastric cancer patients before surgery and outperformed other models in terms of accuracy and sensitivity. The AUC values for the training and validation cohorts were 0.82 and 0.722, respectively. The high-risk and low-risk groups in both the training and validation cohorts showed significant differences in survival rates. Conclusion The radiomics nomogram, based on contrast-enhanced computed tomography (CECT ), is a promising non-invasive tool for preoperatively predicting LNM in gastric cancer patients and postoperative survival.
Collapse
Affiliation(s)
- Weiteng Zhang
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Sujun Wang
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Qiantong Dong
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Wenjing Chen
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Pengfei Wang
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Guanbao Zhu
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Xiaolei Chen
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Yiqi Cai
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
5
|
Li J, Chen Z, Chen Y, Zhao J, He M, Li X, Zhang L, Dong B, Zhang X, Tang L, Shen L. CT-based delta radiomics in predicting the prognosis of stage IV gastric cancer to immune checkpoint inhibitors. Front Oncol 2023; 12:1059874. [PMID: 36686828 PMCID: PMC9847891 DOI: 10.3389/fonc.2022.1059874] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2022] [Accepted: 11/29/2022] [Indexed: 01/06/2023] Open
Abstract
Introduction To explore the prognostic value of CT-based delta radiomics in predicting the prognosis of patients with stage IV gastric cancer treated with immune checkpoint inhibitors (ICI). Materials and methods Forty-two patients with stage IV gastric cancer, who had received ICI monotherapy, were enrolled in this retrospective study. Baseline and first follow-up CT scans were analyzed. Intratumoral and peritumoral regions of interest (ROI) were contoured, enabling the extraction of 192 features from each ROI. The intraclass correlation coefficients were used to select features with high stability. The least absolute shrinkage and selection operator was used to select features with high weights for predicting patient prognosis. Kaplan-Meier analysis and log-rank test were performed to explore the association between features and progression free survival (PFS). Cox regression analyses were used to identify predictors for PFS. The C-index was used to assess the prediction performance of features. Results Two radiomics features of ΔVintra_ZV and postVperi_Sphericity were identified from intratumoral and peritumoral regions, respectively. The Kaplan-Meier analysis revealed significant differences in PFS between patients with low and high feature value (ΔVintra_ZV: P=0.000; postVperi_Sphericity: P=0.012), and the multivariable cox analysis demonstrated that ΔVintra_ZV was independent predictor for PFS (HR, 1.911; 95% CI: 1.163-3.142; P=0.011), with C-index of 0.705. Conclusions Based on CT scans at baseline and first follow-up, the delta radiomics features could efficiently predict the PFS of gastric cancer patients treated with ICI therapy.
Collapse
Affiliation(s)
- Jiazheng Li
- Department of Radiology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital and Institute, Beijing, China
| | - Zifan Chen
- Center for Data Science, Peking University, Beijing, China
| | - Yang Chen
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital and Institute, Beijing, China
| | - Jie Zhao
- National Engineering Laboratory for Big Data Analysis and Applications, Peking University, Beijing, China
| | - Meng He
- Department of Radiology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital and Institute, Beijing, China
| | - Xiaoting Li
- Department of Radiology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital and Institute, Beijing, China
| | - Li Zhang
- Center for Data Science, Peking University, Beijing, China
| | - Bin Dong
- Beijing International Center for Mathematical Research (BICMR), Peking University, Beijing, China,*Correspondence: Bin Dong, ; Xiaotian Zhang, ; Lei Tang, ; Lin Shen,
| | - Xiaotian Zhang
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital and Institute, Beijing, China,*Correspondence: Bin Dong, ; Xiaotian Zhang, ; Lei Tang, ; Lin Shen,
| | - Lei Tang
- Department of Radiology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital and Institute, Beijing, China,*Correspondence: Bin Dong, ; Xiaotian Zhang, ; Lei Tang, ; Lin Shen,
| | - Lin Shen
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital and Institute, Beijing, China,*Correspondence: Bin Dong, ; Xiaotian Zhang, ; Lei Tang, ; Lin Shen,
| |
Collapse
|
6
|
Ma T, Cui J, Wang L, Li H, Ye Z, Gao X. A CT-based radiomics signature for prediction of HER2 overexpression and treatment efficacy of trastuzumab in advanced gastric cancer. Transl Cancer Res 2022; 11:4326-4337. [PMID: 36644192 PMCID: PMC9834583 DOI: 10.21037/tcr-22-1690] [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: 06/15/2022] [Accepted: 09/26/2022] [Indexed: 11/20/2022]
Abstract
Background Accurate evaluation of human epidermal growth factor receptor 2 (HER2) status is very important for appropriate management of advanced gastric cancer (AGC) patients. In this study, we aimed to develop and validate a computed tomography (CT)-based radiomics signature for preoperative prediction of HER2 overexpression and treatment efficacy of trastuzumab in AGC. Methods We retrospectively enrolled 536 consecutive AGC patients (median age, 59 years; interquartile range, 52-65 years; 377 male, 159 female) and separated them into a training set (n=357) and a testing set (n=179). Radiomic features were extracted from 3 different phase images of contrast-enhanced CT scans, and a radiomics signature was built based on highly reproducible features using the least absolute shrinkage and selection operator (LASSO) method. The predictive performance of the radiomics signature was assessed in the training and testing sets. Univariable and multivariable logistical regression analyses were used to identify independent risk factors of HER2 overexpression. Univariable and multivariable Cox regression analyses were used to identify the risk factors of overall survival (OS) and progression-free survival (PFS). The predictive value of the radiomics signature for treatment efficacy of trastuzumab was also evaluated. Results The radiomics signature comprised eight robust features that demonstrated good discrimination ability for HER2 overexpression in the training set [area under the curve (AUC) =0.85] and the testing set (AUC =0.81). Multivariable Cox regression analysis revealed that the radiomics signature was an independent risk factor for OS [hazard ratio (HR) =2.01, P=0.001] and PFS (HR =1.32, P=0.01). The radiomics score of patients who achieved disease control was significantly lower than that of patients with progressive disease (P=0.023). Conclusions The proposed radiomics signature showed favorable accuracy for prediction of HER2 overexpression and prognosis in AGC. It has promising potential as a noninvasive approach for selecting patients for target therapy.
Collapse
Affiliation(s)
- Tingting Ma
- Department of Radiology, Tianjin Cancer Hospital Airport Hospital, Tianjin, China;,Department of Radiology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China;,National Clinical Research Center for Cancer, Tianjin, China;,Tianjin’s Clinical Research Center for Cancer, Tianjin, China
| | - Jingli Cui
- National Clinical Research Center for Cancer, Tianjin, China;,Tianjin’s Clinical Research Center for Cancer, Tianjin, China;,Department of Bone and Soft Tissue Tumor, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China;,Department of General Surgery, Weifang People’s Hospital, Weifang, China
| | - Lingwei Wang
- Department of Radiology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China;,National Clinical Research Center for Cancer, Tianjin, China;,Tianjin’s Clinical Research Center for Cancer, Tianjin, China;,The Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - Hui Li
- National Clinical Research Center for Cancer, Tianjin, China;,Department of Gastrointestinal Cancer Biology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China;,Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, China
| | - Zhaoxiang Ye
- Department of Radiology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China;,National Clinical Research Center for Cancer, Tianjin, China;,Tianjin’s Clinical Research Center for Cancer, Tianjin, China;,The Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - Xujie Gao
- Department of Radiology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China;,National Clinical Research Center for Cancer, Tianjin, China;,Tianjin’s Clinical Research Center for Cancer, Tianjin, China;,The Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| |
Collapse
|
7
|
Ma T, Cui J, Wang L, Li H, Ye Z, Gao X. A multiphase contrast-enhanced CT radiomics model for prediction of human epidermal growth factor receptor 2 status in advanced gastric cancer. Front Genet 2022; 13:968027. [PMID: 36276942 PMCID: PMC9585247 DOI: 10.3389/fgene.2022.968027] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Accepted: 09/27/2022] [Indexed: 11/16/2022] Open
Abstract
Background: Accurate evaluation of human epidermal growth factor receptor 2 (HER2) status is of great importance for appropriate management of advanced gastric cancer (AGC) patients. This study aims to develop and validate a CT-based radiomics model for prediction of HER2 overexpression in AGC. Materials and Methods: Seven hundred and forty-five consecutive AGC patients (median age, 59 years; interquartile range, 52–66 years; 515 male and 230 female) were enrolled and separated into training set (n = 521) and testing set (n = 224) in this retrospective study. Radiomics features were extracted from three phases images of contrast-enhanced CT scans. A radiomics signature was built based on highly reproducible features using the least absolute shrinkage and selection operator method. Univariable and multivariable logistical regression analysis were used to establish predictive model with independent risk factors of HER2 overexpression. The predictive performance of radiomics model was assessed in the training and testing sets. Results: The positive rate of HER2 was 15.9% and 13.8% in the training set and testing set, respectively. The positive rate of HER2 in intestinal-type GC was significantly higher than that in diffuse-type GC. The radiomics signature comprised eight robust features demonstrated good discrimination ability for HER2 overexpression in the training set (AUC = 0.84) and the testing set (AUC = 0.78). A radiomics-based model that incorporated radiomics signature and pathological type showed good discrimination and calibration in the training (AUC = 0.85) and testing (AUC = 0.84) sets. Conclusion: The proposed radiomics model showed favorable accuracy for prediction of HER2 overexpression in AGC.
Collapse
Affiliation(s)
- Tingting Ma
- Department of Radiology, Tianjin Cancer Hospital Airport Hospital, Tianjin, China
- Department of Radiology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- National Clinical Research Center for Cancer, Tianjin, China
- Tianjin’s Clinical Research Center for Cancer, Tianjin, China
| | - Jingli Cui
- National Clinical Research Center for Cancer, Tianjin, China
- Tianjin’s Clinical Research Center for Cancer, Tianjin, China
- Department of Bone and Soft Tissue Tumor, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- Department of General Surgery, Weifang People’s Hospital, Weifang, Shandong, China
| | - Lingwei Wang
- Department of Radiology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- National Clinical Research Center for Cancer, Tianjin, China
- Tianjin’s Clinical Research Center for Cancer, Tianjin, China
- The Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - Hui Li
- National Clinical Research Center for Cancer, Tianjin, China
- Department of Gastrointestinal Cancer Biology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, China
| | - Zhaoxiang Ye
- Department of Radiology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- National Clinical Research Center for Cancer, Tianjin, China
- Tianjin’s Clinical Research Center for Cancer, Tianjin, China
- The Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
- *Correspondence: Zhaoxiang Ye, ; Xujie Gao,
| | - Xujie Gao
- Department of Radiology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- National Clinical Research Center for Cancer, Tianjin, China
- Tianjin’s Clinical Research Center for Cancer, Tianjin, China
- The Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
- *Correspondence: Zhaoxiang Ye, ; Xujie Gao,
| |
Collapse
|
8
|
Chen Q, Zhang L, Liu S, You J, Chen L, Jin Z, Zhang S, Zhang B. Radiomics in precision medicine for gastric cancer: opportunities and challenges. Eur Radiol 2022; 32:5852-5868. [PMID: 35316364 DOI: 10.1007/s00330-022-08704-8] [Citation(s) in RCA: 42] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2021] [Revised: 02/20/2022] [Accepted: 02/28/2022] [Indexed: 01/19/2023]
Abstract
OBJECTIVES Radiomic features derived from routine medical images show great potential for personalized medicine in gastric cancer (GC). We aimed to evaluate the current status and quality of radiomic research as well as its potential for identifying biomarkers to predict therapy response and prognosis in patients with GC. METHODS We performed a systematic search of the PubMed and Embase databases for articles published from inception through July 10, 2021. The phase classification criteria for image mining studies and the radiomics quality scoring (RQS) tool were applied to evaluate scientific and reporting quality. RESULTS Twenty-five studies consisting of 10,432 patients were included. 96% of studies extracted radiomic features from CT images. Association between radiomic signature and therapy response was evaluated in seven (28%) studies; association with survival was evaluated in 17 (68%) studies; one (4%) study analyzed both. All results of the included studies showed significant associations. Based on the phase classification criteria for image mining studies, 18 (72%) studies were classified as phase II, with two, four, and one studies as discovery science, phase 0 and phase I, respectively. The median RQS score for the radiomic studies was 44.4% (range, 0 to 55.6%). There was extensive heterogeneity in the study population, tumor stage, treatment protocol, and radiomic workflow amongst the studies. CONCLUSIONS Although radiomic research in GC is highly heterogeneous and of relatively low quality, it holds promise for predicting therapy response and prognosis. Efforts towards standardization and collaboration are needed to utilize radiomics for clinical application. KEY POINTS • Radiomics application of gastric cancer is increasingly being reported, particularly in predicting therapy response and survival. • Although radiomics research in gastric cancer is highly heterogeneous and relatively low quality, it holds promise for predicting clinical outcomes. • Standardized imaging protocols and radiomic workflow are needed to facilitate radiomics into clinical use.
