1
|
Guan J, Gong X, Zeng H, Zhang W, Qin Q, Gou H, Liu X, Song B. Gastrointestinal tumor personalized immunotherapy: an integrated analysis from molecular genetics to imaging biomarkers. Therap Adv Gastroenterol 2025; 18:17562848251333527. [PMID: 40297204 PMCID: PMC12035075 DOI: 10.1177/17562848251333527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Accepted: 03/24/2025] [Indexed: 04/30/2025] Open
Abstract
The immunotherapy landscape for gastrointestinal (GI) tumors is rapidly evolving. There is an urgent need for reliable biomarkers capable of predicting treatment outcomes to optimize therapeutic strategies and enhance patient prognosis. This review presents a comprehensive overview of biomarkers associated with the immunotherapy response of GI tumors, covering advances in molecular genetics, histopathological markers, and imaging. Key molecular biomarkers, such as microsatellite instability, tumor mutational burden, and programmed death-ligand 1 expression, remain critical for identifying patients likely to benefit from immune checkpoint inhibitors. The significance of tumor-infiltrating lymphocytes, notably the CD8+ T cell to regulatory T cell ratio, as a predictor of immunotherapy response is explored. In addition, advanced imaging techniques, including computed tomography (CT), magnetic resonance imaging, and positron emission tomography-CT, facilitate the noninvasive evaluation of tumor biology and therapeutic response. By bridging molecular and imaging data, this integrated strategy enhances precision in patient selection, treatment monitoring, and adaptive therapy design. Future studies should aim to validate these biomarkers in larger, multicenter cohorts and focus on clinical translation to advance precision medicine in GI oncology.
Collapse
Affiliation(s)
- Jian Guan
- Department of Radiology, West China Hospital, Sichuan University, Chengdu, China
- Department of Radiology, Sichuan Provincial Corps Hospital, Chinese People’s Armed Police Forces, Leshan, China
| | - Xiaoling Gong
- Department of Radiology, West China Hospital, Sichuan University, Chengdu, China
| | - Hanjiang Zeng
- Department of Radiology, West China Hospital, Sichuan University, Chengdu, China
| | - Wei Zhang
- Department of Radiology, Sichuan Provincial Corps Hospital, Chinese People’s Armed Police Forces, Leshan, China
| | - Qing Qin
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Hongfeng Gou
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Xijiao Liu
- Department of Radiology, West China Hospital, Sichuan University, No. 37, Guoxue Alley, Chengdu 610041, China
- Department of Radiology, Sanya People’s Hospital, Sanya, Hainan, China
| | - Bin Song
- Department of Radiology, West China Hospital, Sichuan University, No. 37, Guoxue Alley, Chengdu 610041, China
- Department of Radiology, Sanya People’s Hospital, Sanya, Hainan, China
| |
Collapse
|
2
|
Mohammadpour S, Emami H, Rabiei R, Hosseini A, Moghaddasi H, Faeghi F, Bagherzadeh R. Image Analysis as tool for Predicting Colorectal Cancer Molecular Alterations: A Scoping Review. Mol Imaging Radionucl Ther 2025; 34:10-25. [PMID: 39917985 PMCID: PMC11827529 DOI: 10.4274/mirt.galenos.2024.86402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Accepted: 08/25/2024] [Indexed: 02/16/2025] Open
Abstract
Objectives Among the most important diagnostic indicators of colorectal cancer; however, measuring molecular alterations are invasive and expensive. This study aimed to investigate the application of image processing to predict molecular alterations in colorectal cancer. Methods In this scoping review, we searched for relevant literature by searching the Web of Science, Scopus, and PubMed databases. The method of selecting the articles and reporting the findings was according to the guidelines of the Preferred Reporting Items for Systematic Reviews and Meta-Analyses; moreover, the Strengthening the Reporting of Observational Studies in Epidemiology checklist was used to assess the quality of the studies. Results Sixty seven out of 2,223 articles, 67 were relevant to the aim of the study, and finally 41 studies with sufficient quality were reviewed. The prediction of Kirsten Rat Sarcoma Viral Oncogene Homolog (KRAS), Neuroblastoma RAS Viral (NRAS), B-Raf proto-oncogene, serine/threonine kinase (BRAF), Tumor Protein 53 (TP53), Adenomatous Polyposis Coli, and microsatellite instability (MSI) with the help of image analysis has received more attention than other molecular characteristics. The studies used computed tomography (CT), magnetic resonance imaging (MRI), and 18F-FDG positron emission tomography (PET)/CT with radionics and quantitative analysis to predict molecular alterations in colorectal cancer, analyzing features like texture, maximum standard uptake value, and MTV using various statistical methods. In 39 studies, there was a significant relationship between the features extracted from these images and molecular alterations. Different modalities were used to measure the area under the receiver operating characteristic curve for predicting the alterations in KRAS, MSI, BRAF, and TP53, with an average of 78, 81, 80 and 71%, respectively. Conclusion This scoping review underscores the potential of radiogenomics in predicting molecular alterations in colorectal cancer through non-invasive imaging modalities, like CT, MRI, and 18F-FDG PET/CT. The analysis of 41 studies showed the appropriate prediction of key alterations, such as KRAS, NRAS, BRAF, TP53, and MSI, highlighting the promise of radionics and texture features in enhancing predictive accuracy.
Collapse
Affiliation(s)
- Saman Mohammadpour
- Shahid Beheshti University Faculty of Medicine, Department of Health Information Technology and Management, Tehran, Iran
| | - Hassan Emami
- Shahid Beheshti University Faculty of Medicine, Department of Health Information Technology and Management, Tehran, Iran
| | - Reza Rabiei
- Shahid Beheshti University Faculty of Medicine, Department of Health Information Technology and Management, Tehran, Iran
| | - Azamossadat Hosseini
- Shahid Beheshti University Faculty of Medicine, Department of Health Information Technology and Management, Tehran, Iran
| | - Hamid Moghaddasi
- Shahid Beheshti University Faculty of Medicine, Department of Health Information Technology and Management, Tehran, Iran
| | - Fariborz Faeghi
- Shahid Beheshti University Faculty of Medicine, Department of Radiology Technology, Tehran, Iran
| | - Rafat Bagherzadeh
- Iran University of Medical Sciences Faculty of Medicine, Department of English Language, Tehran, Iran
| |
Collapse
|
3
|
Huang X, Yang Z, Qin W, Li X, Su S, Huang J. Construction of machine learning models based on transrectal ultrasound combined with contrast-enhanced ultrasound to predict preoperative regional lymph node metastasis of rectal cancer. Heliyon 2024; 10:e26433. [PMID: 38390137 PMCID: PMC10882134 DOI: 10.1016/j.heliyon.2024.e26433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Revised: 02/12/2024] [Accepted: 02/13/2024] [Indexed: 02/24/2024] Open
Abstract
Purpose Constructing a machine learning model based on transrectal ultrasound (TRUS) combined with contrast-enhanced ultrasound (CEUS) to predict preoperative regional lymph node metastasis (RLNM) of rectal cancer and provide new references for decision-making. Materials and methods 233 patients with rectal cancer were enrolled and underwent TRUS and CEUS prior to surgery. Clinicopathological and ultrasound data were collected to analyze the correlation of RLNM status, clinical features and ultrasound parameters. A 75% training set and 25% test set were utilized to construct seven machine learning algorithms. The DeLong test was used to assess the model's diagnostic performance, then chose the best one to predict RLNM of rectal cancer. Results The diagnostic performance was most dependent on the following: MMT difference (36), length (30), location (29), AUC ratio (27), and PI ratio (24). The prediction accuracy, sensitivity, specificity, precision, and F1 score range of KNN, Bayes, MLP, LR, SVM, RF, and LightGBM were (0.553-0.857), (0.000-0.935), (0.600-1.000), (0.557-0.952), and (0.617-0.852), respectively. The LightGBM model exhibited the optimal accuracy (0.857) and F1 score (0.852). The AUC for machine learning analytics were (0.517-0.941, 95% CI: 0.380-0.986). The LightGBM model exhibited the highest AUC (0.941, 95% CI: 0.843-0.986), though no statistic significant showed in comparison with the SVM, LR, RF, and MLP models (P > 0.05), it was significantly higher than that of the KNN and Bayes models (P < 0.05). Conclusion The LightGBM machine learning model based on TRUS combined with CEUS may help predict RLNM prior to surgery and provide new references for clinical treatment in rectal cancer.
Collapse
Affiliation(s)
- Xuanzhang Huang
- Department of Ultrasound, The First Affiliated Hospital of Guangxi Medical University, Nanning, PR China
| | - Zhendong Yang
- Department of Radiation Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning, PR China
| | - Wanyue Qin
- Department of Ultrasound, The First Affiliated Hospital of Guangxi Medical University, Nanning, PR China
| | - Xigui Li
- Department of Ultrasound, The First Affiliated Hospital of Guangxi Medical University, Nanning, PR China
| | - Shitao Su
- Department of Ultrasound, The First Affiliated Hospital of Guangxi Medical University, Nanning, PR China
| | - Jianyuan Huang
- Department of Ultrasound, The First Affiliated Hospital of Guangxi Medical University, Nanning, PR China
| |
Collapse
|
4
|
Xiang Y, Li S, Song M, Wang H, Hu K, Wang F, Wang Z, Niu Z, Liu J, Cai Y, Li Y, Zhu X, Geng J, Zhang Y, Teng H, Wang W. KRAS status predicted by pretreatment MRI radiomics was associated with lung metastasis in locally advanced rectal cancer patients. BMC Med Imaging 2023; 23:210. [PMID: 38087207 PMCID: PMC10717608 DOI: 10.1186/s12880-023-01173-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Accepted: 12/05/2023] [Indexed: 12/18/2023] Open
Abstract
BACKGROUND Mutated KRAS may indicate an invasive nature and predict prognosis in locally advanced rectal cancer (LARC). We aimed to establish a radiomic model using pretreatment T2W MRIs to predict KRAS status and explore the association between the KRAS status or model predictions and lung metastasis. METHODS In this retrospective multicentre study, LARC patients from two institutions between January 2012 and January 2019 were randomly divided into training and testing cohorts. Least absolute shrinkage and selection operator (LASSO) regression and the support vector machine (SVM) classifier were utilized to select significant radiomic features and establish a prediction model, which was validated by radiomic score distribution and decision curve analysis. The association between the model stratification and lung metastasis was investigated by Cox regression and Kaplan‒Meier survival analysis; the results were compared by the log-rank test. RESULTS Overall, 103 patients were enrolled (73 and 30 in the training and testing cohorts, respectively). The median follow-up was 38.1 months (interquartile range: 26.9, 49.4). The radiomic model had an area under the curve (AUC) of 0.983 in the training cohort and 0.814 in the testing cohort. Using a cut-off of 0.679 defined by the receiver operating characteristic (ROC) curve, patients with a high radiomic score (RS) had a higher risk for lung metastasis (HR 3.565, 95% CI 1.337, 9.505, p = 0.011), showing similar predictive performances for the mutant and wild-type KRAS groups (HR 3.225, 95% CI 1.249, 8.323, p = 0.016, IDI: 1.08%, p = 0.687; NRI 2.23%, p = 0.766). CONCLUSIONS We established and validated a radiomic model for predicting KRAS status in LARC. Patients with high RS experienced more lung metastases. The model could noninvasively detect KRAS status and may help individualize clinical decision-making.
