1
|
Rai HM, Yoo J, Razaque A. Comparative analysis of machine learning and deep learning models for improved cancer detection: A comprehensive review of recent advancements in diagnostic techniques. EXPERT SYSTEMS WITH APPLICATIONS 2024; 255:124838. [DOI: 10.1016/j.eswa.2024.124838] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/11/2024]
|
2
|
Garbarino GM, Polici M, Caruso D, Laghi A, Mercantini P, Pilozzi E, van Berge Henegouwen MI, Gisbertz SS, van Grieken NCT, Berardi E, Costa G. Radiomics in Oesogastric Cancer: Staging and Prediction of Preoperative Treatment Response: A Narrative Review and the Results of Personal Experience. Cancers (Basel) 2024; 16:2664. [PMID: 39123392 PMCID: PMC11311587 DOI: 10.3390/cancers16152664] [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: 07/01/2024] [Revised: 07/20/2024] [Accepted: 07/23/2024] [Indexed: 08/12/2024] Open
Abstract
BACKGROUND Oesophageal, gastroesophageal, and gastric malignancies are often diagnosed at locally advanced stage and multimodal therapy is recommended to increase the chances of survival. However, given the significant variation in treatment response, there is a clear imperative to refine patient stratification. The aim of this narrative review was to explore the existing evidence and the potential of radiomics to improve staging and prediction of treatment response of oesogastric cancers. METHODS The references for this review article were identified via MEDLINE (PubMed) and Scopus searches with the terms "radiomics", "texture analysis", "oesophageal cancer", "gastroesophageal junction cancer", "oesophagogastric junction cancer", "gastric cancer", "stomach cancer", "staging", and "treatment response" until May 2024. RESULTS Radiomics proved to be effective in improving disease staging and prediction of treatment response for both oesophageal and gastric cancer with all imaging modalities (TC, MRI, and 18F-FDG PET/CT). The literature data on the application of radiomics to gastroesophageal junction cancer are very scarce. Radiomics models perform better when integrating different imaging modalities compared to a single radiology method and when combining clinical to radiomics features compared to only a radiomics signature. CONCLUSIONS Radiomics shows potential in noninvasive staging and predicting response to preoperative therapy among patients with locally advanced oesogastric cancer. As a future perspective, the incorporation of molecular subgroup analysis to clinical and radiomic features may even increase the effectiveness of these predictive and prognostic models.
Collapse
Affiliation(s)
| | - Michela Polici
- Department of Medical Surgical Sciences and Translational Medicine, Sapienza University of Rome, Sant’Andrea Hospital, 00189 Rome, Italy
| | - Damiano Caruso
- Department of Medical Surgical Sciences and Translational Medicine, Sapienza University of Rome, Sant’Andrea Hospital, 00189 Rome, Italy
| | - Andrea Laghi
- Department of Medical Surgical Sciences and Translational Medicine, Sapienza University of Rome, Sant’Andrea Hospital, 00189 Rome, Italy
| | - Paolo Mercantini
- Department of Medical Surgical Sciences and Translational Medicine, Sapienza University of Rome, Sant’Andrea Hospital, 00189 Rome, Italy
| | - Emanuela Pilozzi
- Department of Clinical and Molecular Medicine, Sapienza University of Rome, Sant’Andrea Hospital, 00189 Rome, Italy
| | - Mark I. van Berge Henegouwen
- Department of Surgery, Amsterdam UMC Location University of Amsterdam, 1081 HV Amsterdam, The Netherlands
- Cancer Center Amsterdam, Cancer Treatment and Quality of Life, 1081 HV Amsterdam, The Netherlands
| | - Suzanne S. Gisbertz
- Department of Surgery, Amsterdam UMC Location University of Amsterdam, 1081 HV Amsterdam, The Netherlands
- Cancer Center Amsterdam, Cancer Treatment and Quality of Life, 1081 HV Amsterdam, The Netherlands
| | - Nicole C. T. van Grieken
- Department of Pathology, Amsterdam UMC Location Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands
- Cancer Biology and Immunology, Cancer Center Amsterdam, 1081 HV Amsterdam, The Netherlands
| | - Eva Berardi