Collapse
Affiliation(s)
- Qiuying Chen
- Department of Radiology, the First Affiliated Hospital, Jinan University, No.613, Huangpu West Road, Tianhe District, Guangzhou, 510627, Guangdong, China.,Graduate College, Jinan University, Guangzhou, Guangdong, China
| | - Lu Zhang
- Department of Radiology, the First Affiliated Hospital, Jinan University, No.613, Huangpu West Road, Tianhe District, Guangzhou, 510627, Guangdong, China.,Graduate College, Jinan University, Guangzhou, Guangdong, China
| | - Shuyi Liu
- Department of Radiology, the First Affiliated Hospital, Jinan University, No.613, Huangpu West Road, Tianhe District, Guangzhou, 510627, Guangdong, China.,Graduate College, Jinan University, Guangzhou, Guangdong, China
| | - Jingjing You
- Department of Radiology, the First Affiliated Hospital, Jinan University, No.613, Huangpu West Road, Tianhe District, Guangzhou, 510627, Guangdong, China.,Graduate College, Jinan University, Guangzhou, Guangdong, China
| | - Luyan Chen
- Department of Radiology, the First Affiliated Hospital, Jinan University, No.613, Huangpu West Road, Tianhe District, Guangzhou, 510627, Guangdong, China.,Graduate College, Jinan University, Guangzhou, Guangdong, China
| | - Zhe Jin
- Department of Radiology, the First Affiliated Hospital, Jinan University, No.613, Huangpu West Road, Tianhe District, Guangzhou, 510627, Guangdong, China.,Graduate College, Jinan University, Guangzhou, Guangdong, China
| | - Shuixing Zhang
- Department of Radiology, the First Affiliated Hospital, Jinan University, No.613, Huangpu West Road, Tianhe District, Guangzhou, 510627, Guangdong, China. .,Graduate College, Jinan University, Guangzhou, Guangdong, China.
| | - Bin Zhang
- Department of Radiology, the First Affiliated Hospital, Jinan University, No.613, Huangpu West Road, Tianhe District, Guangzhou, 510627, Guangdong, China. .,Graduate College, Jinan University, Guangzhou, Guangdong, China.
| |
Collapse
|
9
|
Computed Tomography Texture Features and Risk Factor Analysis of Postoperative Recurrence of Patients with Advanced Gastric Cancer after Radical Treatment under Artificial Intelligence Algorithm. COMPUTATIONAL INTELLIGENCE AND NEUROSCIENCE 2022; 2022:1852718. [PMID: 35655504 PMCID: PMC9155975 DOI: 10.1155/2022/1852718] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 04/25/2022] [Accepted: 05/05/2022] [Indexed: 12/12/2022]
Abstract
Computer tomography texture analysis (CTTA) based on the V-Net convolutional neural network (CNN) algorithm was used to analyze the recurrence of advanced gastric cancer after radical treatment. Meanwhile, the clinical characteristics of patients were analyzed to explore the recurrence factors. 86 patients who underwent the advanced radical gastrectomy for gastric cancer were retrospectively selected as the research objects. Patients were divided into the no-recurrence group (30 cases) and the recurrence group (56 cases) according to whether there was recurrence after radical treatment. CTTA was performed before and after surgery in both groups to analyze the risk factors for recurrence. The results showed that the dice coefficient (0.9209) and the intersection over union (IOU) value (0.8392) of the V–CNN segmentation effect were signally higher than those of CNN, V-Net, and context encoder network (CE-Net) (P < 0.05). The mean value of arterial phase and portal phase (65.29 ± 9.23)/(79.89 ± 10.83), kurtosis (3.22)/(3.13), entropy (9.99 ± 0.53)/(9.97 ± 0.83), and correlation (4.12 × 10−5/4.21 × 10−5) of the recurrence group was higher than the no-recurrence group, while the skewness (0.01)/(−0.06) of the recurrence group was lower than that of the no-recurrence group (P < 0.05). Patients aged 60 years old and above, with a tumor diameter of 6 cm and above, and in the stage III/IV in the recurrence group were higher than those in the no-recurrence group, and patients with chemotherapy were lower (P < 0.05). To sum up, age, tumor diameter, whether chemotherapy should be performed, and tumor staging were all the risk factors of postoperative recurrence among patients with gastric cancer. Besides, CT texture parameter could be used to predict and analyze the postoperative recurrence of gastric cancer with good clinical application values.
Collapse
|
10
|
Kashyap A, Rapsomaniki MA, Barros V, Fomitcheva-Khartchenko A, Martinelli AL, Rodriguez AF, Gabrani M, Rosen-Zvi M, Kaigala G. Quantification of tumor heterogeneity: from data acquisition to metric generation. Trends Biotechnol 2021; 40:647-676. [PMID: 34972597 DOI: 10.1016/j.tibtech.2021.11.006] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Revised: 11/26/2021] [Accepted: 11/29/2021] [Indexed: 01/18/2023]
Abstract
Tumors are unique and complex ecosystems, in which heterogeneous cell subpopulations with variable molecular profiles, aggressiveness, and proliferation potential coexist and interact. Understanding how heterogeneity influences tumor progression has important clinical implications for improving diagnosis, prognosis, and treatment response prediction. Several recent innovations in data acquisition methods and computational metrics have enabled the quantification of spatiotemporal heterogeneity across different scales of tumor organization. Here, we summarize the most promising efforts from a common experimental and computational perspective, discussing their advantages, shortcomings, and challenges. With personalized medicine entering a new era of unprecedented opportunities, our vision is that of future workflows integrating across modalities, scales, and dimensions to capture intricate aspects of the tumor ecosystem and to open new avenues for improved patient care.
Collapse
Affiliation(s)
- Aditya Kashyap
- IBM Research Europe -Säumerstrasse 4, Rüschlikon CH-8803, Zurich, Switzerland
| | | | - Vesna Barros
- Department of Healthcare Informatics, IBM Research, IBM R&D Labs, University of Haifa Campus, Mount Carmel, Haifa, 3498825, Israel; The Hebrew University, The Edmond J. Safra Campus - Givat Ram, Jerusalem, 9190401, Israel
| | - Anna Fomitcheva-Khartchenko
- IBM Research Europe -Säumerstrasse 4, Rüschlikon CH-8803, Zurich, Switzerland; Eidgenössische Technische Hochschule (ETH-Zurich), Vladimir-Prelog-Weg 1-5/10, 8099 Zurich, Switzerland
| | | | | | - Maria Gabrani
- IBM Research Europe -Säumerstrasse 4, Rüschlikon CH-8803, Zurich, Switzerland
| | - Michal Rosen-Zvi
- Department of Healthcare Informatics, IBM Research, IBM R&D Labs, University of Haifa Campus, Mount Carmel, Haifa, 3498825, Israel; The Hebrew University, The Edmond J. Safra Campus - Givat Ram, Jerusalem, 9190401, Israel
| | - Govind Kaigala
- IBM Research Europe -Säumerstrasse 4, Rüschlikon CH-8803, Zurich, Switzerland.
| |
Collapse
|
11
|
Chidambaram S, Sounderajah V, Maynard N, Markar SR. Diagnostic Performance of Artificial Intelligence-Centred Systems in the Diagnosis and Postoperative Surveillance of Upper Gastrointestinal Malignancies Using Computed Tomography Imaging: A Systematic Review and Meta-Analysis of Diagnostic Accuracy. Ann Surg Oncol 2021; 29:1977-1990. [PMID: 34762214 PMCID: PMC8810479 DOI: 10.1245/s10434-021-10882-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Accepted: 09/11/2021] [Indexed: 12/24/2022]
Abstract
Background Upper gastrointestinal cancers are aggressive malignancies with poor prognosis, even following multimodality therapy. As such, they require timely and accurate diagnostic and surveillance strategies; however, such radiological workflows necessitate considerable expertise and resource to maintain. In order to lessen the workload upon already stretched health systems, there has been increasing focus on the development and use of artificial intelligence (AI)-centred diagnostic systems. This systematic review summarizes the clinical applicability and diagnostic performance of AI-centred systems in the diagnosis and surveillance of esophagogastric cancers. Methods A systematic review was performed using the MEDLINE, EMBASE, Cochrane Review, and Scopus databases. Articles on the use of AI and radiomics for the diagnosis and surveillance of patients with esophageal cancer were evaluated, and quality assessment of studies was performed using the QUADAS-2 tool. A meta-analysis was performed to assess the diagnostic accuracy of sequencing methodologies. Results Thirty-six studies that described the use of AI were included in the qualitative synthesis and six studies involving 1352 patients were included in the quantitative analysis. Of these six studies, four studies assessed the utility of AI in gastric cancer diagnosis, one study assessed its utility for diagnosing esophageal cancer, and one study assessed its utility for surveillance. The pooled sensitivity and specificity were 73.4% (64.6–80.7) and 89.7% (82.7–94.1), respectively. Conclusions AI systems have shown promise in diagnosing and monitoring esophageal and gastric cancer, particularly when combined with existing diagnostic methods. Further work is needed to further develop systems of greater accuracy and greater consideration of the clinical workflows that they aim to integrate within.
Collapse
Affiliation(s)
| | - Viknesh Sounderajah
- Department of Surgery and Cancer, Imperial College London, London, UK.,Institute of Global Health Innovation, Imperial College London, London, UK
| | - Nick Maynard
- Department of Surgery, Churchill Hospital, Oxford University Hospitals NHS Trust, Oxford, UK
| | - Sheraz R Markar
- Department of Surgery and Cancer, Imperial College London, London, UK. .,Department of Surgery, Churchill Hospital, Oxford University Hospitals NHS Trust, Oxford, UK. .,Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
12
|
Kim HY, Chang W, Lee YJ, Park JH, Cho J, Na HY, Ahn H, Hwang SI, Lee HJ, Kim YH, Lee KH. Adrenal Nodules Detected at Staging CT in Patients with Resectable Gastric Cancers Have a Low Incidence of Malignancy. Radiology 2021; 302:129-137. [PMID: 34665031 DOI: 10.1148/radiol.2021211210] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Background Guidelines recommending additional imaging for adrenal nodules lack relevant epidemiologic evidence. Purpose To measure the prevalence of adrenal nodules detected at staging CT in patients with potentially resectable gastric cancer and the proportion of patients with malignant nodules among them. Materials and Methods This retrospective study included 10 250 consecutive patients (median age, 63 years; interquartile range, 53-71 years; 6884 men) who underwent staging CT and had potentially resectable gastric cancer in a tertiary center (May 2003 to December 2018). All 10 250 CT studies were retrospectively reviewed, and patients with adrenal nodules (or thickening ≥10 mm) were identified to measure the prevalence of adrenal nodules. Among patients with adrenal nodules, the per-patient proportions of malignant nodules, adrenal metastasis from gastric cancer, and additional adrenal examinations were measured. A secondary analysis was performed by using data from the original CT reports. The same metrics that were used in the retrospective review were assessed. Results The prevalence of adrenal nodules was 4.5% (95% CI: 4.1, 4.9; 462 of 10 250). The proportions of malignant nodules and adrenal metastasis from gastric cancer were 0.4% ( 95% CI: 0.1, 1.6; two of 462) and 0% (95% CI: 0.0, 0.8; 0 of 462), respectively. A total of 27% of the patients (95% CI: 23, 31; 123 of 462) underwent additional adrenal examination. According to original CT reports, the prevalence of adrenal nodules and the proportions of malignant nodules, adrenal metastases from gastric cancer, and additional adrenal examination were 2.7% (95% CI: 2.4, 3.0; 272 of 10 250), 0.7% (95% CI: 0.1, 2.6; two of 272), 0% (95% CI: 0.0, 1.4; 0 of 272), and 42.6% (95% CI: 36.7, 48.8; 116 of 272), respectively. Conclusion Although adrenal nodules were detected frequently on staging CT images of patients with otherwise resectable gastric cancer, these nodules were rarely malignant. ©RSNA, 2021 Online supplemental material is available for this article. See also the editorial by Baumgarten in this issue.
Collapse
Affiliation(s)
- Hae Young Kim
- From the Departments of Radiology (H.Y.K., W.C., Y.J.L., J.H.P., J.C., H.A., S.I.H., H.J.L., Y.H.K., K.H.L.) and Pathology (H.Y.N.), Seoul National University Bundang Hospital, 82 Gumi-ro-173-beon-gil, Bundang-gu, Seongnam-si, Gyeonggi-do 13620, Korea; Department of Pathology, Seoul National University College of Medicine, Seoul, Korea (H.Y.N.); Department of Applied Bioengineering, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, Korea (K.H.L.); Department of Radiology, Seoul National University College of Medicine, Seoul National University Bundang Hospital, Gyeonggi-do, Korea (H.J.L., Y.H.K., K.H.L.); and Interdisciplinary Program in Bioengineering, Seoul National University, Seoul, Korea (K.H.L.)
| | - Won Chang
- From the Departments of Radiology (H.Y.K., W.C., Y.J.L., J.H.P., J.C., H.A., S.I.H., H.J.L., Y.H.K., K.H.L.) and Pathology (H.Y.N.), Seoul National University Bundang Hospital, 82 Gumi-ro-173-beon-gil, Bundang-gu, Seongnam-si, Gyeonggi-do 13620, Korea; Department of Pathology, Seoul National University College of Medicine, Seoul, Korea (H.Y.N.); Department of Applied Bioengineering, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, Korea (K.H.L.); Department of Radiology, Seoul National University College of Medicine, Seoul National University Bundang Hospital, Gyeonggi-do, Korea (H.J.L., Y.H.K., K.H.L.); and Interdisciplinary Program in Bioengineering, Seoul National University, Seoul, Korea (K.H.L.)
| | - Yoon Jin Lee
- From the Departments of Radiology (H.Y.K., W.C., Y.J.L., J.H.P., J.C., H.A., S.I.H., H.J.L., Y.H.K., K.H.L.) and Pathology (H.Y.N.), Seoul National University Bundang Hospital, 82 Gumi-ro-173-beon-gil, Bundang-gu, Seongnam-si, Gyeonggi-do 13620, Korea; Department of Pathology, Seoul National University College of Medicine, Seoul, Korea (H.Y.N.); Department of Applied Bioengineering, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, Korea (K.H.L.); Department of Radiology, Seoul National University College of Medicine, Seoul National University Bundang Hospital, Gyeonggi-do, Korea (H.J.L., Y.H.K., K.H.L.); and Interdisciplinary Program in Bioengineering, Seoul National University, Seoul, Korea (K.H.L.)
| | - Ji Hoon Park
- From the Departments of Radiology (H.Y.K., W.C., Y.J.L., J.H.P., J.C., H.A., S.I.H., H.J.L., Y.H.K., K.H.L.) and Pathology (H.Y.N.), Seoul National University Bundang Hospital, 82 Gumi-ro-173-beon-gil, Bundang-gu, Seongnam-si, Gyeonggi-do 13620, Korea; Department of Pathology, Seoul National University College of Medicine, Seoul, Korea (H.Y.N.); Department of Applied Bioengineering, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, Korea (K.H.L.); Department of Radiology, Seoul National University College of Medicine, Seoul National University Bundang Hospital, Gyeonggi-do, Korea (H.J.L., Y.H.K., K.H.L.); and Interdisciplinary Program in Bioengineering, Seoul National University, Seoul, Korea (K.H.L.)