Collapse
Affiliation(s)
- Yirong Xiang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Radiation Oncology, Peking University Cancer Hospital and Institute, Beijing, 100142, China
| | - Shuai Li
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Radiation Oncology, Peking University Cancer Hospital and Institute, Beijing, 100142, China
| | - Maxiaowei Song
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Radiation Oncology, Peking University Cancer Hospital and Institute, Beijing, 100142, China
| | - Hongzhi Wang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Radiation Oncology, Peking University Cancer Hospital and Institute, Beijing, 100142, China
| | - Ke Hu
- Department of Radiation Oncology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Fengwei Wang
- Department of Oncology, Tianjin Union Medical Center, Tianjin, China
| | - Zhi Wang
- Blot Info & Tech (Beijing) Co. Ltd, Beijing, China
| | - Zhiyong Niu
- Blot Info & Tech (Beijing) Co. Ltd, Beijing, China
| | - Jin Liu
- Blot Info & Tech (Beijing) Co. Ltd, Beijing, China
| | - Yong Cai
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Radiation Oncology, Peking University Cancer Hospital and Institute, Beijing, 100142, China
| | - Yongheng Li
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Radiation Oncology, Peking University Cancer Hospital and Institute, Beijing, 100142, China
| | - Xianggao Zhu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Radiation Oncology, Peking University Cancer Hospital and Institute, Beijing, 100142, China
| | - Jianhao Geng
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Radiation Oncology, Peking University Cancer Hospital and Institute, Beijing, 100142, China
| | - Yangzi Zhang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Radiation Oncology, Peking University Cancer Hospital and Institute, Beijing, 100142, China
| | - Huajing Teng
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Radiation Oncology, Peking University Cancer Hospital and Institute, Beijing, 100142, China
| | - Weihu Wang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Radiation Oncology, Peking University Cancer Hospital and Institute, Beijing, 100142, China.
| |
Collapse
|
5
|
O’Sullivan NJ, Temperley HC, Horan MT, Corr A, Mehigan BJ, Larkin JO, McCormick PH, Kavanagh DO, Meaney JFM, Kelly ME. Radiogenomics: Contemporary Applications in the Management of Rectal Cancer. Cancers (Basel) 2023; 15:5816. [PMID: 38136361 PMCID: PMC10741704 DOI: 10.3390/cancers15245816] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 12/05/2023] [Accepted: 12/11/2023] [Indexed: 12/24/2023] Open
Abstract
Radiogenomics, a sub-domain of radiomics, refers to the prediction of underlying tumour biology using non-invasive imaging markers. This novel technology intends to reduce the high costs, workload and invasiveness associated with traditional genetic testing via the development of 'imaging biomarkers' that have the potential to serve as an alternative 'liquid-biopsy' in the determination of tumour biological characteristics. Radiogenomics also harnesses the potential to unlock aspects of tumour biology which are not possible to assess by conventional biopsy-based methods, such as full tumour burden, intra-/inter-lesion heterogeneity and the possibility of providing the information of tumour biology longitudinally. Several studies have shown the feasibility of developing a radiogenomic-based signature to predict treatment outcomes and tumour characteristics; however, many lack prospective, external validation. We performed a systematic review of the current literature surrounding the use of radiogenomics in rectal cancer to predict underlying tumour biology.
Collapse
Affiliation(s)
- Niall J. O’Sullivan
- Department of Radiology, St. James’s Hospital, D08 NHY1 Dublin, Ireland; (M.T.H.)
- School of Medicine, Trinity College Dublin, D02 PN40 Dublin, Ireland
- The National Centre for Advanced Medical Imaging (CAMI), St. James’s Hospital, D08 NHY1 Dublin, Ireland
| | - Hugo C. Temperley
- Department of Surgery, St. James’s Hospital, D08 NHY1 Dublin, Ireland;
| | - Michelle T. Horan
- Department of Radiology, St. James’s Hospital, D08 NHY1 Dublin, Ireland; (M.T.H.)
- School of Medicine, Trinity College Dublin, D02 PN40 Dublin, Ireland
- The National Centre for Advanced Medical Imaging (CAMI), St. James’s Hospital, D08 NHY1 Dublin, Ireland
| | - Alison Corr
- Department of Radiology, St. James’s Hospital, D08 NHY1 Dublin, Ireland; (M.T.H.)
| | - Brian J. Mehigan
- School of Medicine, Trinity College Dublin, D02 PN40 Dublin, Ireland
- Department of Surgery, St. James’s Hospital, D08 NHY1 Dublin, Ireland;
| | - John O. Larkin
- School of Medicine, Trinity College Dublin, D02 PN40 Dublin, Ireland
- Department of Surgery, St. James’s Hospital, D08 NHY1 Dublin, Ireland;
| | - Paul H. McCormick
- School of Medicine, Trinity College Dublin, D02 PN40 Dublin, Ireland
- Department of Surgery, St. James’s Hospital, D08 NHY1 Dublin, Ireland;
| | - Dara O. Kavanagh
- Department of Surgery, Tallaght University Hospital, D24 NR0A Dublin, Ireland
- Department of Surgery, Royal College of Surgeons, D02 YN77 Dublin, Ireland
| | - James F. M. Meaney
- Department of Radiology, St. James’s Hospital, D08 NHY1 Dublin, Ireland; (M.T.H.)
- The National Centre for Advanced Medical Imaging (CAMI), St. James’s Hospital, D08 NHY1 Dublin, Ireland
| | - Michael E. Kelly
- School of Medicine, Trinity College Dublin, D02 PN40 Dublin, Ireland
- Department of Surgery, St. James’s Hospital, D08 NHY1 Dublin, Ireland;
- Trinity St. James’s Cancer Institute (TSJCI), D08 NHY1 Dublin, Ireland
| |
Collapse
|
6
|
Miranda J, Horvat N, Araujo-Filho JAB, Albuquerque KS, Charbel C, Trindade BMC, Cardoso DL, de Padua Gomes de Farias L, Chakraborty J, Nomura CH. The Role of Radiomics in Rectal Cancer. J Gastrointest Cancer 2023; 54:1158-1180. [PMID: 37155130 PMCID: PMC11301614 DOI: 10.1007/s12029-022-00909-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/26/2022] [Indexed: 05/10/2023]
Abstract
PURPOSE Radiomics is a promising method for advancing imaging assessment in rectal cancer. This review aims to describe the emerging role of radiomics in the imaging assessment of rectal cancer, including various applications of radiomics based on CT, MRI, or PET/CT. METHODS We conducted a literature review to highlight the progress of radiomic research to date and the challenges that need to be addressed before radiomics can be implemented clinically. RESULTS The results suggest that radiomics has the potential to provide valuable information for clinical decision-making in rectal cancer. However, there are still challenges in terms of standardization of imaging protocols, feature extraction, and validation of radiomic models. Despite these challenges, radiomics holds great promise for personalized medicine in rectal cancer, with the potential to improve diagnosis, prognosis, and treatment planning. Further research is needed to validate the clinical utility of radiomics and to establish its role in routine clinical practice. CONCLUSION Overall, radiomics has emerged as a powerful tool for improving the imaging assessment of rectal cancer, and its potential benefits should not be underestimated.
Collapse
Affiliation(s)
- Joao Miranda
- Department of Radiology, Memorial Sloan Kettering Cancer Center, 1275 York Ave, Box 29, New York, NY, 10065, USA
| | - Natally Horvat
- Department of Radiology, Memorial Sloan Kettering Cancer Center, 1275 York Ave, Box 29, New York, NY, 10065, USA.
| | - Jose A B Araujo-Filho
- Department of Radiology, Hospital Sirio-Libanes, 91 Adma Jafet, Sao Paulo, SP, 01308-050, Brazil
| | - Kamila S Albuquerque
- Department of Radiology, Hospital Beneficência Portuguesa, 637 Maestro Cardim, Sao Paulo, SP, 01323-001, Brazil
| | - Charlotte Charbel
- Department of Radiology, Memorial Sloan Kettering Cancer Center, 1275 York Ave, Box 29, New York, NY, 10065, USA
| | - Bruno M C Trindade
- Department of Radiology, University of Sao Paulo, 75 Dr. Ovídio Pires de Campos, Sao Paulo, SP, 05403-010, Brazil
| | - Daniel L Cardoso
- Department of Radiology, Hospital Sirio-Libanes, 91 Adma Jafet, Sao Paulo, SP, 01308-050, Brazil
| | | | - Jayasree Chakraborty
- Department of Surgery, Memorial Sloan Kettering Cancer Center, 1275 York Ave, New York, NY, 10065, USA
| | - Cesar Higa Nomura
- Department of Radiology, University of Sao Paulo, 75 Dr. Ovídio Pires de Campos, Sao Paulo, SP, 05403-010, Brazil
| |
Collapse
|
7
|
O'Sullivan NJ, Kelly ME. Radiomics and Radiogenomics in Pelvic Oncology: Current Applications and Future Directions. Curr Oncol 2023; 30:4936-4945. [PMID: 37232830 DOI: 10.3390/curroncol30050372] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 04/19/2023] [Accepted: 05/08/2023] [Indexed: 05/27/2023] Open
Abstract
Radiomics refers to the conversion of medical imaging into high-throughput, quantifiable data in order to analyse disease patterns, guide prognosis and aid decision making. Radiogenomics is an extension of radiomics that combines conventional radiomics techniques with molecular analysis in the form of genomic and transcriptomic data, serving as an alternative to costly, labour-intensive genetic testing. Data on radiomics and radiogenomics in the field of pelvic oncology remain novel concepts in the literature. We aim to perform an up-to-date analysis of current applications of radiomics and radiogenomics in the field of pelvic oncology, particularly focusing on the prediction of survival, recurrence and treatment response. Several studies have applied these concepts to colorectal, urological, gynaecological and sarcomatous diseases, with individual efficacy yet poor reproducibility. This article highlights the current applications of radiomics and radiogenomics in pelvic oncology, as well as the current limitations and future directions. Despite a rapid increase in publications investigating the use of radiomics and radiogenomics in pelvic oncology, the current evidence is limited by poor reproducibility and small datasets. In the era of personalised medicine, this novel field of research has significant potential, particularly for predicting prognosis and guiding therapeutic decisions. Future research may provide fundamental data on how we treat this cohort of patients, with the aim of reducing the exposure of high-risk patients to highly morbid procedures.