- Department of Radiology, San Camillo Hospital, ASL RM 1, 00152 Rome, Italy
| | - Gianluca Costa
- Department of Life Science, Health and Health Professions, Link Campus University, 00165 Rome, Italy
| |
Collapse
|
3
|
Liu Y, Ma Z, Bao Y, Wang X, Men Y, Sun X, Ye F, Men K, Qin J, Bi N, Xue L, Hui Z. Integrating MR radiomics and dynamic hematological factors predicts pathological response to neoadjuvant chemoradiotherapy in esophageal cancer. Heliyon 2024; 10:e33702. [PMID: 39050414 PMCID: PMC11268188 DOI: 10.1016/j.heliyon.2024.e33702] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 06/08/2024] [Accepted: 06/25/2024] [Indexed: 07/27/2024] Open
Abstract
Purpose We aimed to integrate MR radiomics and dynamic hematological factors to build a model to predict pathological complete response (pCR) to neoadjuvant chemoradiotherapy (NCRT) in esophageal squamous cell carcinoma (ESCC). Methods Patients with ESCC receiving NCRT and esophagectomy between September 2014 and September 2022 were retrospectively included. All patients underwent pre-treatment T2-weighted imaging as well as pre-treatment and post-treatment blood tests. Patients were randomly divided to training set and testing set at a ratio of 7:3. Machine learning models were constructed based on MR radiomics and hematological factors to predict pCR, respectively. A nomogram model was developed to integrate MR radiomics and hematological factors. Model performances were evaluated by areas under curves (AUCs), sensitivity, specificity, positive predictive value and negative. Results A total of 82 patients were included, of whom 39 (47.6 %) achieved pCR. The hematological model built with four hematological factors had an AUC of 0.628 (95%CI 0.391-0.852) in the testing set. Two out of 1106 extracted features were selected to build the radiomics model with an AUC of 0.821 (95%CI 0.641-0.981). The nomogram model integrating hematological factors and MR radiomics had best predictive performance, with an AUC of 0.904 (95%CI 0.770-1.000) in the testing set. Conclusion An integrated model using dynamic hematological factors and MR radiomics is constructed to accurately predicted pCR to NCRT in ESCC, which may be potentially useful to assist individualized preservation treatment of the esophagus.
Collapse
Affiliation(s)
- Yunsong Liu
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zeliang Ma
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yongxing Bao
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xin Wang
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yu Men
- Department of VIP Medical Services, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xujie Sun
- Department of Pathology, National Cancer Center/ National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Feng Ye
- Department of Diagnostic Radiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Kuo Men
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jianjun Qin
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Nan Bi
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Liyan Xue
- Department of Pathology, National Cancer Center/ National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zhouguang Hui
- Department of VIP Medical Services, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
4
|
Kawahara D, Murakami Y, Awane S, Emoto Y, Iwashita K, Kubota H, Sasaki R, Nagata Y. Radiomics and dosiomics for predicting complete response to definitive chemoradiotherapy patients with oesophageal squamous cell cancer using the hybrid institution model. Eur Radiol 2024; 34:1200-1209. [PMID: 37589902 DOI: 10.1007/s00330-023-10020-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Revised: 05/08/2023] [Accepted: 06/12/2023] [Indexed: 08/18/2023]
Abstract