| | - Jungheum Cho
- From the Departments of Radiology (H.Y.K., W.C., Y.J.L., J.H.P., J.C., H.A., S.I.H., H.J.L., Y.H.K., K.H.L.) and Pathology (H.Y.N.), Seoul National University Bundang Hospital, 82 Gumi-ro-173-beon-gil, Bundang-gu, Seongnam-si, Gyeonggi-do 13620, Korea; Department of Pathology, Seoul National University College of Medicine, Seoul, Korea (H.Y.N.); Department of Applied Bioengineering, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, Korea (K.H.L.); Department of Radiology, Seoul National University College of Medicine, Seoul National University Bundang Hospital, Gyeonggi-do, Korea (H.J.L., Y.H.K., K.H.L.); and Interdisciplinary Program in Bioengineering, Seoul National University, Seoul, Korea (K.H.L.)
| | - Hee Young Na
- From the Departments of Radiology (H.Y.K., W.C., Y.J.L., J.H.P., J.C., H.A., S.I.H., H.J.L., Y.H.K., K.H.L.) and Pathology (H.Y.N.), Seoul National University Bundang Hospital, 82 Gumi-ro-173-beon-gil, Bundang-gu, Seongnam-si, Gyeonggi-do 13620, Korea; Department of Pathology, Seoul National University College of Medicine, Seoul, Korea (H.Y.N.); Department of Applied Bioengineering, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, Korea (K.H.L.); Department of Radiology, Seoul National University College of Medicine, Seoul National University Bundang Hospital, Gyeonggi-do, Korea (H.J.L., Y.H.K., K.H.L.); and Interdisciplinary Program in Bioengineering, Seoul National University, Seoul, Korea (K.H.L.)
| | - Hyungwoo Ahn
- From the Departments of Radiology (H.Y.K., W.C., Y.J.L., J.H.P., J.C., H.A., S.I.H., H.J.L., Y.H.K., K.H.L.) and Pathology (H.Y.N.), Seoul National University Bundang Hospital, 82 Gumi-ro-173-beon-gil, Bundang-gu, Seongnam-si, Gyeonggi-do 13620, Korea; Department of Pathology, Seoul National University College of Medicine, Seoul, Korea (H.Y.N.); Department of Applied Bioengineering, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, Korea (K.H.L.); Department of Radiology, Seoul National University College of Medicine, Seoul National University Bundang Hospital, Gyeonggi-do, Korea (H.J.L., Y.H.K., K.H.L.); and Interdisciplinary Program in Bioengineering, Seoul National University, Seoul, Korea (K.H.L.)
| | - Sung Il Hwang
- From the Departments of Radiology (H.Y.K., W.C., Y.J.L., J.H.P., J.C., H.A., S.I.H., H.J.L., Y.H.K., K.H.L.) and Pathology (H.Y.N.), Seoul National University Bundang Hospital, 82 Gumi-ro-173-beon-gil, Bundang-gu, Seongnam-si, Gyeonggi-do 13620, Korea; Department of Pathology, Seoul National University College of Medicine, Seoul, Korea (H.Y.N.); Department of Applied Bioengineering, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, Korea (K.H.L.); Department of Radiology, Seoul National University College of Medicine, Seoul National University Bundang Hospital, Gyeonggi-do, Korea (H.J.L., Y.H.K., K.H.L.); and Interdisciplinary Program in Bioengineering, Seoul National University, Seoul, Korea (K.H.L.)
| | - Hak Jong Lee
- From the Departments of Radiology (H.Y.K., W.C., Y.J.L., J.H.P., J.C., H.A., S.I.H., H.J.L., Y.H.K., K.H.L.) and Pathology (H.Y.N.), Seoul National University Bundang Hospital, 82 Gumi-ro-173-beon-gil, Bundang-gu, Seongnam-si, Gyeonggi-do 13620, Korea; Department of Pathology, Seoul National University College of Medicine, Seoul, Korea (H.Y.N.); Department of Applied Bioengineering, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, Korea (K.H.L.); Department of Radiology, Seoul National University College of Medicine, Seoul National University Bundang Hospital, Gyeonggi-do, Korea (H.J.L., Y.H.K., K.H.L.); and Interdisciplinary Program in Bioengineering, Seoul National University, Seoul, Korea (K.H.L.)
| | - Young Hoon Kim
- From the Departments of Radiology (H.Y.K., W.C., Y.J.L., J.H.P., J.C., H.A., S.I.H., H.J.L., Y.H.K., K.H.L.) and Pathology (H.Y.N.), Seoul National University Bundang Hospital, 82 Gumi-ro-173-beon-gil, Bundang-gu, Seongnam-si, Gyeonggi-do 13620, Korea; Department of Pathology, Seoul National University College of Medicine, Seoul, Korea (H.Y.N.); Department of Applied Bioengineering, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, Korea (K.H.L.); Department of Radiology, Seoul National University College of Medicine, Seoul National University Bundang Hospital, Gyeonggi-do, Korea (H.J.L., Y.H.K., K.H.L.); and Interdisciplinary Program in Bioengineering, Seoul National University, Seoul, Korea (K.H.L.)
| | - Kyoung Ho Lee
- From the Departments of Radiology (H.Y.K., W.C., Y.J.L., J.H.P., J.C., H.A., S.I.H., H.J.L., Y.H.K., K.H.L.) and Pathology (H.Y.N.), Seoul National University Bundang Hospital, 82 Gumi-ro-173-beon-gil, Bundang-gu, Seongnam-si, Gyeonggi-do 13620, Korea; Department of Pathology, Seoul National University College of Medicine, Seoul, Korea (H.Y.N.); Department of Applied Bioengineering, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, Korea (K.H.L.); Department of Radiology, Seoul National University College of Medicine, Seoul National University Bundang Hospital, Gyeonggi-do, Korea (H.J.L., Y.H.K., K.H.L.); and Interdisciplinary Program in Bioengineering, Seoul National University, Seoul, Korea (K.H.L.)
| |
Collapse
|
13
|
Shaker R, Wilke C, Ober C, Lawrence J. Machine learning model development for quantitative analysis of CT heterogeneity in canine hepatic masses may predict histologic malignancy. Vet Radiol Ultrasound 2021; 62:711-719. [PMID: 34448312 DOI: 10.1111/vru.13012] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 06/28/2021] [Accepted: 06/29/2021] [Indexed: 01/24/2023] Open
Abstract
Tumor heterogeneity is a well-established marker of biologically aggressive neoplastic processes and is associated with local recurrence and distant metastasis. Quantitative analysis of CT textural features is an indirect measure of tumor heterogeneity and therefore may help predict malignant disease. The purpose of this retrospective, secondary analysis study was to quantitatively evaluate CT heterogeneity in dogs with histologically confirmed liver masses to build a predictive model for malignancy. Forty dogs with liver tumors and corresponding histopathologic evaluation from a previous prospective study were included. Triphasic image acquisition was standardized across dogs and whole liver and liver mass were contoured on each precontrast and delayed postcontrast dataset. First-order and second-order indices were extracted from contoured regions. Univariate analysis identified potentially significant indices that were subsequently used for top-down model construction. Multiple quadratic discriminatory models were constructed and tested, including individual models using both postcontrast and precontrast whole liver or liver mass volumes. The best performing model utilized the CT features voxel volume and uniformity from postcontrast mass contours; this model had an accuracy of 0.90, sensitivity of 0.67, specificity of 1.0, positive predictive value of 1.0, negative predictive value of 0.88, and precision of 1.0. Heterogeneity indices extracted from delayed postcontrast CT hepatic mass contours were more informative about tumor type compared to indices from whole liver contours, or from precontrast hepatic mass and whole liver contours. Results demonstrate that CT radiomic feature analysis may hold clinical utility as a noninvasive method of predicting hepatic malignancy and may influence diagnostic or therapeutic approaches.
Collapse
Affiliation(s)
- Rami Shaker
- Institute for Health Informatics, University of Minnesota, Minneapolis, Minnesota, USA.,Medical School, University of Minnesota, Minneapolis, Minnesota, USA
| | - Christopher Wilke
- Department of Radiation Oncology, Medical School, University of Minnesota, Minneapolis, Minnesota, USA.,Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota, USA
| | - Christopher Ober
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, University of Minnesota, St Paul, Minnesota, USA
| | - Jessica Lawrence
- Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota, USA.,Department of Veterinary Clinical Sciences, College of Veterinary Medicine, University of Minnesota, St Paul, Minnesota, USA
| |
Collapse
|
14
|
Qin Y, Deng Y, Jiang H, Hu N, Song B. Artificial Intelligence in the Imaging of Gastric Cancer: Current Applications and Future Direction. Front Oncol 2021; 11:631686. [PMID: 34367946 PMCID: PMC8335156 DOI: 10.3389/fonc.2021.631686] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Accepted: 07/07/2021] [Indexed: 02/05/2023] Open
Abstract
Gastric cancer (GC) is one of the most common cancers and one of the leading causes of cancer-related death worldwide. Precise diagnosis and evaluation of GC, especially using noninvasive methods, are fundamental to optimal therapeutic decision-making. Despite the recent rapid advancements in technology, pretreatment diagnostic accuracy varies between modalities, and correlations between imaging and histological features are far from perfect. Artificial intelligence (AI) techniques, particularly hand-crafted radiomics and deep learning, have offered hope in addressing these issues. AI has been used widely in GC research, because of its ability to convert medical images into minable data and to detect invisible textures. In this article, we systematically reviewed the methodological processes (data acquisition, lesion segmentation, feature extraction, feature selection, and model construction) involved in AI. We also summarized the current clinical applications of AI in GC research, which include characterization, differential diagnosis, treatment response monitoring, and prognosis prediction. Challenges and opportunities in AI-based GC research are highlighted for consideration in future studies.
Collapse
Affiliation(s)
- Yun Qin
- Department of Radiology, West China Hospital, Sichuan University, Chengdu, China
| | - Yiqi Deng
- Department of Laboratory Medicine, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Hanyu Jiang
- Department of Radiology, West China Hospital, Sichuan University, Chengdu, China
| | - Na Hu
- Department of Radiology, West China Hospital, Sichuan University, Chengdu, China
| | - Bin Song
- Department of Radiology, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
15
|
Mazzei MA, Di Giacomo L, Bagnacci G, Nardone V, Gentili F, Lucii G, Tini P, Marrelli D, Morgagni P, Mura G, Baiocchi GL, Pittiani F, Volterrani L, Roviello F. Delta-radiomics and response to neoadjuvant treatment in locally advanced gastric cancer-a multicenter study of GIRCG (Italian Research Group for Gastric Cancer). Quant Imaging Med Surg 2021; 11:2376-2387. [PMID: 34079708 DOI: 10.21037/qims-20-683] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Background To predict response to neoadjuvant chemotherapy (NAC) of gastric cancer (GC), prior to surgery, would be pivotal to customize patient treatment. The aim of this study is to investigate the reliability of computed tomography (CT) texture analysis (TA) in predicting the histo-pathological response to NAC in patients with resectable locally advanced gastric cancer (AGC). Methods Seventy (40 male, mean age 63.3 years) patients with resectable locally AGC, treated with NAC and radical surgery, were included in this retrospective study from 5 centers of the Italian Research Group for Gastric Cancer (GIRCG). Population was divided into two groups: 29 patients from one center (internal cohort for model development and internal validation) and 41 from other four centers (external cohort for independent external validation). Gross tumor volume (GTV) was segmented on each pre- and post-NAC multidetector CT (MDCT) image by using a dedicated software (RayStation), and 14 TA parameters were then extrapolated. Correlation between TA parameters and complete pathological response (tumor regression grade, TRG1), was initially investigated for the internal cohort. The univariate significant variables were tested on the external cohort and multivariate logistic analysis was performed. Results In multivariate logistic regression the only significant TA variable was delta gray-level co-occurrence matrix (GLCM) contrast (P=0.001, Nagelkerke R2: 0.546 for the internal cohort and P=0.014, Nagelkerke R2: 0.435 for the external cohort). Receiver operating characteristic (ROC) curves, generated from the logistic regression of all the patients, showed an area under the curve (AUC) of 0.763. Conclusions Post-NAC GLCM contrast and dissimilarity and delta GLCM contrast TA parameters seem to be reliable for identifying patients with locally AGC responder to NAC.
Collapse
Affiliation(s)
- Maria Antonietta Mazzei
- Department of Medical, Surgical and Neuro Sciences, University of Siena and Department of Radiological Sciences, Unit of Diagnostic Imaging, Azienda Ospedaliera Universitaria Senese, Siena, Italy
| | - Letizia Di Giacomo
- Department of Medical, Surgical and Neuro Sciences, University of Siena and Department of Radiological Sciences, Unit of Diagnostic Imaging, Azienda Ospedaliera Universitaria Senese, Siena, Italy
| | - Giulio Bagnacci
- Department of Medical, Surgical and Neuro Sciences, University of Siena and Department of Radiological Sciences, Unit of Diagnostic Imaging, Azienda Ospedaliera Universitaria Senese, Siena, Italy
| | | | - Francesco Gentili
- Section of Radiology, Unit of Surgical Sciences, University of Parma, Parma, Italy
| | - Gabriele Lucii
- Department of Medical, Surgical and Neuro Sciences, University of Siena and Department of Radiological Sciences, Unit of Diagnostic Imaging, Azienda Ospedaliera Universitaria Senese, Siena, Italy
| | - Paolo Tini
- Unit of Radiation Oncology, Azienda Ospedaliera Universitaria Senese, Siena, Italy
| | - Daniele Marrelli
- Department of Medical, Surgical and Neuro Sciences, Unit of Surgical Oncology, University of Siena, Azienda Ospedaliera Universitaria Senese, Siena, Italy
| | - Paolo Morgagni
- Department of General Surgery, Morgagni-Pierantoni Hospital, Forlì, Italy
| | - Gianni Mura
- Department of Surgery, San Donato Hospital, Arezzo, Italy
| | - Gian Luca Baiocchi
- Department of Clinical and Experimental Studies, Surgical Clinic, University of Brescia, Brescia, Italy
| | - Frida Pittiani
- Department of Radiology, ASST Spedali Civili Brescia, Brescia, Italy
| | - Luca Volterrani
- Department of Medical, Surgical and Neuro Sciences, University of Siena and Department of Radiological Sciences, Unit of Diagnostic Imaging, Azienda Ospedaliera Universitaria Senese, Siena, Italy
| | - Franco Roviello
- Department of Medical, Surgical and Neuro Sciences, Unit of Surgical Oncology, University of Siena, Azienda Ospedaliera Universitaria Senese, Siena, Italy
| |
Collapse
|
16
|
Foley KG, Pearson B, Riddell Z, Taylor SA. Opportunities in cancer imaging: a review of oesophageal, gastric and colorectal malignancies. Clin Radiol 2021; 76:748-762. [PMID: 33579518 DOI: 10.1016/j.crad.2021.01.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Accepted: 01/13/2021] [Indexed: 02/07/2023]
Abstract
The incidence of gastrointestinal (GI) malignancy is increasing worldwide. In particular, there is a concerning rise in incidence of GI cancer in younger adults. Direct endoscopic visualisation of luminal tumour sites requires invasive procedures, which are associated with certain risks, but remain necessary because of limitations in current imaging techniques and the continuing need to obtain tissue for diagnosis and genetic analysis; however, management of GI cancer is increasingly reliant on non-invasive, radiological imaging to diagnose, stage, and treat these malignancies. Oesophageal, gastric, and colorectal malignancies require specialist investigation and treatment due to the complex nature of the anatomy, biology, and subsequent treatment strategies. As cancer imaging techniques develop, many opportunities to improve tumour detection, diagnostic accuracy and treatment monitoring present themselves. This review article aims to report current imaging practice, advances in various radiological modalities in relation to GI luminal tumour sites and describes opportunities for GI radiologists to improve patient outcomes.