Collapse
Affiliation(s)
- Niall J O'Sullivan
- The Trinity St. James's Cancer Institute, D08 NHY1 Dublin, Ireland
- School of Medicine, Trinity College Dublin, D02 PN40 Dublin, Ireland
| | - Michael E Kelly
- The Trinity St. James's Cancer Institute, D08 NHY1 Dublin, Ireland
- School of Medicine, Trinity College Dublin, D02 PN40 Dublin, Ireland
| |
Collapse
|
8
|
Liu Y, Wei X, Feng X, Liu Y, Feng G, Du Y. Repeatability of radiomics studies in colorectal cancer: a systematic review. BMC Gastroenterol 2023; 23:125. [PMID: 37059990 PMCID: PMC10105401 DOI: 10.1186/s12876-023-02743-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Accepted: 03/22/2023] [Indexed: 04/16/2023] Open
Abstract
BACKGROUND Recently, radiomics has been widely used in colorectal cancer, but many variable factors affect the repeatability of radiomics research. This review aims to analyze the repeatability of radiomics studies in colorectal cancer and to evaluate the current status of radiomics in the field of colorectal cancer. METHODS The included studies in this review by searching from the PubMed and Embase databases. Then each study in our review was evaluated using the Radiomics Quality Score (RQS). We analyzed the factors that may affect the repeatability in the radiomics workflow and discussed the repeatability of the included studies. RESULTS A total of 188 studies was included in this review, of which only two (2/188, 1.06%) studies controlled the influence of individual factors. In addition, the median score of RQS was 11 (out of 36), range-1 to 27. CONCLUSIONS The RQS score was moderately low, and most studies did not consider the repeatability of radiomics features, especially in terms of Intra-individual, scanners, and scanning parameters. To improve the generalization of the radiomics model, it is necessary to further control the variable factors of repeatability.
Collapse
Affiliation(s)
- Ying Liu
- School of Medical Imaging, North Sichuan Medical College, Sichuan Province, Nanchong City, 637000, China
| | - Xiaoqin Wei
- School of Medical Imaging, North Sichuan Medical College, Sichuan Province, Nanchong City, 637000, China
| | | | - Yan Liu
- Department of Radiology, the Affiliated Hospital of North Sichuan Medical College, 1 Maoyuannan Road, Sichuan Province, 637000, Nanchong City, China
| | - Guiling Feng
- Department of Radiology, the Affiliated Hospital of North Sichuan Medical College, 1 Maoyuannan Road, Sichuan Province, 637000, Nanchong City, China
| | - Yong Du
- Department of Radiology, the Affiliated Hospital of North Sichuan Medical College, 1 Maoyuannan Road, Sichuan Province, 637000, Nanchong City, China.
| |
Collapse
|
9
|
Jia L, Wu W, Hou G, Zhao J, Qiang Y, Zhang Y, Cai M. Residual neural network with mixed loss based on batch training technique for identification of EGFR mutation status in lung cancer. MULTIMEDIA TOOLS AND APPLICATIONS 2023; 82:1-21. [PMID: 37362735 PMCID: PMC10020767 DOI: 10.1007/s11042-023-14876-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/26/2021] [Revised: 11/11/2022] [Accepted: 02/06/2023] [Indexed: 06/28/2023]
Abstract
Epidermal growth factor receptor (EGFR) is the key to targeted therapy with tyrosine kinase inhibitors in lung cancer. Traditional identification of EGFR mutation status requires biopsy and sequence testing, which may not be suitable for certain groups who cannot perform biopsy. In this paper, using easily accessible and non-invasive CT images, the residual neural network (ResNet) with mixed loss based on batch training technique is proposed for identification of EGFR mutation status in lung cancer. In this model, the ResNet is regarded as the baseline for feature extraction to avoid the gradient disappearance. Besides, a new mixed loss based on the batch similarity and the cross entropy is proposed to guide the network to better learn the model parameters. The proposed mixed loss utilizes the similarity among batch samples to evaluate the distribution of training data, which can reduce the similarity of different classes and the difference of the same classes. In the experiments, VGG16Net, DenseNet, ResNet18, ResNet34 and ResNet50 models with the mixed loss are trained on the public CT dataset with 155 patients including EGFR mutation status from TCIA. The trained networks are employed to the collected preoperative CT dataset with 56 patients from the cooperative hospital for validating the efficiency of the proposed models. Experimental results show that the proposed models are more appropriate and effective on the lung cancer dataset for identifying the EGFR mutation status. In these models, the ResNet34 with mixed loss is optimal (accuracy = 81.58%, AUC = 0.8861, sensitivity = 80.02%, specificity = 82.90%).
Collapse
Affiliation(s)
- Liye Jia
- College of Information and Computer, Taiyuan University of Technology, Taiyuan, 030600 China
| | - Wei Wu
- Department of Physiology, Shanxi Medical University, Taiyuan, 030051 China
| | - Guojie Hou
- College of Information and Computer, Taiyuan University of Technology, Taiyuan, 030600 China
| | - Juanjuan Zhao
- College of Information and Computer, Taiyuan University of Technology, Taiyuan, 030600 China
| | - Yan Qiang
- College of Information and Computer, Taiyuan University of Technology, Taiyuan, 030600 China
| | - Yanan Zhang
- College of Information and Computer, Taiyuan University of Technology, Taiyuan, 030600 China
| | - Meiling Cai
- College of Information and Computer, Taiyuan University of Technology, Taiyuan, 030600 China
| |
Collapse
|
10
|
Mohammadi M, Kaye EA, Alus O, Kee Y, Golia Pernicka JS, El Homsi M, Petkovska I, Otazo R. Accelerated Diffusion-Weighted MRI of Rectal Cancer Using a Residual Convolutional Network. Bioengineering (Basel) 2023; 10:bioengineering10030359. [PMID: 36978750 PMCID: PMC10045764 DOI: 10.3390/bioengineering10030359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 02/28/2023] [Accepted: 03/09/2023] [Indexed: 03/16/2023] Open
Abstract
This work presents a deep-learning-based denoising technique to accelerate the acquisition of high b-value diffusion-weighted MRI for rectal cancer. A denoising convolutional neural network (DCNN) with a combined L1–L2 loss function was developed to denoise high b-value diffusion-weighted MRI data acquired with fewer repetitions (NEX: number of excitations) using the low b-value image as an anatomical guide. DCNN was trained using 85 datasets acquired on patients with rectal cancer and tested on 20 different datasets with NEX = 1, 2, and 4, corresponding to acceleration factors of 16, 8, and 4, respectively. Image quality was assessed qualitatively by expert body radiologists. Reader 1 scored similar overall image quality between denoised images with NEX = 1 and NEX = 2, which were slightly lower than the reference. Reader 2 scored similar quality between NEX = 1 and the reference, while better quality for NEX = 2. Denoised images with fourfold acceleration (NEX = 4) received even higher scores than the reference, which is due in part to the effect of gas-related motion in the rectum, which affects longer acquisitions. The proposed deep learning denoising technique can enable eightfold acceleration with similar image quality (average image quality = 2.8 ± 0.5) and fourfold acceleration with higher image quality (3.0 ± 0.6) than the clinical standard (2.5 ± 0.8) for improved diagnosis of rectal cancer.
Collapse
Affiliation(s)
- Mohaddese Mohammadi
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Elena A. Kaye
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Or Alus
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Youngwook Kee
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | | | - Maria El Homsi
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Iva Petkovska
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Ricardo Otazo
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
- Correspondence:
| |
Collapse
|
11
|
Predicting gene mutation status via artificial intelligence technologies based on multimodal integration (MMI) to advance precision oncology. Semin Cancer Biol 2023; 91:1-15. [PMID: 36801447 DOI: 10.1016/j.semcancer.2023.02.006] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 01/30/2023] [Accepted: 02/15/2023] [Indexed: 02/21/2023]
Abstract
Personalized treatment strategies for cancer frequently rely on the detection of genetic alterations which are determined by molecular biology assays. Historically, these processes typically required single-gene sequencing, next-generation sequencing, or visual inspection of histopathology slides by experienced pathologists in a clinical context. In the past decade, advances in artificial intelligence (AI) technologies have demonstrated remarkable potential in assisting physicians with accurate diagnosis of oncology image-recognition tasks. Meanwhile, AI techniques make it possible to integrate multimodal data such as radiology, histology, and genomics, providing critical guidance for the stratification of patients in the context of precision therapy. Given that the mutation detection is unaffordable and time-consuming for a considerable number of patients, predicting gene mutations based on routine clinical radiological scans or whole-slide images of tissue with AI-based methods has become a hot issue in actual clinical practice. In this review, we synthesized the general framework of multimodal integration (MMI) for molecular intelligent diagnostics beyond standard techniques. Then we summarized the emerging applications of AI in the prediction of mutational and molecular profiles of common cancers (lung, brain, breast, and other tumor types) pertaining to radiology and histology imaging. Furthermore, we concluded that there truly exist multiple challenges of AI techniques in the way of its real-world application in the medical field, including data curation, feature fusion, model interpretability, and practice regulations. Despite these challenges, we still prospect the clinical implementation of AI as a highly potential decision-support tool to aid oncologists in future cancer treatment management.