OBJECTIVES To develop a multi-institutional prediction model to estimate the local response to oesophageal squamous cell carcinoma (ESCC) treated with definitive radiotherapy based on radiomics and dosiomics features. METHODS The local responses were categorised into two groups (incomplete and complete). An external validation model and a hybrid model that the patients from two institutions were mixed randomly were proposed. The ESCC patients at stages I-IV who underwent chemoradiotherapy from 2012 to 2017 and had follow-up duration of more than 5 years were included. The patients who received palliative or pre-operable radiotherapy and had no FDG PET images were excluded. The segmentations included the GTV, CTV, and PTV which are used in treatment planning. In addition, shrinkage, expansion, and shell regions were created. Radiomic and dosiomic features were extracted from CT, FDG PET images, and dose distribution. Machine learning-based prediction models were developed using decision tree, support vector machine, k-nearest neighbour (kNN) algorithm, and neural network (NN) classifiers. RESULTS A total of 116 and 26 patients enrolled at Centre 1 and Centre 2, respectively. The external validation model exhibited the highest accuracy with 65.4% for CT-based radiomics, 77.9% for PET-based radiomics, and 72.1% for dosiomics based on the NN classifiers. The hybrid model exhibited the highest accuracy of 84.4% for CT-based radiomics based on the kNN classifier, 86.0% for PET-based radiomics, and 79.0% for dosiomics based on the NN classifiers. CONCLUSION The proposed hybrid model exhibited promising predictive performance for the local response to definitive radiotherapy in ESCC patients. CLINICAL RELEVANCE STATEMENT The prediction of the complete response for oesophageal cancer patients may contribute to improving overall survival. The hybrid model has the potential to improve prediction performance than the external validation model that was conventionally proposed. KEY POINTS • Radiomics and dosiomics used to predict response in patients with oesophageal cancer receiving definitive radiotherapy. • Hybrid model with neural network classifier of PET-based radiomics improved prediction accuracy by 8.1%. • The hybrid model has the potential to improve prediction performance.
Collapse
Affiliation(s)
- Daisuke Kawahara
- Department of Radiation Oncology, Graduate School of Biomedical Health Sciences, Hiroshima University, Hiroshima, 734-8551, Japan.
| | - Yuji Murakami
- Department of Radiation Oncology, Graduate School of Biomedical Health Sciences, Hiroshima University, Hiroshima, 734-8551, Japan
| | - Shota Awane
- School of Medicine, Hiroshima University, Hiroshima, 734-8551, Japan
| | - Yuki Emoto
- Department of Radiation Oncology, Hyogo Cancer Center, 70, Kitaoji-Cho 13, Akashi-Shi, Hyogo, Japan
| | - Kazuma Iwashita
- Division of Radiation Oncology, Kobe University Graduate School of Medicine, 7-5-2 Kusunokicho, Chuouku, Kobe, Hyogo, 650-0017, Japan
| | - Hikaru Kubota
- Division of Radiation Oncology, Kobe University Graduate School of Medicine, 7-5-2 Kusunokicho, Chuouku, Kobe, Hyogo, 650-0017, Japan
| | - Ryohei Sasaki
- Division of Radiation Oncology, Kobe University Graduate School of Medicine, 7-5-2 Kusunokicho, Chuouku, Kobe, Hyogo, 650-0017, Japan
| | - Yasushi Nagata
- Department of Radiation Oncology, Graduate School of Biomedical Health Sciences, Hiroshima University, Hiroshima, 734-8551, Japan
- Hiroshima High-Precision Radiotherapy Cancer Center, Hiroshima, 732-0057, Japan
| |
Collapse
|
5
|
Rai HM, Yoo J, Atif Moqurrab S, Dashkevych S. Advancements in traditional machine learning techniques for detection and diagnosis of fatal cancer types: Comprehensive review of biomedical imaging datasets. MEASUREMENT 2024; 225:114059. [DOI: 10.1016/j.measurement.2023.114059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/11/2024]
|
6
|
Yang Z, Guan F, Bronk L, Zhao L. Multi-omics approaches for biomarker discovery in predicting the response of esophageal cancer to neoadjuvant therapy: A multidimensional perspective. Pharmacol Ther 2024; 254:108591. [PMID: 38286161 DOI: 10.1016/j.pharmthera.2024.108591] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 12/02/2023] [Accepted: 01/04/2024] [Indexed: 01/31/2024]
Abstract