Collapse
Affiliation(s)
- K G Foley
- Department of Clinical Radiology, Royal Glamorgan Hospital, Llantrisant, UK.
| | - B Pearson
- National Imaging Academy Wales (NIAW), Pencoed, UK
| | - Z Riddell
- National Imaging Academy Wales (NIAW), Pencoed, UK
| | - S A Taylor
- Centre for Medical Imaging, UCL, London, UK
| |
Collapse
|
17
|
Prospective evaluation of metabolic intratumoral heterogeneity in patients with advanced gastric cancer receiving palliative chemotherapy. Sci Rep 2021; 11:296. [PMID: 33436659 PMCID: PMC7804009 DOI: 10.1038/s41598-020-78963-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Accepted: 12/01/2020] [Indexed: 12/23/2022] Open
Abstract
Although metabolic intratumoral heterogeneity (ITH) gives important value on treatment responses and prognoses, its association with treatment outcomes have not been reported in gastric cancer (GC). We aimed to evaluate temporal changes in metabolic ITH and the associations with treatment responses, progression-free survival (PFS), and overall survival (OS) in advanced GC patients. Eighty-five patients with unresectable, locally advanced, or metastatic GC were prospectively enrolled before the first-line palliative chemotherapy and underwent [18F]FDG PET at baseline (TP1) and the first response follow-up evaluation (TP2). Standardized uptake values (SUVs), volumetric parameters, and textural features were evaluated in primary gastric tumor at TP1 and TP2. Of 85 patients, 44 had partial response, 33 had stable disease, and 8 progressed. From TP1 to TP2, metabolic ITH was significantly reduced (P < 0.01), and the degree of the decrease was greater in responders than in non-responders (P < 0.01). Using multiple Cox regression analyses, a low SUVmax at TP2, a high kurtosis at TP2 and larger decreases in the coefficient of variance were associated with better PFS. A low SUVmax at TP2, larger decreases in the metabolic tumor volume and larger decreased in the energy were associated with better OS. Age older than 60 years and responders also showed better OS. An early reduction in metabolic ITH is useful to predict treatment outcomes in advanced GC patients.
Collapse
|
18
|
Contrast-Enhanced CT-based Textural Parameters as Potential Prognostic Factors of Survival for Colorectal Cancer Patients Receiving Targeted Therapy. Mol Imaging Biol 2020; 23:427-435. [PMID: 33108800 DOI: 10.1007/s11307-020-01552-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Revised: 09/07/2020] [Accepted: 10/05/2020] [Indexed: 02/08/2023]
Abstract
PURPOSE This study was designed to estimate the clinical significance of the contrast-enhanced computed tomography (CT) textural features for prediction of survival in colorectal cancer (CRC) patients receiving targeted therapy (bevacizumab and cetuximab). PROCEDURES The LifeX software was used to extract the textural parameters of the tumor lesions in the contrast-enhanced CT. We used the least absolute shrinkage and selection operator (LASSO) Cox regression and random forest method to screen the non-redundant radiomic features and constructed the CT imaging score. Univariate and multivariate analyses through the Cox proportional hazards model were performed to assess the prognostic clinical factor. Based on the result of multivariate analysis and CT imaging score, combined nomogram model was constructed to predict the overall survival (OS) of patients. Decision curves analysis was employed to evaluate the performance of the combined model and clinical model. RESULTS After comparative analysis of the area under curve of the receiver operating characteristic (ROC) curve, we chose the result of random forest model as CT imaging score. Considering the clinical practice and the result of analysis, age, surgery, and lactate dehydrogenase (LDH) level have been introduced into clinical model. Based on the result of analysis and the CT imaging score, we constructed the nomogram combined model. C-index and calibration curve verified the goodness of fit and discrimination of the combined model. Decision curve analysis (DCA) demonstrated that the combined model showed the better net benefit for a 3-year OS than clinical model. CONCLUSIONS In conclusion, the study provides preliminary evidences that several radiomic parameters of tumor lesions derived from CT images were prognostic factors and predictive markers for CRC patients who are candidates for targeted therapy (bevacizumab and cetuximab).
Collapse
|
19
|
Zhang WT, Zhu C, Wang X, Wang SJ, Chen XD, Chen XL, Shen X. Preoperative Splenic Density for the Prediction of Survival and Adjuvant Chemotherapy Benefits in Gastric Cancer. J Cancer 2020; 11:6133-6139. [PMID: 32922553 PMCID: PMC7477414 DOI: 10.7150/jca.47559] [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: 04/29/2020] [Accepted: 08/13/2020] [Indexed: 11/18/2022] Open
Abstract
Background: We aimed to determine whether splenic features change during tumor progression by evaluating the clinicopathological characteristics relevant to splenic density in patients with gastric cancer (GC) and identify a new predictive indicator of prognosis and chemotherapy benefits. Methods: In the present analysis, 408 patients who underwent gastrectomy were included. Density was expressed in mean spleen Hounsfield units on computed tomography. Other clinical characteristics and detailed follow-up data were collected. The cutoff splenic density was 47.8 by the Xtile software. The R software was used for characteristic differential analysis in patients with different splenic densities. The Cox proportional hazards model and forest plot were used for prognosis and chemotherapy benefit analyses. Results: Patients with low splenic density had significantly worse 3-year disease-free survival (DFS) and overall survival (OS) rates (high vs low splenic density: DFS, 63.4% vs 44.6%, p<0.001; OS, 69.8% vs 52.4%, p<0.001). Splenic density showed strong negative correlations with age, number of metastasized lymph nodes, tumor size, and depth of tumor invasion. The benefits of adjuvant chemotherapy were better in the low splenic density group (hazard ratio of OS, 0.546; p=0.001) than in the high-density group (hazard ratio of OS, 0.701; p=0.106). Conclusions: Patients with low splenic density tended to have more advanced tumors and poor prognosis, but better chemotherapy benefits. Splenic density can be regarded as a new indicator of chemotherapy benefits and increase the accuracy of preoperative staging evaluation. Moreover, preoperative evaluation of splenic density may help establish individualized treatment strategies.
Collapse
Affiliation(s)
- Wei-Teng Zhang
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Ce Zhu
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Xiang Wang
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Su-Jun Wang
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Wenzhou Medical University, ShangCai Village, Wenzhou, Zhejiang Province, China
| | - Xiao-Dong Chen
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Xiao-Lei Chen
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Wenzhou Medical University, ShangCai Village, Wenzhou, Zhejiang Province, China
| | - Xian Shen
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| |
Collapse
|
20
|
Feng P, Wang ZD, Fan W, Liu H, Pan JJ. Diagnostic advances of artificial intelligence and radiomics in gastroenterology. Artif Intell Gastroenterol 2020; 1:37-50. [DOI: 10.35712/aig.v1.i2.37] [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: 05/27/2020] [Revised: 08/22/2020] [Accepted: 08/27/2020] [Indexed: 02/06/2023] Open
Abstract
Traditional medical imaging, including ultrasound, computed tomography, magnetic resonance imaging, or positron emission tomography, remains widely used diagnostic modalities for gastrointestinal diseases at present. These modalities are used to assess changes in morphology, attenuation, signal intensity, and enhancement characteristics. Gastrointestinal tumors, especially malignant tumors, are commonly seen in clinical practice with an increasing number of deaths each year. Because the imaging manifestations of different diseases usually overlap, accurate early diagnosis of tumor lesions, noninvasive and effective evaluation of tumor staging, and prediction of prognosis remain challenging. Fortunately, traditional medical images contain a great deal of important information that cannot be recognized by human eyes but can be extracted by artificial intelligence (AI) technology, which can quantitatively assess the heterogeneity of lesions and provide valuable information, including therapeutic effects and patient prognosis. With the development of computer technology, the combination of medical imaging and AI technology is considered to represent a promising field in medical image analysis. This new emerging field is called “radiomics”, which makes big data mining and extraction from medical imagery possible and can help clinicians make effective decisions and develop personalized treatment plans. Recently, AI and radiomics have been gradually applied to lesion detection, qualitative and quantitative diagnosis, histopathological grading and staging of tumors, therapeutic efficacy assessment, and prognosis evaluation. In this minireview, we briefly introduce the basic principles and technology of radiomics. Then, we review the research and application of AI and radiomics in gastrointestinal diseases, especially diagnostic advancements of radiomics in the differential diagnosis, treatment option, assessment of therapeutic efficacy, and prognosis evaluation of esophageal, gastric, hepatic, pancreatic, and colorectal diseases.
Collapse
Affiliation(s)
- Pei Feng
- Department of Radiology, PLA Rocket Force Characteristic Medical Center, Beijing 100088, China
| | - Zhen-Dong Wang
- Department of Ultrasound, Beijing Sihui Hospital of Traditional Chinese Medicine, Beijing 100022, China
| | - Wei Fan
- Department of Radiology, PLA Rocket Force Characteristic Medical Center, Beijing 100088, China
| | - Heng Liu
- Department of Radiology, PLA Rocket Force Characteristic Medical Center, Beijing 100088, China
| | - Jing-Jing Pan
- Department of Radiology, PLA Rocket Force Characteristic Medical Center, Beijing 100088, China
| |
Collapse
|
21
|
Wang S, Feng C, Dong D, Li H, Zhou J, Ye Y, Liu Z, Tian J, Wang Y. Preoperative computed tomography-guided disease-free survival prediction in gastric cancer: a multicenter radiomics study. Med Phys 2020; 47:4862-4871. [PMID: 32592224 DOI: 10.1002/mp.14350] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 05/24/2020] [Accepted: 06/17/2020] [Indexed: 12/12/2022] Open
Abstract
PURPOSE Preoperative and noninvasive prognosis evaluation remains challenging for gastric cancer. Novel preoperative prognostic biomarkers should be investigated. This study aimed to develop multidetector-row computed tomography (MDCT)-guided prognostic models to direct follow-up strategy and improve prognosis. METHODS A retrospective dataset of 353 gastric cancer patients were enrolled from two centers and allocated to three cohorts: training cohort (n = 166), internal validation cohort (n = 83), and external validation cohort (n = 104). Quantitative radiomic features were extracted from MDCT images. The least absolute shrinkage and selection operator penalized Cox regression was adopted to construct a radiomic signature. A radiomic nomogram was established by integrating the radiomic signature and significant clinical risk factors. We also built a preoperative tumor-node-metastasis staging model for comparison. All models were evaluated considering the abilities of risk stratification, discrimination, calibration, and clinical use. RESULTS In the two validation cohorts, the established four-feature radiomic signature showed robust risk stratification power (P = 0.0260 and 0.0003, log-rank test). The radiomic nomogram incorporated radiomic signature, extramural vessel invasion, clinical T stage, and clinical N stage, outperforming all the other models (concordance index = 0.720 and 0.727) with good calibration and decision benefits. Also, the 2-yr disease-free survival (DFS) prediction was most effective (time-dependent area under curve = 0.771 and 0.765). Moreover, subgroup analysis indicated that the radiomic signature was more sensitive in risk stratifying patients with advanced clinical T/N stage. CONCLUSIONS The proposed MDCT-guided radiomic signature was verified as a prognostic factor for gastric cancer. The radiomic nomogram was a noninvasive auxiliary model for preoperative individualized DFS prediction, holding potential in promoting treatment strategy and clinical prognosis.