Collapse
|
12
|
Pan X, Yi X, Lan M, Su X, Zhou F, Wu W. Research on the pathological mechanism of rectal adenocarcinoma based on DNA methylation. Medicine (Baltimore) 2023; 102:e32763. [PMID: 36705386 PMCID: PMC9876001 DOI: 10.1097/md.0000000000032763] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Colorectal cancer is one of the 3 most common cancers worldwide. In this study, a weighted network-based analysis method was proposed to explore the pathological mechanisms and prognostic targets of rectal adenocarcinoma (READ) at the deoxyribonucleic acid (DNA) methylation level. In this study, we downloaded clinical information and DNA methylation data from The Cancer Genome Atlas database. Differentially methylated gene analysis was used to identify the differential methylated genes in READ. Canonical correlation analysis was used to construct the weighted gene regulatory network for READ. Multilevel analysis and association analyses between gene modules and clinical information were used to mine key modules related to tumor metastasis evaluation. Genetic significance analysis was used to identify methylation sites in key modules. Finally, the importance of these methylation sites was confirmed using survival analysis. DNA methylation datasets from 90 cancer tissue samples and 6 paracancerous tissue samples were selected. A weighted gene regulatory network was constructed, and a multilevel algorithm was used to divide the gene co-expression network into 20 modules. From gene ontology enrichment analysis, characteristic M was related to biological processes such as the chemotaxis of fibroblast growth factors and the activation and regulation of immune cells etc and characteristic N was associated with the regulation of cytoskeleton formation, mainly microtubules and flagella, regulation of synapses, and regulation of cell mitosis. Based on the results of survival analysis, 7 key methylation sites were found closely correlated to the survival rate of READ, such as cg04441191 (microtubule-associated protein 4 [MAP4]), cg05658717 (KSR2), cg09622330 (GRIN2A), cg10698404 (YWHAG), cg17047993 (SPAG9), cg24504843 (CEP135), and cg24531267 (CEP250). Mutational and transcriptomic level studies revealed significant differences in DNA methylation, single nucleotide polymorphism, and transcript levels between YWHAG and MAP4 in normal tissues compared to tumor tissues, and differential expression of the 2 proteins in immunohistochemistry. Therefore, potential targeting drugs were screened for these 2 proteins for molecular docking, and artenimol was found to bind to MAP4 protein and 27-hydroxycholesterol to YWHAG. Our study found that key methylation sites played an important role in tumor metastasis and were associated with the prognosis of READ. Mutations and methylation may jointly regulate the transcription and translation of related genes, which in turn affect cancer progression. This may provide some new potential therapeutic targets and thoughts for the prognosis of READ.
Collapse
Affiliation(s)
- Xiaoqiang Pan
- Department of Neurosurgery, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Xingcheng Yi
- Laboratory of Cancer Precision Medicine, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Maozhuo Lan
- Laboratory of Cancer Precision Medicine, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Xiaoyun Su
- School of Pharmaceutical Sciences, Jilin University, Changchun, China
| | - Fang Zhou
- Equipment Division, Zhijiang People’s Hospital, Yichang, Hubei, China
| | - Wei Wu
- Department of Neurosurgery, The First Hospital of Jilin University, Changchun, Jilin, China
- * Correspondence: Wei Wu, Department of Neurosurgery, The First Hospital of Jilin University, Changchun, Jilin 130012, China (e-mail: )
| |
Collapse
|
13
|
Fernandes MC, Horvat N. Editorial for "A Deep Learning Model Based on MRI and Clinical Factors Facilitates Noninvasive Prediction of KRAS Mutation in Rectal Cancer". J Magn Reson Imaging 2022; 56:1669-1670. [PMID: 35575434 DOI: 10.1002/jmri.28233] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Accepted: 05/04/2022] [Indexed: 01/04/2023] Open
Affiliation(s)
- Maria Clara Fernandes
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Natally Horvat
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| |
Collapse
|
14
|
Mao Q, Zhou MT, Zhao ZP, Liu N, Yang L, Zhang XM. Role of radiomics in the diagnosis and treatment of gastrointestinal cancer. World J Gastroenterol 2022; 28:6002-6016. [PMID: 36405385 PMCID: PMC9669820 DOI: 10.3748/wjg.v28.i42.6002] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 09/24/2022] [Accepted: 10/27/2022] [Indexed: 11/10/2022] Open
Abstract
Gastrointestinal cancer (GIC) has high morbidity and mortality as one of the main causes of cancer death. Preoperative risk stratification is critical to guide patient management, but traditional imaging studies have difficulty predicting its biological behavior. The emerging field of radiomics allows the conversion of potential pathophysiological information in existing medical images that cannot be visually recognized into high-dimensional quantitative image features. Tumor lesion characterization, therapeutic response evaluation, and survival prediction can be achieved by analyzing the relationships between these features and clinical and genetic data. In recent years, the clinical application of radiomics to GIC has increased dramatically. In this editorial, we describe the latest progress in the application of radiomics to GIC and discuss the value of its potential clinical applications, as well as its limitations and future directions.
Collapse
Affiliation(s)
- Qi Mao
- Department of Radiology, Affiliated Hospital of North Sichuan Medical College, Nanchong 637000, Sichuan Province, China
| | - Mao-Ting Zhou
- Department of Radiology, Affiliated Hospital of North Sichuan Medical College, Nanchong 637000, Sichuan Province, China
| | - Zhang-Ping Zhao
- Department of Radiology, Panzhihua Central Hospital, Panzhihua 617000, Sichuan Province, China
| | - Ning Liu
- Department of Radiology, Affiliated Hospital of North Sichuan Medical College, Nanchong 637000, Sichuan Province, China
| | - Lin Yang
- Department of Radiology, Affiliated Hospital of North Sichuan Medical College, Nanchong 637000, Sichuan Province, China
| | - Xiao-Ming Zhang
- Department of Radiology, Affiliated Hospital of North Sichuan Medical College, Nanchong 637000, Sichuan Province, China
| |
Collapse
|
15
|
Capelli G, Campi C, Bao QR, Morra F, Lacognata C, Zucchetta P, Cecchin D, Pucciarelli S, Spolverato G, Crimì F. 18F-FDG-PET/MRI texture analysis in rectal cancer after neoadjuvant chemoradiotherapy. Nucl Med Commun 2022; 43:815-822. [PMID: 35471653 PMCID: PMC9177153 DOI: 10.1097/mnm.0000000000001570] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Accepted: 04/05/2022] [Indexed: 12/27/2022]
Abstract
OBJECTIVE Reliable markers to predict the response to neoadjuvant chemoradiotherapy (nCRT) in locally advanced rectal cancer (LARC) are lacking. We aimed to assess the ability of 18F-FDG PET/MRI to predict response to nCRT among patients undergoing curative-intent surgery. METHODS Patients with histological-confirmed LARC who underwent curative-intent surgery following nCRT and restaging with 18F-FDG PET/MRI were included. Statistical correlation between radiomic features extracted in PET, apparent diffusion coefficient (ADC) and T2w images and patients' histopathologic response to chemoradiotherapy using a multivariable logistic regression model ROC-analysis. RESULTS Overall, 50 patients were included in the study. A pathological complete response was achieved in 28.0% of patients. Considering second-order textural features, nine parameters showed a statistically significant difference between the two groups in ADC images, six parameters in PET images and four parameters in T2w images. Combining all the features selected for the three techniques in the same multivariate ROC curve analysis, we obtained an area under ROC curve of 0.863 (95% CI, 0.760-0.966), showing a sensitivity, specificity and accuracy at the Youden's index of 100% (14/14), 64% (23/36) and 74% (37/50), respectively. CONCLUSION PET/MRI texture analysis seems to represent a valuable tool in the identification of rectal cancer patients with a complete pathological response to nCRT.
Collapse
Affiliation(s)
- Giulia Capelli
- General Surgery 3, Department of Surgical, Oncological and Gastroenterological Sciences, University of Padova, Padova
| | | | - Quoc Riccardo Bao
- General Surgery 3, Department of Surgical, Oncological and Gastroenterological Sciences, University of Padova, Padova
| | - Francesco Morra
- Institute of Radiology, Department of Medicine, University of Padova
| | | | - Pietro Zucchetta
- Nuclear Medicine Unit, Department of Medicine, University of Padova, Padova, Italy
| | - Diego Cecchin
- Nuclear Medicine Unit, Department of Medicine, University of Padova, Padova, Italy
| | - Salvatore Pucciarelli
- General Surgery 3, Department of Surgical, Oncological and Gastroenterological Sciences, University of Padova, Padova
| | - Gaya Spolverato
- General Surgery 3, Department of Surgical, Oncological and Gastroenterological Sciences, University of Padova, Padova
| | - Filippo Crimì
- Institute of Radiology, Department of Medicine, University of Padova
| |
Collapse
|
16
|
Quraishi MI. Radiomics-Guided Precision Medicine Approaches for Colorectal Cancer. Front Oncol 2022; 12:872656. [PMID: 35756680 PMCID: PMC9218262 DOI: 10.3389/fonc.2022.872656] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Accepted: 05/06/2022] [Indexed: 11/13/2022] Open
Abstract
The concept of precision oncology entails molecular profiling of tumors to guide therapeutic interventions. Genomic testing through next-generation sequencing (NGS) molecular analysis provides the basis of such highly targeted therapeutics in oncology. As radiomic analysis delivers an array of structural and functional imaging-based biomarkers that depict these molecular mechanisms and correlate with key genetic alterations related to cancers. There is an opportunity to synergize these two big-data approaches to determine the molecular guidance for precision therapeutics. Colorectal cancer is one such disease whose therapeutic management is being guided by genetic and genomic analyses. We review the rationale and utility of radiomics as a combinative strategy for these approaches in the management of colorectal cancer.
Collapse
Affiliation(s)
- Mohammed I Quraishi
- Department of Radiology, University of Tennessee Medical Center, Knoxville, TN, United States
| |
Collapse
|
17
|
Wang F, Tan BF, Poh SS, Siow TR, Lim FLWT, Yip CSP, Wang MLC, Nei W, Tan HQ. Predicting outcomes for locally advanced rectal cancer treated with neoadjuvant chemoradiation with CT-based radiomics. Sci Rep 2022; 12:6167. [PMID: 35418656 PMCID: PMC9008122 DOI: 10.1038/s41598-022-10175-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2021] [Accepted: 03/31/2022] [Indexed: 12/24/2022] Open
Abstract
A feasibility study was performed to determine if CT-based radiomics could play an augmentative role in predicting neoadjuvant rectal score (NAR), locoregional failure free survival (LRFFS), distant metastasis free survival (DMFS), disease free survival (DFS) and overall survival (OS) in locally advanced rectal cancer (LARC). The NAR score, which takes into account the pathological tumour and nodal stage as well as clinical tumour stage, is a validated surrogate endpoint used for early determination of treatment response whereby a low NAR score (< 8) has been correlated with better outcomes and high NAR score (> 16) has been correlated with poorer outcomes. CT images of 191 patients with LARC were used in this study. Primary tumour (GTV) and mesorectum (CTV) were contoured separately and radiomics features were extracted from both segments. Two NAR models (NAR > 16 and NAR < 8) models were constructed using Least Absolute Shrinkage and Selection Operator (LASSO) and the survival models were constructed using regularized Cox regressions. Area under curve (AUC) and time-dependent AUC were used to quantify the performance of the LASSO and Cox regression respectively, using ten folds cross validations. The NAR > 16 and NAR < 8 models have an average AUCs of 0.68 ± 0.13 and 0.59 ± 0.14 respectively. There are statistically significant differences between the clinical and combined model for LRFFS (from 0.68 ± 0.04 to 0.72 ± 0.04), DMFS (from 0.68 ± 0.05 to 0.70 ± 0.05) and OS (from 0.64 ± 0.06 to 0.66 ± 0.06). CTV radiomics features were also found to be more important than GTV features in the NAR prediction model. The most important clinical features are age and CEA for NAR > 16 and NAR < 8 models respectively, while the most significant clinical features are age, surgical margin and NAR score across all the four survival models.