Neoadjuvant chemoradiotherapy (NCRT) followed by surgery has been established as the standard treatment strategy for operable locally advanced esophageal cancer (EC). However, achieving pathologic complete response (pCR) or near pCR to NCRT is significantly associated with a considerable improvement in survival outcomes, while pCR patients may help organ preservation for patients by active surveillance to avoid planned surgery. Thus, there is an urgent need for improved biomarkers to predict EC chemoradiation response in research and clinical settings. Advances in multiple high-throughput technologies such as next-generation sequencing have facilitated the discovery of novel predictive biomarkers, specifically based on multi-omics data, including genomic/transcriptomic sequencings and proteomic/metabolomic mass spectra. The application of multi-omics data has shown the benefits in improving the understanding of underlying mechanisms of NCRT sensitivity/resistance in EC. Particularly, the prominent development of artificial intelligence (AI) has introduced a new direction in cancer research. The integration of multi-omics data has significantly advanced our knowledge of the disease and enabled the identification of valuable biomarkers for predicting treatment response from diverse dimension levels, especially with rapid advances in biotechnological and AI methodologies. Herein, we summarize the current status of research on the use of multi-omics technologies in predicting NCRT response for EC patients. Current limitations, challenges, and future perspectives of these multi-omics platforms will be addressed to assist in experimental designs and clinical use for further integrated analysis.
Collapse
Affiliation(s)
- Zhi Yang
- Department of Radiation Oncology, Xijing Hospital, Fourth Military Medical University, 15 West Changle Road, Xi'an, China
| | - Fada Guan
- Department of Therapeutic Radiology, Yale University School of Medicine, New Haven, CT 06510, United States of America
| | - Lawrence Bronk
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, United States of America
| | - Lina Zhao
- Department of Radiation Oncology, Xijing Hospital, Fourth Military Medical University, 15 West Changle Road, Xi'an, China.
| |
Collapse
|
7
|
Liu Y, Wang Y, Wang X, Xue L, Zhang H, Ma Z, Deng H, Yang Z, Sun X, Men Y, Ye F, Men K, Qin J, Bi N, Wang Q, Hui Z. MR radiomics predicts pathological complete response of esophageal squamous cell carcinoma after neoadjuvant chemoradiotherapy: a multicenter study. Cancer Imaging 2024; 24:16. [PMID: 38263134 PMCID: PMC10804642 DOI: 10.1186/s40644-024-00659-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2023] [Accepted: 01/10/2024] [Indexed: 01/25/2024] Open
Abstract
BACKGROUND More than 40% of patients with resectable esophageal squamous cell cancer (ESCC) achieve pathological complete response (pCR) after neoadjuvant chemoradiotherapy (nCRT), who have favorable prognosis and may benefit from an organ-preservation strategy. Our study aims to develop and validate a machine learning model based on MR radiomics to accurately predict the pCR of ESCC patients after nCRT. METHODS In this retrospective multicenter study, eligible patients with ESCC who underwent baseline MR (T2-weighted imaging) and nCRT plus surgery were enrolled between September 2014 and September 2022 at institution 1 (training set) and between December 2017 and August 2021 at institution 2 (testing set). Models were constructed using machine learning algorithms based on clinical factors and MR radiomics to predict pCR after nCRT. The area under the curve (AUC) and cutoff analysis were used to evaluate model performance. RESULTS A total of 155 patients were enrolled in this study, 82 in the training set and 73 in the testing set. The radiomics model was constructed based on two radiomics features, achieving AUCs of 0.968 (95%CI 0.933-0.992) in the training set and 0.885 (95%CI 0.800-0.958) in the testing set. The cutoff analysis resulted in an accuracy of 82.2% (95%CI 72.6-90.4%), a sensitivity of 75.0% (95%CI 58.3-91.7%), and a specificity of 85.7% (95%CI 75.5-96.0%) in the testing set. CONCLUSION A machine learning model based on MR radiomics was developed and validated to accurately predict pCR after nCRT in patients with ESCC.