Collapse
Affiliation(s)
- Siwen Wang
- CAS Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing, 100190, China.,School of Artificial Intelligence, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Caizhen Feng
- Department of Radiology, Peking University People's Hospital, Beijing, 100044, China
| | - Di Dong
- CAS Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing, 100190, China.,School of Artificial Intelligence, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Hailin Li
- CAS Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing, 100190, China.,School of Artificial Intelligence, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Jing Zhou
- Department of Gastrointestinal Surgery, Peking University People's Hospital, Beijing, 100044, China
| | - Yingjiang Ye
- Department of Gastrointestinal Surgery, Peking University People's Hospital, Beijing, 100044, China
| | - Zaiyi Liu
- Department of Radiology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080, China
| | - Jie Tian
- CAS Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing, 100190, China.,Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, Beihang University, Beijing, 100191, China
| | - Yi Wang
- Department of Radiology, Peking University People's Hospital, Beijing, 100044, China
| |
Collapse
|
22
|
Sun KY, Hu HT, Chen SL, Ye JN, Li GH, Chen LD, Peng JJ, Feng ST, Yuan YJ, Hou X, Wu H, Li X, Wu TF, Wang W, Xu JB. CT-based radiomics scores predict response to neoadjuvant chemotherapy and survival in patients with gastric cancer. BMC Cancer 2020; 20:468. [PMID: 32450841 PMCID: PMC7249312 DOI: 10.1186/s12885-020-06970-7] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Accepted: 05/18/2020] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Neoadjuvant chemotherapy is a promising treatment option for potential resectable gastric cancer, but patients' responses vary. We aimed to develop and validate a radiomics score (rad_score) to predict treatment response to neoadjuvant chemotherapy and to investigate its efficacy in survival stratification. METHODS A total of 106 patients with neoadjuvant chemotherapy before gastrectomy were included (training cohort: n = 74; validation cohort: n = 32). Radiomics features were extracted from the pre-treatment portal venous-phase CT. After feature reduction, a rad_score was established by Randomised Tree algorithm. A rad_clinical_score was constructed by integrating the rad_score with clinical variables, so was a clinical score by clinical variables only. The three scores were validated regarding their discrimination and clinical usefulness. The patients were stratified into two groups according to the score thresholds (updated with post-operative clinical variables), and their survivals were compared. RESULTS In the validation cohort, the rad_score demonstrated a good predicting performance in treatment response to the neoadjuvant chemotherapy (AUC [95% CI] =0.82 [0.67, 0.98]), which was better than the clinical score (based on pre-operative clinical variables) without significant difference (0.62 [0.42, 0.83], P = 0.09). The rad_clinical_score could not further improve the performance of the rad_score (0.70 [0.51, 0.88], P = 0.16). Based on the thresholds of these scores, the high-score groups all achieved better survivals than the low-score groups in the whole cohort (all P < 0.001). CONCLUSION The rad_score that we developed was effective in predicting treatment response to neoadjuvant chemotherapy and in stratifying patients with gastric cancer into different survival groups. Our proposed strategy is useful for individualised treatment planning.
Collapse
Affiliation(s)
- Kai-Yu Sun
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Sun Yat-Sen University, 58 Zhongshan Road 2, Guangzhou, 510080, People's Republic of China
| | - Hang-Tong Hu
- Department of Medical Ultrasonics, Institute of Diagnostic and Interventional Ultrasound, The First Affiliated Hospital of Sun Yat-Sen University, 58 Zhongshan Road 2, Guangzhou, 510080, People's Republic of China
| | - Shu-Ling Chen
- Department of Medical Ultrasonics, Institute of Diagnostic and Interventional Ultrasound, The First Affiliated Hospital of Sun Yat-Sen University, 58 Zhongshan Road 2, Guangzhou, 510080, People's Republic of China
| | - Jin-Ning Ye
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Sun Yat-Sen University, 58 Zhongshan Road 2, Guangzhou, 510080, People's Republic of China
| | - Guang-Hua Li
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Sun Yat-Sen University, 58 Zhongshan Road 2, Guangzhou, 510080, People's Republic of China
| | - Li-Da Chen
- Department of Medical Ultrasonics, Institute of Diagnostic and Interventional Ultrasound, The First Affiliated Hospital of Sun Yat-Sen University, 58 Zhongshan Road 2, Guangzhou, 510080, People's Republic of China
| | - Jian-Jun Peng
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Sun Yat-Sen University, 58 Zhongshan Road 2, Guangzhou, 510080, People's Republic of China
| | - Shi-Ting Feng
- Department of Radiology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, China
| | - Yu-Jie Yuan
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Sun Yat-Sen University, 58 Zhongshan Road 2, Guangzhou, 510080, People's Republic of China
| | - Xun Hou
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Sun Yat-Sen University, 58 Zhongshan Road 2, Guangzhou, 510080, People's Republic of China
| | - Hui Wu
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Sun Yat-Sen University, 58 Zhongshan Road 2, Guangzhou, 510080, People's Republic of China
| | - Xin Li
- Research Center of GE Healthcare, Shanghai, 200000, China
| | - Ting-Fan Wu
- Research Center of GE Healthcare, Shanghai, 200000, China
| | - Wei Wang
- Department of Medical Ultrasonics, Institute of Diagnostic and Interventional Ultrasound, The First Affiliated Hospital of Sun Yat-Sen University, 58 Zhongshan Road 2, Guangzhou, 510080, People's Republic of China.
| | - Jian-Bo Xu
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Sun Yat-Sen University, 58 Zhongshan Road 2, Guangzhou, 510080, People's Republic of China.
| |
Collapse
|
23
|
Ieni A, Cardia R, Pizzimenti C, Zeppa P, Tuccari G. HER2 Heterogeneity in Personalized Therapy of Gastro-Oesophageal Malignancies: An Overview by Different Methodologies. J Pers Med 2020; 10:jpm10010010. [PMID: 32098203 PMCID: PMC7151629 DOI: 10.3390/jpm10010010] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Revised: 02/06/2020] [Accepted: 02/12/2020] [Indexed: 12/15/2022] Open
Abstract
Human epidermal growth factor receptor-2 (HER2)-expression gastro-oesophageal adenocarcinomas (GEA) gained interest as an important target for therapy with trastuzumab. In the current review, we focused the current knowledge on HER2 status in dysplastic and neoplastic gastric conditions, analyzing the methodological procedures to identify HER2 expression/amplification, as well as the proposed scoring recommendations. One of the most relevant questions to evaluate the useful impact of HER2 status on therapeutic choice in GEAs is represented by the significant heterogeneity of HER2 protein and gene expression that may affect the targeted treatment selection. Future development of biotechnology will continue to evolve in order to offer more powerful detection systems for the assessment of HER2 status. Finally, liquid biopsy as well as mutation/amplification of several additional genes may furnish an early detection of secondary HER2 resistance mechanisms in GEAs with a better monitoring of the treatment response.
Collapse
Affiliation(s)
- Antonio Ieni
- Department of Human Pathology in Adult and Developmental Age “Gaetano Barresi”, Section of Pathology, University of Messina, 98125 Messina, Italy; (R.C.); (C.P.); (G.T.)
- Correspondence: ; Tel.: +39-90-221-2536; Fax: +39-90-292-8150
| | - Roberta Cardia
- Department of Human Pathology in Adult and Developmental Age “Gaetano Barresi”, Section of Pathology, University of Messina, 98125 Messina, Italy; (R.C.); (C.P.); (G.T.)
| | - Cristina Pizzimenti
- Department of Human Pathology in Adult and Developmental Age “Gaetano Barresi”, Section of Pathology, University of Messina, 98125 Messina, Italy; (R.C.); (C.P.); (G.T.)
| | - Pio Zeppa
- Department of Medicine and Surgery, University of Salerno, 84131 Salerno, Italy;
| | - Giovanni Tuccari
- Department of Human Pathology in Adult and Developmental Age “Gaetano Barresi”, Section of Pathology, University of Messina, 98125 Messina, Italy; (R.C.); (C.P.); (G.T.)
| |
Collapse
|
24
|
Li Y, Cheng Z, Gevaert O, He L, Huang Y, Chen X, Huang X, Wu X, Zhang W, Dong M, Huang J, Huang Y, Xia T, Liang C, Liu Z. A CT-based radiomics nomogram for prediction of human epidermal growth factor receptor 2 status in patients with gastric cancer. Chin J Cancer Res 2020; 32:62-71. [PMID: 32194306 PMCID: PMC7072015 DOI: 10.21147/j.issn.1000-9604.2020.01.08] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2019] [Accepted: 12/03/2019] [Indexed: 02/05/2023] Open
Abstract
OBJECTIVE To develop and validate a computed tomography (CT)-based radiomics nomogram for predicting human epidermal growth factor receptor 2 (HER2) status in patients with gastric cancer. METHODS This retrospective study included 134 patients with gastric cancer (HER2-negative: n=87; HER2-positive: n=47) from April 2013 to March 2018, who were then randomly divided into training (n=94) and validation (n=40) cohorts. Radiomics features were obtained from the CT images showing gastric cancer. Least absolute shrinkage and selection operator (LASSO) regression analysis was utilized for building the radiomics signature. A multivariable logistic regression method was applied to develop a prediction model incorporating the radiomics signature and independent clinicopathologic risk predictors, which were then visualized as a radiomics nomogram. The predictive performance of the nomogram was assessed in the training and validation cohorts. RESULTS The radiomics signature was significantly associated with HER2 status in both training (P<0.001) and validation (P=0.023) cohorts. The prediction model that incorporated the radiomics signature and carcinoembryonic antigen (CEA) level demonstrated good discriminative performance for HER2 status prediction, with an area under the curve (AUC) of 0.799 [95% confidence interval (95% CI): 0.704-0.894] in the training cohort and 0.771 (95% CI: 0.607-0.934) in the validation cohort. The calibration curve of the radiomics nomogram also showed good calibration. Decision curve analysis showed that the radiomics nomogram was useful. CONCLUSIONS We built and validated a radiomics nomogram with good performance for HER2 status prediction in gastric cancer. This radiomics nomogram could serve as a non-invasive tool to predict HER2 status and guide clinical treatment.
Collapse
Affiliation(s)
- Yexing Li
- Department of Radiology, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, China
- Graduate College, Shantou University Medical College, Shantou 515041, China
| | - Zixuan Cheng
- Department of Radiology, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, China
- School of Medicine, South China University of Technology, Guangzhou 510006, China
| | - Olivier Gevaert
- Stanford Center for Biomedical Informatics Research, Department of Medicine, and Department of Biomedical Data Science, Stanford University, California 94305, USA
| | - Lan He
- Department of Radiology, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, China
| | - Yanqi Huang
- Department of Radiology, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, China
| | - Xin Chen
- Department of Radiology, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, China
- Graduate College, Southern Medical University, Guangzhou 510515, China
- Department of Radiology, Guangzhou First People’s Hospital, Guangzhou 510180, China
| | - Xiaomei Huang
- Department of Radiology, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, China
- Graduate College, Southern Medical University, Guangzhou 510515, China
| | - Xiaomei Wu
- Department of Radiology, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, China
- School of Medicine, South China University of Technology, Guangzhou 510006, China
| | - Wen Zhang
- Department of Radiology, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, China
- Graduate College, Southern Medical University, Guangzhou 510515, China
- Department of Radiology, Zhuhai Hospital of Traditional Chinese and Western Medicine, Zhuhai 519000, China
| | - Mengyi Dong
- Department of Radiology, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, China
- Graduate College, Southern Medical University, Guangzhou 510515, China
| | - Jia Huang
- Department of Radiology, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, China
- Graduate College, Shantou University Medical College, Shantou 515041, China
| | - Yucun Huang
- Department of Radiology, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, China
- Graduate College, Southern Medical University, Guangzhou 510515, China
- Department of Radiology, The Fifth People’s Hospital of Zhuhai, Zhuhai 519055, China
| | - Ting Xia
- Department of Radiology, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, China
- School of Medicine, South China University of Technology, Guangzhou 510006, China
| | - Changhong Liang
- Department of Radiology, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, China
- Graduate College, Shantou University Medical College, Shantou 515041, China
- Changhong Liang, PhD. Department of Radiology, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, No.106 Zhongshan Er Road, Guangzhou 510080, China.
| | - Zaiyi Liu
- Department of Radiology, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, China
- Graduate College, Shantou University Medical College, Shantou 515041, China
- Zaiyi Liu, PhD. Department of Radiology, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, No.106 Zhongshan Er Road, Guangzhou 510080, China.
| |
Collapse
|
25
|
Abstract
Objective: To review the application of radiomics in gastric cancer and its challenges as well as future prospects. Data sources: A research for relevant studies were performed in PubMed with the terms of “radiomics,” “texture analysis,” and “gastric cancer.” The search was updated until February 28th, 2019. Study selection: All original articles regarding the investigation of texture analysis or radiomics in gastric cancer were retrieved. Only papers written in English were included. Results: A total of 17 original articles were selected in final. It is shown that radiomics has yielded moderate to excellent performance in a spectrum of respects including differential diagnosis, assessment of histological differential degree, evaluation of tumor stage, prediction of response to therapy, and prognosis in gastric cancer. Yet, a number of challenges are facing both radiomics itself and its application in gastric cancer. Conclusions: Radiomics holds great potential in facilitating decision-making in gastric cancer. With the standardization of work-flow and advancement of machine learning methods, radiomics is expected to make great breakthroughs in precision medicine of gastric cancer.
Collapse
|
26
|
de Maar JS, Sofias AM, Porta Siegel T, Vreeken RJ, Moonen C, Bos C, Deckers R. Spatial heterogeneity of nanomedicine investigated by multiscale imaging of the drug, the nanoparticle and the tumour environment. Am J Cancer Res 2020; 10:1884-1909. [PMID: 32042343 PMCID: PMC6993242 DOI: 10.7150/thno.38625] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Accepted: 11/13/2019] [Indexed: 02/07/2023] Open
Abstract
Genetic and phenotypic tumour heterogeneity is an important cause of therapy resistance. Moreover, non-uniform spatial drug distribution in cancer treatment may cause pseudo-resistance, meaning that a treatment is ineffective because the drug does not reach its target at sufficient concentrations. Together with tumour heterogeneity, non-uniform drug distribution causes “therapy heterogeneity”: a spatially heterogeneous treatment effect. Spatial heterogeneity in drug distribution occurs on all scales ranging from interpatient differences to intratumour differences on tissue or cellular scale. Nanomedicine aims to improve the balance between efficacy and safety of drugs by targeting drug-loaded nanoparticles specifically to tumours. Spatial heterogeneity in nanoparticle and payload distribution could be an important factor that limits their efficacy in patients. Therefore, imaging spatial nanoparticle distribution and imaging the tumour environment giving rise to this distribution could help understand (lack of) clinical success of nanomedicine. Imaging the nanoparticle, drug and tumour environment can lead to improvements of new nanotherapies, increase understanding of underlying mechanisms of heterogeneous distribution, facilitate patient selection for nanotherapies and help assess the effect of treatments that aim to reduce heterogeneity in nanoparticle distribution. In this review, we discuss three groups of imaging modalities applied in nanomedicine research: non-invasive clinical imaging methods (nuclear imaging, MRI, CT, ultrasound), optical imaging and mass spectrometry imaging. Because each imaging modality provides information at a different scale and has its own strengths and weaknesses, choosing wisely and combining modalities will lead to a wealth of information that will help bring nanomedicine forward.