Collapse
Affiliation(s)
- Fuqiang Wang
- Division of Radiation Oncology, National Cancer Centre Singapore, Singapore, Singapore.
| | - Boon Fei Tan
- Division of Radiation Oncology, National Cancer Centre Singapore, Singapore, Singapore
| | - Sharon Shuxian Poh
- Division of Radiation Oncology, National Cancer Centre Singapore, Singapore, Singapore
| | - Tian Rui Siow
- Division of Radiation Oncology, National Cancer Centre Singapore, Singapore, Singapore
| | | | - Connie Siew Poh Yip
- Division of Radiation Oncology, National Cancer Centre Singapore, Singapore, Singapore
| | | | - Wenlong Nei
- Division of Radiation Oncology, National Cancer Centre Singapore, Singapore, Singapore
| | - Hong Qi Tan
- Division of Radiation Oncology, National Cancer Centre Singapore, Singapore, Singapore.
| |
Collapse
|
18
|
Noninvasive Method for Predicting the Expression of Ki67 and Prognosis in Non-Small-Cell Lung Cancer Patients: Radiomics. JOURNAL OF HEALTHCARE ENGINEERING 2022; 2022:7761589. [PMID: 35340222 PMCID: PMC8942651 DOI: 10.1155/2022/7761589] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 12/03/2021] [Accepted: 12/06/2021] [Indexed: 11/18/2022]
Abstract
Purpose In this study, we aimed to develop and validate a noninvasive method based on radiomics to evaluate the expression of Ki67 and prognosis of patients with non-small-cell lung cancer (NSCLC). Patients and Methods. A total of 120 patients with NSCLC were enrolled in this retrospective study. All patients were randomly assigned to a training dataset (n = 85) and test dataset (n = 35). According to the preprocessed F-FDG PET/CT image of each patient, a total of 384 radiomics features were extracted from the segmentation of regions of interest (ROIs). The Spearman correlation test and least absolute shrinkage and selection operator (LASSO), after normalization on the features matrix, were applied to reduce the dimensionality of the features. Furthermore, multivariable logistic regression analysis was used to propose a model for predicting Ki67. The survival curve was used to explore the prognostic significance of radiomics features. Results A total of 62 Ki67 positive patients and 58 Ki67 negative patients formed the training set and test training dataset and test dataset. Radiomics signatures showed good performance in predicting the expression of Ki67 with AUCs of 0.86 (training dataset) and 0.85 (test dataset). Validation and calibration showed that the radiomics had a strong predictive power in patients with NSCLC survival, which was significantly close to the effect of Ki67 expression on the survival of patients with NSCLC. Conclusion Radiomics signatures based on preoperative F-FDG PET/CT could distinguish the expression of Ki67, which also had a strong predictive performance for the survival outcome.
Collapse
|
19
|
Fernandes MC, Gollub MJ, Brown G. The importance of MRI for rectal cancer evaluation. Surg Oncol 2022; 43:101739. [PMID: 35339339 PMCID: PMC9464708 DOI: 10.1016/j.suronc.2022.101739] [Citation(s) in RCA: 64] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Accepted: 02/20/2022] [Indexed: 12/19/2022]
Abstract
Magnetic resonance imaging (MRI) has gained increasing importance in the management of rectal cancer over the last two decades. The role of MRI in patients with rectal cancer has expanded beyond the tumor-node-metastasis (TNM) system in both staging and restaging scenarios and has contributed to identifying "high" and "low" risk features that can be used to tailor and personalize patient treatment; for instance, selecting the patients for neoadjuvant chemoradiation (NCRT) before the total mesorectal excision (TME) surgery based on risk of recurrence. Among those features, the status of the circumferential resection margin (CRM), extramural vascular invasion (EMVI), and tumor deposits (TD) have stood out. Moreover, MRI also has played a role in surgical planning, especially when the tumor is located in the low rectum, when the relationship between tumor and the anal canal is important to choose the best surgical approach, and in cases of locally advanced or recurrent tumors invading adjacent pelvic organs that may require more complex surgeries such as pelvic exenteration. As approaches using organ preservation emerge, including transanal local excision and "watch-and-wait", MRI may help in the patient selection for those treatments, follow up, and detection of tumor regrowth. Additionally, potential MRI-based prognostic and predictive biomarkers, such as quantitative and semi-quantitative metrics derived from functional sequences like diffusion-weighted imaging (DWI) and dynamic contrast-enhanced (DCE), and radiomics, are under investigation. This review provides an overview of the current role of MRI in rectal cancer in staging and restaging and highlights the main areas under investigation and future perspectives.
Collapse
|
20
|
The application of radiomics in predicting gene mutations in cancer. Eur Radiol 2022; 32:4014-4024. [DOI: 10.1007/s00330-021-08520-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Revised: 12/11/2021] [Accepted: 12/14/2021] [Indexed: 12/24/2022]
|
21
|
Stanzione A, Verde F, Romeo V, Boccadifuoco F, Mainenti PP, Maurea S. Radiomics and machine learning applications in rectal cancer: Current update and future perspectives. World J Gastroenterol 2021; 27:5306-5321. [PMID: 34539134 PMCID: PMC8409167 DOI: 10.3748/wjg.v27.i32.5306] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 03/13/2021] [Accepted: 07/22/2021] [Indexed: 02/06/2023] Open
Abstract
The high incidence of rectal cancer in both sexes makes it one of the most common tumors, with significant morbidity and mortality rates. To define the best treatment option and optimize patient outcome, several rectal cancer biological variables must be evaluated. Currently, medical imaging plays a crucial role in the characterization of this disease, and it often requires a multimodal approach. Magnetic resonance imaging is the first-choice imaging modality for local staging and restaging and can be used to detect high-risk prognostic factors. Computed tomography is widely adopted for the detection of distant metastases. However, conventional imaging has recognized limitations, and many rectal cancer characteristics remain assessable only after surgery and histopathology evaluation. There is a growing interest in artificial intelligence applications in medicine, and imaging is by no means an exception. The introduction of radiomics, which allows the extraction of quantitative features that reflect tumor heterogeneity, allows the mining of data in medical images and paved the way for the identification of potential new imaging biomarkers. To manage such a huge amount of data, the use of machine learning algorithms has been proposed. Indeed, without prior explicit programming, they can be employed to build prediction models to support clinical decision making. In this review, current applications and future perspectives of artificial intelligence in medical imaging of rectal cancer are presented, with an imaging modality-based approach and a keen eye on unsolved issues. The results are promising, but the road ahead for translation in clinical practice is rather long.
Collapse
Affiliation(s)
- Arnaldo Stanzione
- Department of Advanced Biomedical Sciences, University of Naples "Federico II", Naples 80131, Italy
| | - Francesco Verde
- Department of Advanced Biomedical Sciences, University of Naples "Federico II", Naples 80131, Italy
| | - Valeria Romeo
- Department of Advanced Biomedical Sciences, University of Naples "Federico II", Naples 80131, Italy
| | - Francesca Boccadifuoco
- Department of Advanced Biomedical Sciences, University of Naples "Federico II", Naples 80131, Italy
| | - Pier Paolo Mainenti
- Institute of Biostructures and Bioimaging, National Council of Research, Napoli 80131, Italy
| | - Simone Maurea
- Department of Advanced Biomedical Sciences, University of Naples "Federico II", Naples 80131, Italy
| |
Collapse
|
22
|
Lorkowski J, Kolaszyńska O, Pokorski M. Artificial Intelligence and Precision Medicine: A Perspective. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1375:1-11. [PMID: 34138457 DOI: 10.1007/5584_2021_652] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
This article aims to present how the advanced solutions of artificial intelligence and precision medicine work together to refine medical management. Multi-omics seems the most suitable approach for biological analysis of data on precision medicine and artificial intelligence. We searched PubMed and Google Scholar databases to collect pertinent articles appearing up to 5 March 2021. Genetics, oncology, radiology, and the recent coronavirus disease (COVID-19) pandemic were chosen as representative fields addressing the cross-compliance of artificial intelligence (AI) and precision medicine based on the highest number of articles, topicality, and interconnectedness of the issue. Overall, we identified and perused 1572 articles. AI is a breakthrough that takes part in shaping the Fourth Industrial Revolution in medicine and health care, changing the long-time accepted diagnostic and treatment regimens and approaches. AI-based link prediction models may be outstandingly helpful in the literature search for drug repurposing or finding new therapeutical modalities in rapidly erupting wide-scale diseases such as the recent COVID-19.
Collapse
Affiliation(s)
- Jacek Lorkowski
- Department of Orthopedics, Traumatology and Sports Medicine, Central Clinical Hospital of the Ministry of Internal Affairs and Administration, Warsaw, Poland. .,Faculty of Health Sciences, Medical University of Mazovia, Warsaw, Poland.
| | - Oliwia Kolaszyńska
- Department of Cardiology, Independent Public Regional Hospital, Szczecin, Poland
| | - Mieczysław Pokorski
- Institute of Health Sciences, Opole University, Opole, Poland.,Faculty of Health Sciences, The Jan Długosz University in Częstochowa, Częstochowa, Poland
| |
Collapse
|
23
|
Granata V, Caruso D, Grassi R, Cappabianca S, Reginelli A, Rizzati R, Masselli G, Golfieri R, Rengo M, Regge D, Lo Re G, Pradella S, Fusco R, Faggioni L, Laghi A, Miele V, Neri E, Coppola F. Structured Reporting of Rectal Cancer Staging and Restaging: A Consensus Proposal. Cancers (Basel) 2021; 13:cancers13092135. [PMID: 33925250 PMCID: PMC8125446 DOI: 10.3390/cancers13092135] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 04/22/2021] [Accepted: 04/26/2021] [Indexed: 12/23/2022] Open
Abstract
Simple Summary Structured reporting in oncologic imaging is becoming necessary and has recently been recognized by major scientific societies. Structured reports collect all Patient Clinical Data, Clinical Evaluations and relevant key findings of Rectal Cancer, both in staging and restaging, and can facilitate clinical decision-making. Abstract Background: Structured reporting (SR) in oncologic imaging is becoming necessary and has recently been recognized by major scientific societies. The aim of this study was to build MRI-based structured reports for rectal cancer (RC) staging and restaging in order to provide clinicians all critical tumor information. Materials and Methods: A panel of radiologist experts in abdominal imaging, called the members of the Italian Society of Medical and Interventional Radiology, was established. The modified Delphi process was used to build the SR and to assess the level of agreement in all sections. The Cronbach’s alpha (Cα) correlation coefficient was used to assess the internal consistency of each section and to measure the quality analysis according to the average inter-item correlation. The intraclass correlation coefficient (ICC) was also evaluated. Results: After the second Delphi round of the SR RC staging, the panelists’ single scores and sum of scores were 3.8 (range 2–4) and 169, and the SR RC restaging panelists’ single scores and sum of scores were 3.7 (range 2–4) and 148, respectively. The Cα correlation coefficient was 0.79 for SR staging and 0.81 for SR restaging. The ICCs for the SR RC staging and restaging were 0.78 (p < 0.01) and 0.82 (p < 0.01), respectively. The final SR version was built and included 53 items for RC staging and 50 items for RC restaging. Conclusions: The final version of the structured reports of MRI-based RC staging and restaging should be a helpful and promising tool for clinicians in managing cancer patients properly. Structured reports collect all Patient Clinical Data, Clinical Evaluations and relevant key findings of Rectal Cancer, both in staging and restaging, and can facilitate clinical decision-making.