Collapse
Affiliation(s)
- Yunsong Liu
- Department of Radiation Oncology, National Clinical Research Center for Cancer/Cancer Hospital, National Cancer Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Panjiayuan Nanli #17, Chaoyang District, Beijing, China
| | - Yi Wang
- Department of Radiation Oncology, Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, No.55.Section 4, South Renmin Road, Chengdu, 610042, China
| | - Xin Wang
- Department of Radiation Oncology, National Clinical Research Center for Cancer/Cancer Hospital, National Cancer Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Panjiayuan Nanli #17, Chaoyang District, Beijing, China
| | - Liyan Xue
- Department of Pathology, National Clinical Research Center for Cancer/Cancer Hospital, National Cancer Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Panjiayuan Nanli #17, Chaoyang District, Beijing, China
| | - Huan Zhang
- Department of Radiation Oncology, Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, No.55.Section 4, South Renmin Road, Chengdu, 610042, China
| | - Zeliang Ma
- Department of Radiation Oncology, National Clinical Research Center for Cancer/Cancer Hospital, National Cancer Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Panjiayuan Nanli #17, Chaoyang District, Beijing, China
| | - Heping Deng
- Department of Diagnostic Radiology, Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, No.55.Section 4, South Renmin Road, Chengdu, China
| | - Zhaoyang Yang
- Department of Pathology, National Clinical Research Center for Cancer/Cancer Hospital, National Cancer Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Panjiayuan Nanli #17, Chaoyang District, Beijing, China
| | - Xujie Sun
- Department of Pathology, National Clinical Research Center for Cancer/Cancer Hospital, National Cancer Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Panjiayuan Nanli #17, Chaoyang District, Beijing, China
| | - Yu Men
- Department of VIP Medical Services & Radiation Oncology, National Clinical Research Center for Cancer/Cancer Hospital, National Cancer Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Panjiayuan Nanli #17, Chaoyang District, Beijing, 100021, China
| | - Feng Ye
- Department of Diagnostic Radiology, National Clinical Research Center for Cancer/Cancer Hospital, National Cancer Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Panjiayuan Nanli #17, Chaoyang District, Beijing, China
| | - Kuo Men
- Department of Radiation Oncology, National Clinical Research Center for Cancer/Cancer Hospital, National Cancer Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Panjiayuan Nanli #17, Chaoyang District, Beijing, China
| | - Jianjun Qin
- Department of Thoracic Surgery, National Clinical Research Center for Cancer/Cancer Hospital, National Cancer Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Panjiayuan Nanli #17, Chaoyang District, Beijing, China
| | - Nan Bi
- Department of Radiation Oncology, National Clinical Research Center for Cancer/Cancer Hospital, National Cancer Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Panjiayuan Nanli #17, Chaoyang District, Beijing, China
| | - Qifeng Wang
- Department of Radiation Oncology, Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, No.55.Section 4, South Renmin Road, Chengdu, 610042, China.
| | - Zhouguang Hui
- Department of VIP Medical Services & Radiation Oncology, National Clinical Research Center for Cancer/Cancer Hospital, National Cancer Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Panjiayuan Nanli #17, Chaoyang District, Beijing, 100021, China.