Collapse
|
27
|
Li Z, Ding Z, Rong D, Tang W, Cao H. Overexpression of lncRNA AFAP1-AS1 promotes cell proliferation and invasion in gastric cancer. Oncol Lett 2019; 18:3211-3217. [PMID: 31452798 PMCID: PMC6704292 DOI: 10.3892/ol.2019.10640] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2018] [Accepted: 06/12/2019] [Indexed: 12/31/2022] Open
Abstract
Long non-coding RNA (lncRNA) actin filament-associated protein 1 antisense RNA 1 (AFAP1-AS1) has been revealed to be associated with certain types of cancer. However, whether the lncRNA AFAP1-AS1 is involved in the development and progression of gastric cancer (GC) remains unknown. The present study investigated the clinical significance and biological functions of AFAP1-AS1 in GC. The expression levels of lncRNA AFAP1-AS1 in 52 patients with GC, and in 1 normal gastric mucosal cell line and 3 GC cell lines, were evaluated by reverse transcription quantitative polymerase chain reaction analysis. Small interfering RNAs were used to suppress AFAP1-AS1 expression in GC cell lines. The results indicated that AFAP1-AS1 expression levels were significantly increased in GC tissues and cell lines compared with the corresponding noncancerous tissues and normal gastric cells. In addition, the patients with GC with increased AFAP1-AS1 expression exhibited an advanced clinical stage and an association with the occurrence of lymph node metastasis compared with those with decreased AFAP1-AS1 expression. In vitro assays demonstrated that knockdown of AFAP1-AS1 decreased levels of cell proliferation and migration. In addition, the results of flow cytometry demonstrated that knockdown of AFAP1-AS1 caused cell cycle arrest. In conclusion, AFAP1-AS1 is a novel molecule involved in GC progression, which may be a potential prognostic biomarker and target for therapeutic intervention.
Collapse
Affiliation(s)
- Zhouxiao Li
- Department of General Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu 210001, P.R. China.,Department of Plastic and Hand Surgery-Campus Innenstadt, University Hospital Munich, D-80336 Munich, Germany
| | - Zhili Ding
- Department of Pediatric Intensive Care Unit, Changzhou Children's Hospital, Changzhou, Jiangsu 213018, P.R. China
| | - Dawei Rong
- Department of General Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu 210001, P.R. China
| | - Weiwei Tang
- Department of General Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu 210001, P.R. China
| | - Hongyong Cao
- Department of General Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu 210001, P.R. China
| |
Collapse
|
28
|
Korn RL, Rahmanuddin S, Borazanci E. Use of Precision Imaging in the Evaluation of Pancreas Cancer. Cancer Treat Res 2019; 178:209-236. [PMID: 31209847 DOI: 10.1007/978-3-030-16391-4_8] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Pancreas cancer is an aggressive and fatal disease that will become one of the leading causes of cancer mortality by 2030. An all-out effort is underway to better understand the basic biologic mechanisms of this disease ranging from early development to metastatic disease. In order to change the course of this disease, diagnostic radiology imaging may play a vital role in providing a precise, noninvasive method for early diagnosis and assessment of treatment response. Recent progress in combining medical imaging, advanced image analysis and artificial intelligence, termed radiomics, can offer an innovate approach in detecting the earliest changes of tumor development as well as a rapid method for the detection of response. In this chapter, we introduce the principles of radiomics and demonstrate how it can provide additional information into tumor biology, early detection, and response assessments advancing the goals of precision imaging to deliver the right treatment to the right person at the right time.
Collapse
Affiliation(s)
- Ronald L Korn
- Virginia G Piper Cancer Center at HonorHealth, Scottsdale, AZ, USA. .,Translational Genomics Research Institute, An Affiliate of City of Hope, Phoenix, AZ, USA. .,Imaging Endpoints Core Lab, Scottsdale, AZ, USA.
| | | | - Erkut Borazanci
- Virginia G Piper Cancer Center at HonorHealth, Scottsdale, AZ, USA.,Translational Genomics Research Institute, An Affiliate of City of Hope, Phoenix, AZ, USA
| |
Collapse
|
29
|
Abstract
Esophageal, esophago-gastric, and gastric cancers are major causes of cancer morbidity and cancer death. For patients with potentially resectable disease, multi-modality treatment is recommended as it provides the best chance of survival. However, quality of life may be adversely affected by therapy, and with a wide variation in outcome despite multi-modality therapy, there is a clear need to improve patient stratification. Radiomic approaches provide an opportunity to improve tumor phenotyping. In this review we assess the evidence to date and discuss how these approaches could improve outcome in esophageal, esophago-gastric, and gastric cancer.
Collapse
Affiliation(s)
- Bert-Ram Sah
- Department of Cancer Imaging, School of Biomedical Engineering and Imaging Sciences, King's College London, London, UK
| | - Kasia Owczarczyk
- Department of Cancer Imaging, School of Biomedical Engineering and Imaging Sciences, King's College London, London, UK
| | - Musib Siddique
- Department of Cancer Imaging, School of Biomedical Engineering and Imaging Sciences, King's College London, London, UK
| | - Gary J R Cook
- Department of Cancer Imaging, School of Biomedical Engineering and Imaging Sciences, King's College London, London, UK
- King's College London and Guy's and St Thomas' PET Centre, St Thomas' Hospital, London, UK
| | - Vicky Goh
- Department of Cancer Imaging, School of Biomedical Engineering and Imaging Sciences, King's College London, London, UK.
- Department of Radiology, Guy's & St Thomas' Hospitals NHS Foundation Trust, London, UK.
- Radiology, Level 1, Lambeth Wing, St Thomas' Hospital, Westminster Bridge Road, London, SE1 7EH, UK.
| |
Collapse
|
30
|
Jiang Y, Wang W, Chen C, Zhang X, Zha X, Lv W, Xie J, Huang W, Sun Z, Hu Y, Yu J, Li T, Zhou Z, Xu Y, Li G. Radiomics Signature on Computed Tomography Imaging: Association With Lymph Node Metastasis in Patients With Gastric Cancer. Front Oncol 2019; 9:340. [PMID: 31106158 PMCID: PMC6498894 DOI: 10.3389/fonc.2019.00340] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Accepted: 04/12/2019] [Indexed: 12/24/2022] Open
Abstract
Background: To evaluate whether radiomic feature-based computed tomography (CT) imaging signatures allow prediction of lymph node (LN) metastasis in gastric cancer (GC) and to develop a preoperative nomogram for predicting LN status. Methods: We retrospectively analyzed radiomics features of CT images in 1,689 consecutive patients from three cancer centers. The prediction model was developed in the training cohort and validated in internal and external validation cohorts. Lasso regression model was utilized to select features and build radiomics signature. Multivariable logistic regression analysis was utilized to develop the model. We integrated the radiomics signature, clinical T and N stage, and other independent clinicopathologic variables, and this was presented as a radiomics nomogram. The performance of the nomogram was assessed with calibration, discrimination, and clinical usefulness. Results: The radiomics signature was significantly associated with pathological LN stage in training and validation cohorts. Multivariable logistic analysis found the radiomics signature was an independent predictor of LN metastasis. The nomogram showed good discrimination and calibration. Conclusions: The newly developed radiomic signature was a powerful predictor of LN metastasis and the radiomics nomogram could facilitate the preoperative individualized prediction of LN status.
Collapse
Affiliation(s)
- Yuming Jiang
- Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China.,Guangdong Key Laboratory of Liver Disease Research, The 3rd Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Wei Wang
- Department of Gastric Surgery, Sun Yat-sen University Cancer Center, Guangzhou, China.,State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Chuanli Chen
- Department of Medical Imaging Center, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xiaodong Zhang
- Department of Radiology, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Xuefan Zha
- School of Biomedical Engineering and Guangdong Provincal Key Laboratory of Medical Image Processing, Southern Medical University, Guangzhou, China
| | - Wenbing Lv
- School of Biomedical Engineering and Guangdong Provincal Key Laboratory of Medical Image Processing, Southern Medical University, Guangzhou, China
| | - Jingjing Xie
- Center for Drug and Clinical Research, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Weicai Huang
- Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Zepang Sun
- Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yanfeng Hu
- Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jiang Yu
- Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Tuanjie Li
- Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Zhiwei Zhou
- Department of Gastric Surgery, Sun Yat-sen University Cancer Center, Guangzhou, China.,State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Yikai Xu
- Department of Medical Imaging Center, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Guoxin Li
- Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
31
|
Liu Z, Wang S, Dong D, Wei J, Fang C, Zhou X, Sun K, Li L, Li B, Wang M, Tian J. The Applications of Radiomics in Precision Diagnosis and Treatment of Oncology: Opportunities and Challenges. Theranostics 2019; 9:1303-1322. [PMID: 30867832 PMCID: PMC6401507 DOI: 10.7150/thno.30309] [Citation(s) in RCA: 512] [Impact Index Per Article: 102.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2018] [Accepted: 01/10/2019] [Indexed: 12/14/2022] Open
Abstract
Medical imaging can assess the tumor and its environment in their entirety, which makes it suitable for monitoring the temporal and spatial characteristics of the tumor. Progress in computational methods, especially in artificial intelligence for medical image process and analysis, has converted these images into quantitative and minable data associated with clinical events in oncology management. This concept was first described as radiomics in 2012. Since then, computer scientists, radiologists, and oncologists have gravitated towards this new tool and exploited advanced methodologies to mine the information behind medical images. On the basis of a great quantity of radiographic images and novel computational technologies, researchers developed and validated radiomic models that may improve the accuracy of diagnoses and therapy response assessments. Here, we review the recent methodological developments in radiomics, including data acquisition, tumor segmentation, feature extraction, and modelling, as well as the rapidly developing deep learning technology. Moreover, we outline the main applications of radiomics in diagnosis, treatment planning and evaluations in the field of oncology with the aim of developing quantitative and personalized medicine. Finally, we discuss the challenges in the field of radiomics and the scope and clinical applicability of these methods.
Collapse
Affiliation(s)
- Zhenyu Liu
- CAS Key Laboratory of Molecular Imaging, Institute of Automation, Beijing, 100190, China
- University of Chinese Academy of Sciences, Beijing, 100080, China
| | - Shuo Wang
- CAS Key Laboratory of Molecular Imaging, Institute of Automation, Beijing, 100190, China
- University of Chinese Academy of Sciences, Beijing, 100080, China
| | - Di Dong
- CAS Key Laboratory of Molecular Imaging, Institute of Automation, Beijing, 100190, China
- University of Chinese Academy of Sciences, Beijing, 100080, China
| | - Jingwei Wei
- CAS Key Laboratory of Molecular Imaging, Institute of Automation, Beijing, 100190, China
- University of Chinese Academy of Sciences, Beijing, 100080, China
| | - Cheng Fang
- Department of Hepatobiliary Surgery, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, 646000, China
| | - Xuezhi Zhou
- CAS Key Laboratory of Molecular Imaging, Institute of Automation, Beijing, 100190, China
- Engineering Research Center of Molecular and Neuro Imaging of Ministry of Education, School of Life Science and Technology, Xidian University, Xi'an, Shaanxi, 710126, China
| | - Kai Sun
- CAS Key Laboratory of Molecular Imaging, Institute of Automation, Beijing, 100190, China
- Engineering Research Center of Molecular and Neuro Imaging of Ministry of Education, School of Life Science and Technology, Xidian University, Xi'an, Shaanxi, 710126, China
| | - Longfei Li
- CAS Key Laboratory of Molecular Imaging, Institute of Automation, Beijing, 100190, China
- Collaborative Innovation Center for Internet Healthcare, Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Bo Li
- Department of Hepatobiliary Surgery, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, 646000, China
| | - Meiyun Wang
- Department of Radiology, Henan Provincial People's Hospital & the People's Hospital of Zhengzhou University, Zhengzhou, Henan, 450003, China
| | - Jie Tian
- CAS Key Laboratory of Molecular Imaging, Institute of Automation, Beijing, 100190, China
- Engineering Research Center of Molecular and Neuro Imaging of Ministry of Education, School of Life Science and Technology, Xidian University, Xi'an, Shaanxi, 710126, China
- Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, Beihang University, Beijing, 100191, China
| |
Collapse
|
32
|
Klaassen R, Larue RTHM, Mearadji B, van der Woude SO, Stoker J, Lambin P, van Laarhoven HWM. Feasibility of CT radiomics to predict treatment response of individual liver metastases in esophagogastric cancer patients. PLoS One 2018; 13:e0207362. [PMID: 30440002 PMCID: PMC6237370 DOI: 10.1371/journal.pone.0207362] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Accepted: 10/30/2018] [Indexed: 02/06/2023] Open
Abstract
In this study we investigate a CT radiomics approach to predict response to chemotherapy of individual liver metastases in patients with esophagogastric cancer (EGC). In eighteen patients with metastatic EGC treated with chemotherapy, all liver metastases were manually delineated in 3D on the pre-treatment and evaluation CT. From the pre-treatment CT scans 370 radiomics features were extracted per lesion. Random forest (RF) models were generated to discriminate partial responding (PR, >65% volume decrease, including 100% volume decrease), and complete remission (CR, only 100% volume decrease) lesions from other lesions. RF-models were build using a leave one out strategy where all lesions of a single patient were removed from the dataset and used as validation set for a model trained on the lesions of the remaining patients. This process was repeated for all patients, resulting in 18 trained models and one validation set for both the PR and CR datasets. Model performance was evaluated by receiver operating characteristics with corresponding area under the curve (AUC). In total 196 liver metastases were delineated on the pre-treatment CT, of which 99 (51%) lesions showed a decrease in size of more than 65% (PR). From the PR set a total of 47 (47% of RL, 24% of initial) lesions were no longer detected in CT scan 2 (CR). The RF-model for PR lesions showed an average training AUC of 0.79 (range: 0.74-0.83) and 0.65 (95% ci: 0.57-0.73) for the combined validation set. The RF-model for CR lesions had an average training AUC of 0.87 (range: 0.83-0.90) and 0.79 (95% ci 0.72-0.87) for the validation set. Our findings show that individual response of liver metastases varies greatly within and between patients. A CT radiomics approach shows potential in discriminating responding from non-responding liver metastases based on the pre-treatment CT scan, although further validation in an independent patient cohort is needed to validate these findings.