Collapse
Affiliation(s)
- Vincenza Granata
- Division of Radiology, Istituto Nazionale Tumori IRCCS Fondazione Pascale—IRCCS di Napoli, 80131 Naples, Italy; (V.G.); (R.F.)
| | - Damiano Caruso
- Department of Medical-Surgical and Translational Medicine-Radiology Unit, Sapienza University of Rome, 00185 Rome, Italy; (D.C.); (M.R.); (A.L.)
| | - Roberto Grassi
- Division of Radiology, Università degli Studi della Campania Luigi Vanvitelli, 80127 Naples, Italy; (R.G.); (S.C.); (A.R.)
- SIRM Foundation, Italian Society of Medical and Interventional Radiology, 20122 Milan, Italy
| | - Salvatore Cappabianca
- Division of Radiology, Università degli Studi della Campania Luigi Vanvitelli, 80127 Naples, Italy; (R.G.); (S.C.); (A.R.)
| | - Alfonso Reginelli
- Division of Radiology, Università degli Studi della Campania Luigi Vanvitelli, 80127 Naples, Italy; (R.G.); (S.C.); (A.R.)
| | - Roberto Rizzati
- Division of Radiology, SS.ma Annunziata Hospital, Azienda USL di Ferrara, 44121 Ferrara, Italy;
| | - Gabriele Masselli
- Department of Radiological Sciences, Oncology and Pathology, Sapienza University of Rome, 00161 Rome, Italy;
| | - Rita Golfieri
- Division of Radiology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy; (R.G.); (F.C.)
| | - Marco Rengo
- Department of Medical-Surgical and Translational Medicine-Radiology Unit, Sapienza University of Rome, 00185 Rome, Italy; (D.C.); (M.R.); (A.L.)
| | - Daniele Regge
- Department of Surgical Sciences, University of Turin, 10124 Turin, Italy;
- Radiology Unit, Candiolo Cancer Institute, FPO-IRCCS, Candiolo, 10060 Turin, Italy
| | - Giuseppe Lo Re
- Section of Radiological Sciences, DIBIMED, University of Palermo, 90127 Palermo, Italy;
| | - Silvia Pradella
- Division of Radiology, Azienda Ospedaliera Universitaria Careggi, 50139 Florence, Italy; (S.P.); (V.M.)
| | - Roberta Fusco
- Division of Radiology, Istituto Nazionale Tumori IRCCS Fondazione Pascale—IRCCS di Napoli, 80131 Naples, Italy; (V.G.); (R.F.)
| | - Lorenzo Faggioni
- Department of Translational Research, University of Pisa, 56126 Pisa, Italy;
| | - Andrea Laghi
- Department of Medical-Surgical and Translational Medicine-Radiology Unit, Sapienza University of Rome, 00185 Rome, Italy; (D.C.); (M.R.); (A.L.)
| | - Vittorio Miele
- Division of Radiology, Azienda Ospedaliera Universitaria Careggi, 50139 Florence, Italy; (S.P.); (V.M.)
| | - Emanuele Neri
- SIRM Foundation, Italian Society of Medical and Interventional Radiology, 20122 Milan, Italy
- Department of Translational Research, University of Pisa, 56126 Pisa, Italy;
- Correspondence: ; Tel.: +39-050-997313 or +39-050-992913
| | - Francesca Coppola
- Division of Radiology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy; (R.G.); (F.C.)
| |
Collapse
|
24
|
Badic B, Tixier F, Cheze Le Rest C, Hatt M, Visvikis D. Radiogenomics in Colorectal Cancer. Cancers (Basel) 2021; 13:cancers13050973. [PMID: 33652647 PMCID: PMC7956421 DOI: 10.3390/cancers13050973] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2021] [Revised: 02/07/2021] [Accepted: 02/20/2021] [Indexed: 12/12/2022] Open
Abstract
Simple Summary Colorectal carcinoma is characterized by intratumoral heterogeneity that can be assessed by radiogenomics. Radiomics, high-throughput quantitative data extracted from medical imaging, combined with molecular analysis, through genomic and transcriptomic data, is expected to lead to significant advances in personalized medicine. However, a radiogenomics approach in colorectal cancer is still in its early stages and many problems remain to be solved. Here we review the progress and challenges in this field at its current stage, as well as future developments. Abstract The steady improvement of high-throughput technologies greatly facilitates the implementation of personalized precision medicine. Characterization of tumor heterogeneity through image-derived features—radiomics and genetic profile modifications—genomics, is a rapidly evolving field known as radiogenomics. Various radiogenomics studies have been dedicated to colorectal cancer so far, highlighting the potential of these approaches to enhance clinical decision-making. In this review, a general outline of colorectal radiogenomics literature is provided, discussing the current limitations and suggested further developments.
Collapse
Affiliation(s)
- Bogdan Badic
- National Institute of Health and Medical Research, LaTIM—Laboratory of Medical Information Processing (INSERM LaTIM), UMR 1101, Université Bretagne Occidentale, 29238 Brest, France; (F.T.); (C.C.L.R.); (M.H.); (D.V.)
- Correspondence: ; Tel.: +33-298-347-215
| | - Florent Tixier
- National Institute of Health and Medical Research, LaTIM—Laboratory of Medical Information Processing (INSERM LaTIM), UMR 1101, Université Bretagne Occidentale, 29238 Brest, France; (F.T.); (C.C.L.R.); (M.H.); (D.V.)
| | - Catherine Cheze Le Rest
- National Institute of Health and Medical Research, LaTIM—Laboratory of Medical Information Processing (INSERM LaTIM), UMR 1101, Université Bretagne Occidentale, 29238 Brest, France; (F.T.); (C.C.L.R.); (M.H.); (D.V.)
- Department of Nuclear Medicine, University Hospital of Poitiers, 86021 Poitiers, France
| | - Mathieu Hatt
- National Institute of Health and Medical Research, LaTIM—Laboratory of Medical Information Processing (INSERM LaTIM), UMR 1101, Université Bretagne Occidentale, 29238 Brest, France; (F.T.); (C.C.L.R.); (M.H.); (D.V.)
| | - Dimitris Visvikis
- National Institute of Health and Medical Research, LaTIM—Laboratory of Medical Information Processing (INSERM LaTIM), UMR 1101, Université Bretagne Occidentale, 29238 Brest, France; (F.T.); (C.C.L.R.); (M.H.); (D.V.)
| |
Collapse
|
25
|
Shui L, Ren H, Yang X, Li J, Chen Z, Yi C, Zhu H, Shui P. The Era of Radiogenomics in Precision Medicine: An Emerging Approach to Support Diagnosis, Treatment Decisions, and Prognostication in Oncology. Front Oncol 2021; 10:570465. [PMID: 33575207 PMCID: PMC7870863 DOI: 10.3389/fonc.2020.570465] [Citation(s) in RCA: 65] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Accepted: 12/08/2020] [Indexed: 02/05/2023] Open
Abstract
With the rapid development of new technologies, including artificial intelligence and genome sequencing, radiogenomics has emerged as a state-of-the-art science in the field of individualized medicine. Radiogenomics combines a large volume of quantitative data extracted from medical images with individual genomic phenotypes and constructs a prediction model through deep learning to stratify patients, guide therapeutic strategies, and evaluate clinical outcomes. Recent studies of various types of tumors demonstrate the predictive value of radiogenomics. And some of the issues in the radiogenomic analysis and the solutions from prior works are presented. Although the workflow criteria and international agreed guidelines for statistical methods need to be confirmed, radiogenomics represents a repeatable and cost-effective approach for the detection of continuous changes and is a promising surrogate for invasive interventions. Therefore, radiogenomics could facilitate computer-aided diagnosis, treatment, and prediction of the prognosis in patients with tumors in the routine clinical setting. Here, we summarize the integrated process of radiogenomics and introduce the crucial strategies and statistical algorithms involved in current studies.
Collapse
Affiliation(s)
- Lin Shui
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Haoyu Ren
- Department of General, Visceral and Transplantation Surgery, University Hospital, LMU Munich, Munich, Germany
| | - Xi Yang
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Jian Li
- Department of Pharmacy, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, China
| | - Ziwei Chen
- Department of Nephrology, Chengdu Integrated TCM and Western Medicine Hospital, Chengdu, China
| | - Cheng Yi
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Hong Zhu
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Pixian Shui
- School of Pharmacy, Southwest Medical University, Luzhou, China
| |
Collapse
|
26
|
Horvat N, Araujo-Filho JDAB, Assuncao-Jr AN, Machado FADM, Sims JA, Rocha CCT, Oliveira BC, Horvat JV, Maccali C, Puga ALBL, Chagas AL, Menezes MR, Cerri GG. Radiomic analysis of MRI to Predict Sustained Complete Response after Radiofrequency Ablation in Patients with Hepatocellular Carcinoma - A Pilot Study. Clinics (Sao Paulo) 2021; 76:e2888. [PMID: 34287480 PMCID: PMC8266162 DOI: 10.6061/clinics/2021/e2888] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Accepted: 05/31/2021] [Indexed: 01/04/2023] Open
Abstract
OBJECTIVES To investigate whether quantitative textural features, extracted from pretreatment MRI, can predict sustained complete response to radiofrequency ablation (RFA) in patients with hepatocellular carcinoma (HCC). METHODS In this IRB-approved study, patients were selected from a maintained six-year database of consecutive patients who underwent both pretreatment MRI imaging with a probable or definitive imaging diagnosis of HCC (LI-RADS 4 or 5) and loco-regional treatment with RFA. An experienced radiologist manually segmented the hepatic nodules in MRI arterial and equilibrium phases to obtain the volume of interest (VOI) for extraction of 107 quantitative textural features, including shape and first- and second-order features. Statistical analysis was performed to evaluate associations between textural features and complete response. RESULTS The study consisted of 34 patients with 51 treated hepatic nodules. Sustained complete response was achieved by 6 patients (4 with single nodule and 2 with multiple nodules). Of the 107 features from the arterial and equilibrium phases, 20 (18%) and 25 (23%) achieved AUC >0.7, respectively. The three best performing features were found in the equilibrium phase: Dependence Non-Uniformity Normalized and Dependence Variance (both GLDM class, with AUC of 0.78 and 0.76, respectively) and Maximum Probability (GLCM class, AUC of 0.76). CONCLUSIONS This pilot study demonstrates that a radiomic analysis of pre-treatment MRI might be useful in identifying patients with HCC who are most likely to have a sustained complete response to RFA. Second-order features (GLDM and GLCM) extracted from equilibrium phase obtained highest discriminatory performance.