Collapse
Affiliation(s)
- Remy Klaassen
- Amsterdam UMC, University of Amsterdam, Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam, Netherlands
- Amsterdam UMC, University of Amsterdam, LEXOR, Laboratory for Experimental Oncology and Radiobiology, Cancer Center Amsterdam, Amsterdam, Netherlands
| | - Ruben T. H. M. Larue
- The D-Lab: Decision Support for Precision Medicine, GROW-School for Oncology and Developmental Biology, Maastricht Comprehensive Cancer Centre, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Banafsche Mearadji
- Amsterdam UMC, University of Amsterdam, Department of Radiology and Nuclear Medicine, Cancer Center Amsterdam, Amsterdam, Netherlands
| | - Stephanie O. van der Woude
- Amsterdam UMC, University of Amsterdam, Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam, Netherlands
| | - Jaap Stoker
- Amsterdam UMC, University of Amsterdam, Department of Radiology and Nuclear Medicine, Cancer Center Amsterdam, Amsterdam, Netherlands
| | - Philippe Lambin
- The D-Lab: Decision Support for Precision Medicine, GROW-School for Oncology and Developmental Biology, Maastricht Comprehensive Cancer Centre, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Hanneke W. M. van Laarhoven
- Amsterdam UMC, University of Amsterdam, Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam, Netherlands
| |
Collapse
|
33
|
Jiang Y, Yuan Q, Lv W, Xi S, Huang W, Sun Z, Chen H, Zhao L, Liu W, Hu Y, Lu L, Ma J, Li T, Yu J, Wang Q, Li G. Radiomic signature of 18F fluorodeoxyglucose PET/CT for prediction of gastric cancer survival and chemotherapeutic benefits. Am J Cancer Res 2018; 8:5915-5928. [PMID: 30613271 PMCID: PMC6299427 DOI: 10.7150/thno.28018] [Citation(s) in RCA: 98] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Accepted: 10/22/2018] [Indexed: 12/13/2022] Open
Abstract
We aimed to evaluate whether radiomic feature-based fluorine 18 (18F) fluorodeoxyglucose (FDG) positron emission tomography (PET) imaging signatures allow prediction of gastric cancer (GC) survival and chemotherapy benefits. Methods: A total of 214 GC patients (training (n = 132) or validation (n = 82) cohort) were subjected to radiomic feature extraction (80 features). Radiomic features of patients in the training cohort were subjected to a LASSO cox analysis to predict disease-free survival (DFS) and overall survival (OS) and were validated in the validation cohort. A radiomics nomogram with the radiomic signature incorporated was constructed to demonstrate the incremental value of the radiomic signature to the TNM staging system for individualized survival estimation, which was then assessed with respect to calibration, discrimination, and clinical usefulness. The performance was assessed with concordance index (C-index) and integrated Brier scores. Results: Significant differences were found between the high- and low-radiomic score (Rad-score) patients in 5-year DFS and OS in training and validation cohorts. Multivariate analysis revealed that the Rad-score was an independent prognostic factor. Incorporating the Rad-score into the radiomics-based nomogram resulted in better performance (C-index: DFS, 0.800; OS, 0.786; in the training cohort) than TNM staging system and clinicopathologic nomogram. Further analysis revealed that patients with higher Rad-scores were prone to benefit from chemotherapy. Conclusion: The newly developed radiomic signature was a powerful predictor of OS and DFS. Moreover, the radiomic signature could predict which patients could benefit from chemotherapy.
Collapse
|
34
|
Giganti F, Tang L, Baba H. Gastric cancer and imaging biomarkers: Part 1 - a critical review of DW-MRI and CE-MDCT findings. Eur Radiol 2018; 29:1743-1753. [PMID: 30280246 PMCID: PMC6420485 DOI: 10.1007/s00330-018-5732-4] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2018] [Revised: 08/13/2018] [Accepted: 08/28/2018] [Indexed: 12/17/2022]
Abstract
Abstract The current standard of care for gastric cancer imaging includes heterogeneity in image acquisition techniques and qualitative image interpretation. In addition to qualitative assessment, several imaging techniques, including diffusion-weighted magnetic resonance imaging (DW-MRI), contrast-enhanced multidetector computed tomography (CE-MDCT), dynamic-contrast enhanced MRI and 18F-fluorodeoxyglucose positron emission tomography, can allow quantitative analysis. However, so far there is no consensus regarding the application of functional imaging in the management of gastric cancer. The aim of this article is to specifically review two promising biomarkers for gastric cancer with reasonable spatial resolution: the apparent diffusion coefficient (ADC) from DW-MRI and textural features from CE-MDCT. We searched MEDLINE/ PubMed for manuscripts published from inception to 6 February 2018. Initially, we searched for (gastric cancer OR gastric tumour) AND diffusion weighted magnetic resonance imaging. Then, we searched for (gastric cancer OR gastric tumour) AND texture analysis AND computed tomography. We collated the results from the studies related to this query. There is evidence that: (1) the ADC is a promising biomarker for the evaluation of the aggressiveness (T and N stage), treatment response and prognosis of gastric cancer; (2) textural features are related to the degree of differentiation, Lauren classification, treatment response and prognosis of gastric cancer. We conclude that these imaging biomarkers hold promise as effective additional tools in the diagnostic pathway of gastric cancer and may facilitate the multidisciplinary work between the radiologist and clinician, and across different institutions, to provide a greater biological understanding of gastric cancer. Key Points • Quantitative imaging is the extraction of quantifiable features from medical images for the assessment of normal or pathological conditions and represents a promising area for gastric cancer. • Quantitative analysis from CE-MDCT and DW-MRI allows the extrapolation of multiple imaging biomarkers. • ADC from DW-MRI and CE- MDCT-based texture features are non-invasive, quantitative imaging biomarkers that hold promise in the evaluation of the aggressiveness, treatment response and prognosis of gastric cancer.
Collapse
Affiliation(s)
- Francesco Giganti
- Department of Radiology, University College London Hospital NHS Foundation Trust, London, UK. .,Division of Surgery and Interventional Science, Faculty of Medical Sciences, University College London, 3rd Floor, Charles Bell House, 43-45 Foley St, London, W1W 7TS, UK.
| | - Lei Tang
- Department of Radiology, Peking University Cancer Hospital, Beijing, China
| | - Hideo Baba
- Department of Gastroenterological Surgery, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| |
Collapse
|
35
|
Jiang Y, Chen C, Xie J, Wang W, Zha X, Lv W, Chen H, Hu Y, Li T, Yu J, Zhou Z, Xu Y, Li G. Radiomics signature of computed tomography imaging for prediction of survival and chemotherapeutic benefits in gastric cancer. EBioMedicine 2018; 36:171-182. [PMID: 30224313 PMCID: PMC6197796 DOI: 10.1016/j.ebiom.2018.09.007] [Citation(s) in RCA: 127] [Impact Index Per Article: 21.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2018] [Revised: 09/07/2018] [Accepted: 09/07/2018] [Indexed: 02/08/2023] Open
Abstract
To develop and validate a radiomics signature for the prediction of gastric cancer (GC) survival and chemotherapeutic benefits. In this multicenter retrospective analysis, we analyzed the radiomics features of portal venous-phase computed tomography in 1591 consecutive patients. A radiomics signature was generated by using the Lasso-Cox regression model in 228 patients and validated in internal and external validation cohorts. Radiomics nomograms integrating the radiomics signature were constructed, demonstrating the incremental value of the radiomics signature to the traditional staging system for individualized survival estimation. The performance of the nomograms was assessed with respect to calibration, discrimination, and clinical usefulness. The radiomics signature consisted of 19 selected features and was significantly associated with DFS (disease-free survival) and OS (overall survival). Multivariate analysis demonstrated that the radiomics signature was an independent prognostic factor. Incorporating the radiomics signature into the radiomics-based nomograms resulted in better performance for the estimation of DFS and OS than the clinicopathological nomograms and TNM staging system, with improved accuracy of the classification of survival outcomes. Further analysis showed that stage II and III patients with higher radiomics scores exhibited a favorable response to chemotherapy. In conclusion, the newly developed radiomics signature is a powerful predictor of DFS and OS, and it may predict which patients with stage II and III GC benefit from chemotherapy.
Collapse
Affiliation(s)
- Yuming Jiang
- Department of General Surgery, Nanfang Hospital, Southern Medical University, 1838 North Guangzhou Avenue, Guangzhou, China
| | - Chuanli Chen
- Department of Medical Imaging Center, Nanfang Hospital, Southern Medical University, No. 1838, Guangzhou Avenue North, 510515 Guangzhou, China
| | - Jingjing Xie
- Research Center for Clinical Pharmacology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Wei Wang
- State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China; Department of Gastric and Pancreatic Surgery, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Xuefan Zha
- Department of Biomedical Engineering, Southern Medical University, Guangzhou, China
| | - Wenbing Lv
- Department of Biomedical Engineering, Southern Medical University, Guangzhou, China
| | - Hao Chen
- Department of General Surgery, Nanfang Hospital, Southern Medical University, 1838 North Guangzhou Avenue, Guangzhou, China
| | - Yanfeng Hu
- Department of General Surgery, Nanfang Hospital, Southern Medical University, 1838 North Guangzhou Avenue, Guangzhou, China
| | - Tuanjie Li
- Department of General Surgery, Nanfang Hospital, Southern Medical University, 1838 North Guangzhou Avenue, Guangzhou, China
| | - Jiang Yu
- Department of General Surgery, Nanfang Hospital, Southern Medical University, 1838 North Guangzhou Avenue, Guangzhou, China.
| | - Zhiwei Zhou
- State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China; Department of Gastric and Pancreatic Surgery, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China.
| | - Yikai Xu
- Department of Medical Imaging Center, Nanfang Hospital, Southern Medical University, No. 1838, Guangzhou Avenue North, 510515 Guangzhou, China.
| | - Guoxin Li
- Department of General Surgery, Nanfang Hospital, Southern Medical University, 1838 North Guangzhou Avenue, Guangzhou, China.
| |
Collapse
|
36
|
Yun G, Kim YH, Lee YJ, Kim B, Hwang JH, Choi DJ. Tumor heterogeneity of pancreas head cancer assessed by CT texture analysis: association with survival outcomes after curative resection. Sci Rep 2018; 8:7226. [PMID: 29740111 PMCID: PMC5940761 DOI: 10.1038/s41598-018-25627-x] [Citation(s) in RCA: 76] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Accepted: 04/25/2018] [Indexed: 12/12/2022] Open
Abstract
The value of image based texture features as a powerful method to predict prognosis and assist clinical management in cancer patients has been established recently. However, texture analysis using histograms and grey-level co-occurrence matrix in pancreas cancer patients has rarely been reported. We aimed to analyze the association of survival outcomes with texture features in pancreas head cancer patients. Eighty-eight pancreas head cancer patients who underwent preoperative CT images followed by curative resection were included. Texture features using different filter values were obtained. The texture features of average, contrast, correlation, and standard deviation with no filter, and fine to medium filter values as well as the presence of nodal metastasis were significantly different between the recurred (n = 70, 79.5%) and non-recurred group (n = 18, 20.5%). In the multivariate Cox regression analysis, lower standard deviation and contrast and higher correlation with lower average value representing homogenous texture were significantly associated with poorer DFS (disease free survival), along with the presence of lymph node metastasis. Texture parameters from routinely performed pre-operative CT images could be used as an independent imaging tool for predicting the prognosis in pancreas head cancer patients who underwent curative resection.
Collapse
Affiliation(s)
- Gabin Yun
- Seoul National University Bundang Hospital, Department of Radiology, Seongnam, 13620, Korea
| | - Young Hoon Kim
- Seoul National University Bundang Hospital, Department of Radiology, Seongnam, 13620, Korea.
| | - Yoon Jin Lee
- Seoul National University Bundang Hospital, Department of Radiology, Seongnam, 13620, Korea
| | - Bohyoung Kim
- Seoul National University Bundang Hospital, Department of Radiology, Seongnam, 13620, Korea.,Hankuk University of Foreign Studies, Division of Biomedical Engineering, Yongin, 17035, Korea
| | - Jin-Hyeok Hwang
- Seoul National University Bundang Hospital, Department of Internal Medicine, Seongnam, 13620, Korea
| | - Dong Joon Choi
- Seoul National University Bundang Hospital, Department of Radiology, Seongnam, 13620, Korea
| |
Collapse
|
37
|
Liu S, Shi H, Ji C, Zheng H, Pan X, Guan W, Chen L, Sun Y, Tang L, Guan Y, Li W, Ge Y, He J, Liu S, Zhou Z. Preoperative CT texture analysis of gastric cancer: correlations with postoperative TNM staging. Clin Radiol 2018; 73:756.e1-756.e9. [PMID: 29625746 DOI: 10.1016/j.crad.2018.03.005] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2017] [Accepted: 03/08/2018] [Indexed: 02/07/2023]
Abstract
AIM To explore the role of computed tomography (CT) texture analysis in predicting pathologic stage of gastric cancers. MATERIALS AND METHODS Preoperative enhanced CT images of 153 patients (112 men, 41 women) with gastric cancers were reviewed retrospectively. Regions of interest (ROIs) were manually drawn along the margin of the lesion on the section where it appeared largest on the arterial and venous CT images, which yielded texture parameters, including mean, maximum frequency, mode, skewness, kurtosis, and entropy. Correlations between texture parameters and pathological stage were analysed with Spearman's correlation test. The diagnostic performance of CT texture parameters in differentiating different stages was evaluated using receiver operating characteristic (ROC) analysis. RESULTS Maximum frequency in the arterial phase and mean, maximum frequency, mode in the venous phase correlated positively with T stage, N stage, and overall stage (all p<0.05) of gastric cancer. Entropy in the venous phase also correlated positively with N stage (p=0.009) and overall stage (p=0.032). Skewness in the arterial phase had the highest area under the ROC curve (AUC) of 0.822 in identifying early from advanced gastric cancers. Multivariate analysis identified four parameters, including maximum frequency, skewness, entropy in the venous phase, and differentiation degree from biopsy, for predicting lymph node metastasis of gastric cancer. The multivariate model could distinguish gastric cancers with and without lymph node metastasis with an AUC of 0.892. CONCLUSION Multiple CT texture parameters, especially those in the venous phase, correlated well with pathological stage and hold great potential in predicting lymph node metastasis of gastric cancers.