Collapse
Affiliation(s)
- Natally Horvat
- Departamento de Radiologia, Hospital Sirio Libanes, Sao Paulo, SP, BR
- Departamento de Radiologia, Instituto de Radiologia (InRad), Hospital das Clinicas (HCFMUSP), Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, BR
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, USA
- *Corresponding author. E-mail:
| | | | | | - Felipe Augusto de M. Machado
- Instituto de Educacao e Pesquisa, Hospital Sirio Libanes, Sao Paulo, SP, BR
- Escola Politecnica, Universidade de Sao Paulo, Sao Paulo, SP, BR
| | - John A. Sims
- Departamento de Engenharia Biomedica, Centro de Engenharia, Modelagem e Ciencias Sociais Aplicadas, Universidade Federal do ABC (UFABC), Santo Andre, SP, BR
| | - Camila Carlos Tavares Rocha
- Departamento de Radiologia, Instituto de Radiologia (InRad), Hospital das Clinicas (HCFMUSP), Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, BR
| | | | - Joao Vicente Horvat
- Departamento de Radiologia, Hospital Sirio Libanes, Sao Paulo, SP, BR
- Departamento de Radiologia, Instituto de Radiologia (InRad), Hospital das Clinicas (HCFMUSP), Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, BR
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, USA
| | - Claudia Maccali
- Departamento de Gastroenterologia, Divisao de Gastroenterologia e Hepatologia Clinica, Hospital das Clinicas (HCFMUSP), Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, BR
| | | | - Aline Lopes Chagas
- Departamento de Gastroenterologia, Divisao de Gastroenterologia e Hepatologia Clinica, Hospital das Clinicas (HCFMUSP), Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, BR
| | - Marcos Roberto Menezes
- Departamento de Radiologia, Hospital Sirio Libanes, Sao Paulo, SP, BR
- Departamento de Radiologia, Instituto de Radiologia (InRad), Hospital das Clinicas (HCFMUSP), Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, BR
| | - Giovanni Guido Cerri
- Departamento de Radiologia, Hospital Sirio Libanes, Sao Paulo, SP, BR
- Departamento de Radiologia, Instituto de Radiologia (InRad), Hospital das Clinicas (HCFMUSP), Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, BR
| |
Collapse
|
27
|
Liu Z, Wu K, Wu B, Tang X, Yuan H, Pang H, Huang Y, Zhu X, Luo H, Qi Y. Imaging genomics for accurate diagnosis and treatment of tumors: A cutting edge overview. Biomed Pharmacother 2020; 135:111173. [PMID: 33383370 DOI: 10.1016/j.biopha.2020.111173] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2020] [Revised: 11/30/2020] [Accepted: 12/14/2020] [Indexed: 02/07/2023] Open
Abstract
Imaging genomics refers to the establishment of the connection between invasive gene expression features and non-invasive imaging features. Tumor imaging genomics can not only understand the macroscopic phenotype of tumor, but also can deeply analyze the cellular and molecular characteristics of tumor tissue. In recent years, tumor imaging genomics has been a key in the field of medicine. The incidence of cancer in China has increased significantly, which is the main reason of disease death of urban residents. With the rapid development of imaging medicine, depending on imaging genomics, many experts have made remarkable achievements in tumor screening and diagnosis, prognosis evaluation, new treatment targets and understanding of tumor biological mechanism. This review analyzes the relationship between tumor radiology and gene expression, which provides a favorable direction for clinical staging, prognosis evaluation and accurate treatment of tumors.
Collapse
Affiliation(s)
- Zhen Liu
- Southern Marine Science and Engineering Guangdong Laboratory (Zhanjiang), Zhanjiang, China; Guangdong Key Laboratory for Research and Development of Natural Drugs, The Marine Biomedical Research Institute, Guangdong Medical University, Zhanjiang, China; The Key Lab of Zhanjiang for R&D Marine Microbial Resources in the Beibu Gulf Rim, Guangdong Medical University, Zhanjiang, China; The Marine Biomedical Research Institute of Guangdong Zhanjiang, Zhanjiang, China
| | - Kefeng Wu
- Southern Marine Science and Engineering Guangdong Laboratory (Zhanjiang), Zhanjiang, China
| | - Binhua Wu
- Southern Marine Science and Engineering Guangdong Laboratory (Zhanjiang), Zhanjiang, China; Guangdong Key Laboratory for Research and Development of Natural Drugs, The Marine Biomedical Research Institute, Guangdong Medical University, Zhanjiang, China; The Key Lab of Zhanjiang for R&D Marine Microbial Resources in the Beibu Gulf Rim, Guangdong Medical University, Zhanjiang, China; The Marine Biomedical Research Institute of Guangdong Zhanjiang, Zhanjiang, China
| | - Xiaoning Tang
- Southern Marine Science and Engineering Guangdong Laboratory (Zhanjiang), Zhanjiang, China; Guangdong Key Laboratory for Research and Development of Natural Drugs, The Marine Biomedical Research Institute, Guangdong Medical University, Zhanjiang, China
| | - Huiqing Yuan
- Southern Marine Science and Engineering Guangdong Laboratory (Zhanjiang), Zhanjiang, China; Guangdong Key Laboratory for Research and Development of Natural Drugs, The Marine Biomedical Research Institute, Guangdong Medical University, Zhanjiang, China; The Key Lab of Zhanjiang for R&D Marine Microbial Resources in the Beibu Gulf Rim, Guangdong Medical University, Zhanjiang, China; The Marine Biomedical Research Institute of Guangdong Zhanjiang, Zhanjiang, China
| | - Hao Pang
- Southern Marine Science and Engineering Guangdong Laboratory (Zhanjiang), Zhanjiang, China
| | - Yongmei Huang
- Southern Marine Science and Engineering Guangdong Laboratory (Zhanjiang), Zhanjiang, China; Guangdong Key Laboratory for Research and Development of Natural Drugs, The Marine Biomedical Research Institute, Guangdong Medical University, Zhanjiang, China; The Key Lab of Zhanjiang for R&D Marine Microbial Resources in the Beibu Gulf Rim, Guangdong Medical University, Zhanjiang, China; The Marine Biomedical Research Institute of Guangdong Zhanjiang, Zhanjiang, China
| | - Xiao Zhu
- Southern Marine Science and Engineering Guangdong Laboratory (Zhanjiang), Zhanjiang, China; Guangdong Key Laboratory for Research and Development of Natural Drugs, The Marine Biomedical Research Institute, Guangdong Medical University, Zhanjiang, China; The Key Lab of Zhanjiang for R&D Marine Microbial Resources in the Beibu Gulf Rim, Guangdong Medical University, Zhanjiang, China; The Marine Biomedical Research Institute of Guangdong Zhanjiang, Zhanjiang, China.
| | - Hui Luo
- Southern Marine Science and Engineering Guangdong Laboratory (Zhanjiang), Zhanjiang, China; Guangdong Key Laboratory for Research and Development of Natural Drugs, The Marine Biomedical Research Institute, Guangdong Medical University, Zhanjiang, China; The Key Lab of Zhanjiang for R&D Marine Microbial Resources in the Beibu Gulf Rim, Guangdong Medical University, Zhanjiang, China; The Marine Biomedical Research Institute of Guangdong Zhanjiang, Zhanjiang, China.
| | - Yi Qi
- Southern Marine Science and Engineering Guangdong Laboratory (Zhanjiang), Zhanjiang, China; Guangdong Key Laboratory for Research and Development of Natural Drugs, The Marine Biomedical Research Institute, Guangdong Medical University, Zhanjiang, China; The Key Lab of Zhanjiang for R&D Marine Microbial Resources in the Beibu Gulf Rim, Guangdong Medical University, Zhanjiang, China; The Marine Biomedical Research Institute of Guangdong Zhanjiang, Zhanjiang, China.
| |
Collapse
|
28
|
Veeraraghavan H, Vargas HA, Jimenez-Sanchez A, Micco M, Mema E, Lakhman Y, Crispin-Ortuzar M, Huang EP, Levine DA, Grisham RN, Abu-Rustum N, Deasy JO, Snyder A, Miller ML, Brenton JD, Sala E. Integrated Multi-Tumor Radio-Genomic Marker of Outcomes in Patients with High Serous Ovarian Carcinoma. Cancers (Basel) 2020; 12:E3403. [PMID: 33212885 PMCID: PMC7698381 DOI: 10.3390/cancers12113403] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 11/06/2020] [Accepted: 11/11/2020] [Indexed: 02/06/2023] Open
Abstract
Purpose: Develop an integrated intra-site and inter-site radiomics-clinical-genomic marker of high grade serous ovarian cancer (HGSOC) outcomes and explore the biological basis of radiomics with respect to molecular signaling pathways and the tumor microenvironment (TME). Method: Seventy-five stage III-IV HGSOC patients from internal (N = 40) and external factors via the Cancer Imaging Archive (TCGA) (N = 35) with pre-operative contrast enhanced CT, attempted primary cytoreduction, at least two disease sites, and molecular analysis performed within TCGA were retrospectively analyzed. An intra-site and inter-site radiomics (cluDiss) measure was combined with clinical-genomic variables (iRCG) and compared against conventional (volume and number of sites) and average radiomics (N = 75) for prognosticating progression-free survival (PFS) and platinum resistance. Correlation with molecular signaling and TME derived using a single sample gene set enrichment that was measured. Results: The iRCG model had the best platinum resistance classification accuracy (AUROC of 0.78 [95% CI 0.77 to 0.80]). CluDiss was associated with PFS (HR 1.03 [95% CI: 1.01 to 1.05], p = 0.002), negatively correlated with Wnt signaling, and positively to immune TME. Conclusions: CluDiss and the iRCG prognosticated HGSOC outcomes better than conventional and average radiomic measures and could better stratify patient outcomes if validated on larger multi-center trials.
Collapse
Affiliation(s)
- Harini Veeraraghavan
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA;
| | - Herbert Alberto Vargas
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; (H.A.V.); (Y.L.); (E.S.)
| | - Alejandro Jimenez-Sanchez
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Center, Cambridge, Cambridgeshire CB2 0RE, UK; (A.J.-S.); (M.C.-O.); (M.L.M.); (J.D.B.)
| | - Maura Micco
- Radioterapia Oncologica ed Ematologica, Dipartimento Diagnostica per Immagini, Area Diagnostica per Immagini, Radiologica Diagnostica e Interventistica Generale, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Roma, Italy;
| | - Eralda Mema
- Columbia University Medical Center, New York, NY 10032, USA;
| | - Yulia Lakhman
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; (H.A.V.); (Y.L.); (E.S.)
| | - Mireia Crispin-Ortuzar
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Center, Cambridge, Cambridgeshire CB2 0RE, UK; (A.J.-S.); (M.C.-O.); (M.L.M.); (J.D.B.)
| | | | - Douglas A. Levine
- Laura and Issac Perlmutter Cancer Center, New York University Langone Health, New York, NY 10016, USA;
| | - Rachel N. Grisham
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; (R.N.G.); (A.S.)
- Department of Medicine, Weill Cornell Medical College, New York, NY 10065, USA
| | - Nadeem Abu-Rustum
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA;
| | - Joseph O. Deasy
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA;
| | - Alexandra Snyder
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; (R.N.G.); (A.S.)
- Department of Medicine, Weill Cornell Medical College, New York, NY 10065, USA
| | - Martin L. Miller
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Center, Cambridge, Cambridgeshire CB2 0RE, UK; (A.J.-S.); (M.C.-O.); (M.L.M.); (J.D.B.)
| | - James D. Brenton
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Center, Cambridge, Cambridgeshire CB2 0RE, UK; (A.J.-S.); (M.C.-O.); (M.L.M.); (J.D.B.)
| | - Evis Sala
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; (H.A.V.); (Y.L.); (E.S.)
| |
Collapse
|
29
|
Clinical utility of radiomics at baseline rectal MRI to predict complete response of rectal cancer after chemoradiation therapy. Abdom Radiol (NY) 2020; 45:3608-3617. [PMID: 32296896 DOI: 10.1007/s00261-020-02502-w] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
PURPOSE To investigate the value of T2-radiomics combined with anatomical MRI staging criteria from pre-treatment rectal MRI in predicting complete response to neoadjuvant chemoradiation therapy (CRT). METHODS This retrospective study included patients with locally advanced rectal cancer who underwent rectal MRI before neoadjuvant CRT from October 2011 to January 2015 and then surgery. Surgical histopathologic analysis was used as the reference standard for pathologic complete response. Anatomical MRI staging criteria were extracted from our institutional standardized radiology report. In radiomics analysis, one radiologist manually segmented the primary tumor on T2-weighted images for all 102 patients (i.e., training set); two different radiologists independently segmented 66/102 patients (i.e., validation set). 108 radiomics features were extracted. Then, scanner-independent features were identified and least absolute shrinkage operator analysis was used to extract a radiomics score. Finally, a support vector machine model combining the radiomics score and anatomical MRI staging criteria was compared against both anatomical MRI-only and radiomics-only models using the deLong test. RESULTS The study included 102 patients (42 women; median age = 61 years).The radiomics score produced an area under the curve (AUC) of 0.75. Comparable results were found using the validation set (AUCs = 0.75 and 0.71 for each radiologist, respectively). The anatomical MRI-only model had an accuracy of 67% (sensitivity 42%, specificity 72%); when adding the radiomics score, the accuracy increased to 74% (sensitivity 58%, specificity 77%). CONCLUSION Combining T2-radiomics and anatomical MRI staging criteria from pre-treatment rectal MRI may help to stratify patients based on the prediction of treatment response to neoadjuvant therapy.
Collapse
|
30
|
Ogunwobi OO, Mahmood F, Akingboye A. Biomarkers in Colorectal Cancer: Current Research and Future Prospects. Int J Mol Sci 2020; 21:E5311. [PMID: 32726923 PMCID: PMC7432436 DOI: 10.3390/ijms21155311] [Citation(s) in RCA: 162] [Impact Index Per Article: 32.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2020] [Revised: 07/12/2020] [Accepted: 07/20/2020] [Indexed: 12/12/2022] Open
Abstract
Colorectal cancer (CRC) is a leading cause of death worldwide, despite progress made in detection and management through surgery, chemotherapy, radiotherapy, and immunotherapy. Novel therapeutic agents have improved survival in both the adjuvant and advanced disease settings, albeit with an increased risk of toxicity and cost. However, metastatic disease continues to have a poor long-term prognosis and significant challenges remain due to late stage diagnosis and treatment failure. Biomarkers are a key tool in early detection, prognostication, survival, and predicting treatment response. The past three decades have seen advances in genomics and molecular pathology of cancer biomarkers, allowing for greater individualization of therapy with a positive impact on survival outcomes. Clinically useful predictive biomarkers aid clinical decision making, such as the presence of KRAS gene mutations predicting benefit from epidermal growth factor receptor (EGFR) inhibiting antibodies. However, few biomarkers have been translated into clinical practice highlighting the need for further investigation. We review a range of protein, DNA and RNA-based biomarkers under investigation for diagnostic, predictive, and prognostic properties for CRC. In particular, long non-coding RNAs (lncRNA), have been investigated as biomarkers in a range of cancers including colorectal cancer. Specifically, we evaluate the potential role of lncRNA plasmacytoma variant translocation 1 (PVT1), an oncogene, as a diagnostic, prognostic, and therapeutic biomarker in colorectal cancer.
Collapse
Affiliation(s)
- Olorunseun O. Ogunwobi
- Department of Biological Sciences, Hunter College of The City University of New York, New York, NY 10065, USA
- Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medicine, Cornell University, New York, NY 10021, USA
| | - Fahad Mahmood
- The Dudley Group Hospitals, Russells Hall Hospital, The Dudley Group NHS Foundation Trust, Dudley, West Midlands DY1 2HQ, UK;
| | - Akinfemi Akingboye
- The Dudley Group Hospitals, Russells Hall Hospital, The Dudley Group NHS Foundation Trust, Dudley, West Midlands DY1 2HQ, UK;
| |
Collapse
|
31
|
MRI T2-weighted sequences-based texture analysis (TA) as a predictor of response to neoadjuvant chemo-radiotherapy (nCRT) in patients with locally advanced rectal cancer (LARC). Radiol Med 2020; 125:1216-1224. [DOI: 10.1007/s11547-020-01215-w] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Accepted: 04/27/2020] [Indexed: 12/13/2022]
|
32
|
Wang J, Cui Y, Shi G, Zhao J, Yang X, Qiang Y, Du Q, Ma Y, Kazihise NGF. Multi-branch cross attention model for prediction of KRAS mutation in rectal cancer with t2-weighted MRI. APPL INTELL 2020. [DOI: 10.1007/s10489-020-01658-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
33
|
Mainenti PP, Stanzione A, Guarino S, Romeo V, Ugga L, Romano F, Storto G, Maurea S, Brunetti A. Colorectal cancer: Parametric evaluation of morphological, functional and molecular tomographic imaging. World J Gastroenterol 2019; 25:5233-5256. [PMID: 31558870 PMCID: PMC6761241 DOI: 10.3748/wjg.v25.i35.5233] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Revised: 08/06/2019] [Accepted: 08/24/2019] [Indexed: 02/06/2023] Open
Abstract
Colorectal cancer (CRC) represents one of the leading causes of tumor-related deaths worldwide. Among the various tools at physicians' disposal for the diagnostic management of the disease, tomographic imaging (e.g., CT, MRI, and hybrid PET imaging) is considered essential. The qualitative and subjective evaluation of tomographic images is the main approach used to obtain valuable clinical information, although this strategy suffers from both intrinsic and operator-dependent limitations. More recently, advanced imaging techniques have been developed with the aim of overcoming these issues. Such techniques, such as diffusion-weighted MRI and perfusion imaging, were designed for the "in vivo" evaluation of specific biological tissue features in order to describe them in terms of quantitative parameters, which could answer questions difficult to address with conventional imaging alone (e.g., questions related to tissue characterization and prognosis). Furthermore, it has been observed that a large amount of numerical and statistical information is buried inside tomographic images, resulting in their invisibility during conventional assessment. This information can be extracted and represented in terms of quantitative parameters through different processes (e.g., texture analysis). Numerous researchers have focused their work on the significance of these quantitative imaging parameters for the management of CRC patients. In this review, we aimed to focus on evidence reported in the academic literature regarding the application of parametric imaging to the diagnosis, staging and prognosis of CRC while discussing future perspectives and present limitations. While the transition from purely anatomical to quantitative tomographic imaging appears achievable for CRC diagnostics, some essential milestones, such as scanning and analysis standardization and the definition of robust cut-off values, must be achieved before quantitative tomographic imaging can be incorporated into daily clinical practice.
Collapse
Affiliation(s)
- Pier Paolo Mainenti
- Institute of Biostructures and Bioimaging of the National Council of Research (CNR), Naples 80145, Italy
| | - Arnaldo Stanzione
- University of Naples "Federico II", Department of Advanced Biomedical Sciences, Naples 80131, Italy
| | - Salvatore Guarino
- University of Naples "Federico II", Department of Advanced Biomedical Sciences, Naples 80131, Italy
| | - Valeria Romeo
- University of Naples "Federico II", Department of Advanced Biomedical Sciences, Naples 80131, Italy
| | - Lorenzo Ugga
- University of Naples "Federico II", Department of Advanced Biomedical Sciences, Naples 80131, Italy
| | - Federica Romano
- University of Naples "Federico II", Department of Advanced Biomedical Sciences, Naples 80131, Italy
| | - Giovanni Storto
- IRCCS-CROB, Referral Cancer Center of Basilicata, Rionero in Vulture 85028, Italy
| | - Simone Maurea
- University of Naples "Federico II", Department of Advanced Biomedical Sciences, Naples 80131, Italy
| | - Arturo Brunetti
- University of Naples "Federico II", Department of Advanced Biomedical Sciences, Naples 80131, Italy
| |
Collapse
|