Collapse
Affiliation(s)
- S Liu
- Department of Radiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, 210008, China
| | - H Shi
- Department of Radiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, 210008, China
| | - C Ji
- Department of Radiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, 210008, China
| | - H Zheng
- Department of Radiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, 210008, China
| | - X Pan
- Department of Radiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, 210008, China
| | - W Guan
- Department of Gastrointestinal Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, 210008, China
| | - L Chen
- Department of Pathology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, 210008, China
| | - Y Sun
- Department of Radiology, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - L Tang
- Department of Radiology, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Y Guan
- School of Electronic Science and Engineering, Nanjing University, Nanjing, 210046, China
| | - W Li
- School of Electronic Science and Engineering, Nanjing University, Nanjing, 210046, China
| | - Y Ge
- School of Electronic Science and Engineering, Nanjing University, Nanjing, 210046, China
| | - J He
- Department of Radiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, 210008, China.
| | - S Liu
- Department of Radiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, 210008, China.
| | - Z Zhou
- Department of Radiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, 210008, China.
| |
Collapse
|
38
|
Could texture features from preoperative CT image be used for predicting occult peritoneal carcinomatosis in patients with advanced gastric cancer? PLoS One 2018; 13:e0194755. [PMID: 29596522 PMCID: PMC5875782 DOI: 10.1371/journal.pone.0194755] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Accepted: 03/09/2018] [Indexed: 12/19/2022] Open
Abstract
Purpose To retrospectively investigate whether texture features obtained from preoperative CT images of advanced gastric cancer (AGC) patients could be used for the prediction of occult peritoneal carcinomatosis (PC) detected during operation. Materials and methods 51 AGC patients with occult PC detected during operation from January 2009 to December 2012 were included as occult PC group. For the control group, other 51 AGC patients without evidence of distant metastasis including PC, and whose clinical T and N stage could be matched to those of the patients of the occult PC group, were selected from the period of January 2011 to July 2012. Each group was divided into test (n = 41) and validation cohort (n = 10). Demographic and clinical data of these patients were acquired from the hospital database. Texture features including average, standard deviation, kurtosis, skewness, entropy, correlation, and contrast were obtained from manually drawn region of interest (ROI) over the omentum on the axial CT image showing the omentum at its largest cross sectional area. After using Fisher's exact and Wilcoxon signed-rank test for comparison of the clinical and texture features between the two groups of the test cohort, conditional logistic regression analysis was performed to determine significant independent predictor for occult PC. Using the optimal cut-off value from receiver operating characteristic (ROC) analysis for the significant variables, diagnostic sensitivity and specificity were determined in the test cohort. The cut-off value of the significant variables obtained from the test cohort was then applied to the validation cohort. Bonferroni correction was used to adjust P value for multiple comparisons. Results Between the two groups, there was no significant difference in the clinical features. Regarding the texture features, the occult PC group showed significantly higher average, entropy, standard deviation, and significantly lower correlation (P value < 0.004 for all). Conditional logistic regression analysis demonstrated that entropy was significant independent predictor for occult PC. When the cut-off value of entropy (> 7.141) was applied to the validation cohort, sensitivity and specificity for the prediction of occult PC were 80% and 90%, respectively. Conclusion For AGC patients whose PC cannot be detected with routine imaging such as CT, texture analysis may be a useful adjunct for the prediction of occult PC.
Collapse
|
39
|
Li Z, Zhang D, Dai Y, Dong J, Wu L, Li Y, Cheng Z, Ding Y, Liu Z. Computed tomography-based radiomics for prediction of neoadjuvant chemotherapy outcomes in locally advanced gastric cancer: A pilot study. Chin J Cancer Res 2018; 30:406-414. [PMID: 30210220 PMCID: PMC6129565 DOI: 10.21147/j.issn.1000-9604.2018.04.03] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Objective The standard treatment for patients with locally advanced gastric cancer has relied on perioperative radio-chemotherapy or chemotherapy and surgery. The aim of this study was to investigate the wealth of radiomics for pre-treatment computed tomography (CT) in the prediction of the pathological response of locally advanced gastric cancer with preoperative chemotherapy. Methods Thirty consecutive patients with CT-staged II/III gastric cancer receiving neoadjuvant chemotherapy were enrolled in this study between December 2014 and March 2017. All patients underwent upper abdominal CT during the unenhanced, late arterial phase (AP) and portal venous phase (PP) before the administration of neoadjuvant chemotherapy. In total, 19,985 radiomics features were extracted in the AP and PP for each patient. Four methods were adopted during feature selection and eight methods were used in the process of building the classifier model. Thirty-two combinations of feature selection and classification methods were examined. Receiver operating characteristic (ROC) curves were used to evaluate the capability of each combination of feature selection and classification method to predict a non-good response (non-GR) based on tumor regression grade (TRG). Results The mean area under the curve (AUC) ranged from 0.194 to 0.621 in the AP, and from 0.455 to 0.722 in the PP, according to different combinations of feature selection and the classification methods. There was only one cross-combination machine-learning method indicating a relatively higher AUC (>0.600) in the AP, while 12 cross-combination machine-learning methods presented relatively higher AUCs (all >0.600) in the PP. The feature selection method adopted by a filter based on linear discriminant analysis + classifier of random forest achieved a significantly prognostic performance in the PP (AUC, 0.722±0.108; accuracy, 0.793; sensitivity, 0.636; specificity, 0.889; Z=2.039; P=0.041). Conclusions It is possible to predict non-GR after neoadjuvant chemotherapy in locally advanced gastric cancers based on the radiomics of CT.
Collapse
Affiliation(s)
- Zhenhui Li
- Department of Radiology, the Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Kunming 650118, China
| | - Dafu Zhang
- Department of Radiology, the Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Kunming 650118, China
| | - Youguo Dai
- Department of Abdominal Surgery, the Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Kunming 650118, China
| | - Jian Dong
- Department of Abdominal Surgery, the Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Kunming 650118, China
| | - Lin Wu
- Department of Pathology, the Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Kunming 650118, China
| | - Yajun Li
- Department of Radiology, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, China.,School of Medicine, South China University of Technology, Guangzhou 510641, China
| | - Zixuan Cheng
- Department of Radiology, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, China.,School of Medicine, South China University of Technology, Guangzhou 510641, China
| | - Yingying Ding
- Department of Radiology, the Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Kunming 650118, China
| | - Zaiyi Liu
- Department of Radiology, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, China
| |
Collapse
|
40
|
Lubner MG, Smith AD, Sandrasegaran K, Sahani DV, Pickhardt PJ. CT Texture Analysis: Definitions, Applications, Biologic Correlates, and Challenges. Radiographics 2017; 37:1483-1503. [PMID: 28898189 DOI: 10.1148/rg.2017170056] [Citation(s) in RCA: 536] [Impact Index Per Article: 76.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
This review discusses potential oncologic and nononcologic applications of CT texture analysis ( CTTA CT texture analysis ), an emerging area of "radiomics" that extracts, analyzes, and interprets quantitative imaging features. CTTA CT texture analysis allows objective assessment of lesion and organ heterogeneity beyond what is possible with subjective visual interpretation and may reflect information about the tissue microenvironment. CTTA CT texture analysis has shown promise in lesion characterization, such as differentiating benign from malignant or more biologically aggressive lesions. Pretreatment CT texture features are associated with histopathologic correlates such as tumor grade, tumor cellular processes such as hypoxia or angiogenesis, and genetic features such as KRAS or epidermal growth factor receptor (EGFR) mutation status. In addition, and likely as a result, these CT texture features have been linked to prognosis and clinical outcomes in some tumor types. CTTA CT texture analysis has also been used to assess response to therapy, with decreases in tumor heterogeneity generally associated with pathologic response and improved outcomes. A variety of nononcologic applications of CTTA CT texture analysis are emerging, particularly quantifying fibrosis in the liver and lung. Although CTTA CT texture analysis seems to be a promising imaging biomarker, there is marked variability in methods, parameters reported, and strength of associations with biologic correlates. Before CTTA CT texture analysis can be considered for widespread clinical implementation, standardization of tumor segmentation and measurement techniques, image filtration and postprocessing techniques, and methods for mathematically handling multiple tumors and time points is needed, in addition to identification of key texture parameters among hundreds of potential candidates, continued investigation and external validation of histopathologic correlates, and structured reporting of findings. ©RSNA, 2017.
Collapse
Affiliation(s)
- Meghan G Lubner
- From the Department of Radiology, University of Wisconsin School of Medicine and Public Health, E3/311 Clinical Sciences Center, 600 Highland Ave, Madison, WI 35792 (M.G.L., P.J.P.); Department of Radiology, University of Mississippi Medical Center, Jackson, Miss (A.D.S.); Department of Radiology, Indiana University School of Medicine, Indianapolis, Ind (K.S.); and Department of Radiology, Harvard Medical School, Boston, Mass (D.V.S.)
| | - Andrew D Smith
- From the Department of Radiology, University of Wisconsin School of Medicine and Public Health, E3/311 Clinical Sciences Center, 600 Highland Ave, Madison, WI 35792 (M.G.L., P.J.P.); Department of Radiology, University of Mississippi Medical Center, Jackson, Miss (A.D.S.); Department of Radiology, Indiana University School of Medicine, Indianapolis, Ind (K.S.); and Department of Radiology, Harvard Medical School, Boston, Mass (D.V.S.)
| | - Kumar Sandrasegaran
- From the Department of Radiology, University of Wisconsin School of Medicine and Public Health, E3/311 Clinical Sciences Center, 600 Highland Ave, Madison, WI 35792 (M.G.L., P.J.P.); Department of Radiology, University of Mississippi Medical Center, Jackson, Miss (A.D.S.); Department of Radiology, Indiana University School of Medicine, Indianapolis, Ind (K.S.); and Department of Radiology, Harvard Medical School, Boston, Mass (D.V.S.)
| | - Dushyant V Sahani
- From the Department of Radiology, University of Wisconsin School of Medicine and Public Health, E3/311 Clinical Sciences Center, 600 Highland Ave, Madison, WI 35792 (M.G.L., P.J.P.); Department of Radiology, University of Mississippi Medical Center, Jackson, Miss (A.D.S.); Department of Radiology, Indiana University School of Medicine, Indianapolis, Ind (K.S.); and Department of Radiology, Harvard Medical School, Boston, Mass (D.V.S.)
| | - Perry J Pickhardt
- From the Department of Radiology, University of Wisconsin School of Medicine and Public Health, E3/311 Clinical Sciences Center, 600 Highland Ave, Madison, WI 35792 (M.G.L., P.J.P.); Department of Radiology, University of Mississippi Medical Center, Jackson, Miss (A.D.S.); Department of Radiology, Indiana University School of Medicine, Indianapolis, Ind (K.S.); and Department of Radiology, Harvard Medical School, Boston, Mass (D.V.S.)
| |
Collapse
|
41
|
Liu S, Pan X, Liu R, Zheng H, Chen L, Guan W, Wang H, Sun Y, Tang L, Guan Y, Ge Y, He J, Zhou Z. Texture analysis of CT images in predicting malignancy risk of gastrointestinal stromal tumours. Clin Radiol 2017; 73:266-274. [PMID: 28969853 DOI: 10.1016/j.crad.2017.09.003] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2017] [Revised: 07/27/2017] [Accepted: 09/04/2017] [Indexed: 12/14/2022]
Abstract
AIM To explore the role of texture analysis of computed tomography (CT) images in predicting the malignancy risk of gastrointestinal stromal tumours (GISTs). MATERIALS AND METHODS Seventy-eight patients with histopathologically confirmed GISTs underwent preoperative CT. Texture analysis was performed on unenhanced and contrast-enhanced CT images, respectively. Fourteen CT texture parameters were obtained and compared among GISTs at different malignancy risks with one-way analysis of variance or independent-samples Kruskal-Wallis test. Correlations between CT texture parameters and malignancy risk were analysed with Spearman's correlation test. Diagnostic performance of CT texture parameters in differentiating GISTs at low/very low malignancy risk was tested with receiver operating characteristic (ROC) analysis. RESULTS Three parameters on unenhanced images (r=-0.268-0.506), four parameters on arterial phase (r=-0.365-0.508), and six parameters on venous phase (r=-0.343-0.481) imaging correlated significantly with malignancy risk of GISTs, respectively (all p<0.05). For identifying GISTs at low/very low malignancy risk, three parameters on unenhanced images (area under ROC curve [AUC], 0.676-0.802), four parameters on arterial phase (AUC, 0.637-0.811), and six parameters on venous phase (AUC, 0.636-0.791) imaging showed significant diagnostic performance, respectively (all p<0.05), especially maximum frequency on both unenhanced and contrast-enhanced images (AUC, 0.791-0.811). CONCLUSION Texture analysis of CT images holds great potential to predict the malignancy risk of GISTs preoperatively.
Collapse
Affiliation(s)
- S Liu
- Department of Radiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, 210008, China
| | - X Pan
- Department of Radiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, 210008, China
| | - R Liu
- Department of Radiology, Xi'an Central Hospital, Affiliated to Xi'an Jiaotong University, Xi'an, 710004, China
| | - H Zheng
- Department of Radiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, 210008, China
| | - L Chen
- Department of Pathology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, 210008, China
| | - W Guan
- Department of General Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, 210008, China
| | - H Wang
- Department of General Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, 210008, China
| | - Y Sun
- Department of Radiology, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - L Tang
- Department of Radiology, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Y Guan
- School of Electronic Science and Engineering, Nanjing University, Nanjing, 210046, China
| | - Y Ge
- School of Electronic Science and Engineering, Nanjing University, Nanjing, 210046, China.
| | - J He
- Department of Radiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, 210008, China. ,
| | - Z Zhou
- Department of Radiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, 210008, China.
| |
Collapse
|