| |
Collapse
|
8
|
Lu S, Wang C, Liu Y, Chu F, Jia Z, Zhang H, Wang Z, Lu Y, Wang S, Yang G, Qu J. The MRI radiomics signature can predict the pathologic response to neoadjuvant chemotherapy in locally advanced esophageal squamous cell carcinoma. Eur Radiol 2024; 34:485-494. [PMID: 37540319 DOI: 10.1007/s00330-023-10040-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 05/26/2023] [Accepted: 06/19/2023] [Indexed: 08/05/2023]
Abstract
OBJECTIVES To investigate the MRI radiomics signatures in predicting pathologic response among patients with locally advanced esophageal squamous cell carcinoma (ESCC), who received neoadjuvant chemotherapy (NACT). METHODS Patients who underwent NACT from March 2015 to October 2019 were prospectively included. Each patient underwent esophageal MR scanning within one week before NACT and within 2-3 weeks after completion of NACT, prior to surgery. Radiomics features extracted from T2-TSE-BLADE were randomly split into the training and validation sets at a ratio of 7:3. According to the progressive tumor regression grade (TRG), patients were stratified into two groups: good responders (GR, TRG 0 + 1) and poor responders (non-GR, TRG 2 + 3). We constructed the Pre/Post-NACT model (Pre/Post-model) and the Delta-NACT model (Delta-model). Kruskal-Wallis was used to select features, logistic regression was used to develop the final model. RESULTS A total of 108 ESCC patients were included, and 3/2/4 out of 107 radiomics features were selected for constructing the Pre/Post/Delta-model, respectively. The selected radiomics features were statistically different between GR and non-GR groups. The highest area under the curve (AUC) was for the Delta-model, which reached 0.851 in the training set and 0.831 in the validation set. Among the three models, Pre-model showed the poorest performance in the training and validation sets (AUC, 0.466 and 0.596), and the Post-model showed better performance than the Pre-model in the training and validation sets (AUC, 0.753 and 0.781). CONCLUSIONS MRI-based radiomics models can predict the pathological response after NACT in ESCC patients, with the Delta-model exhibiting optimal predictive efficacy. CLINICAL RELEVANCE STATEMENT MRI radiomics features could be used as a useful tool for predicting the efficacy of neoadjuvant chemotherapy in esophageal carcinoma patients, especially in selecting responders among those patients who may be candidates to benefit from neoadjuvant chemotherapy. KEY POINTS • The MRI radiomics features based on T2WI-TSE-BLADE could potentially predict the pathologic response to NACT among ESCC patients. • The Delta-model exhibited the best predictive ability for pathologic response, followed by the Post-model, which similarly had better predictive ability, while the Pre-model performed less well in predicting TRG.
Collapse
Affiliation(s)
- Shuang Lu
- Department of Radiology, the Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, No. 127 Dongming Road, Zhengzhou, 450008, Henan, China
| | - Chenglong Wang
- Shanghai Key Laboratory of Magnetic Resonance, East China Normal University, Shanghai, 200062, China
| | - Yun Liu
- Shanghai Key Laboratory of Magnetic Resonance, East China Normal University, Shanghai, 200062, China
| | - Funing Chu
- Department of Radiology, the Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, No. 127 Dongming Road, Zhengzhou, 450008, Henan, China
| | - Zhengyan Jia
- Department of Radiology, the Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, No. 127 Dongming Road, Zhengzhou, 450008, Henan, China
| | - Hongkai Zhang
- Department of Radiology, the Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, No. 127 Dongming Road, Zhengzhou, 450008, Henan, China
| | - Zhaoqi Wang
- Department of Radiology, the Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, No. 127 Dongming Road, Zhengzhou, 450008, Henan, China
| | - Yanan Lu
- Department of Radiology, the Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, No. 127 Dongming Road, Zhengzhou, 450008, Henan, China
| | - Shuting Wang
- Department of Radiology, the Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, No. 127 Dongming Road, Zhengzhou, 450008, Henan, China
| | - Guang Yang
- Shanghai Key Laboratory of Magnetic Resonance, East China Normal University, Shanghai, 200062, China.
| | - Jinrong Qu
- Department of Radiology, the Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, No. 127 Dongming Road, Zhengzhou, 450008, Henan, China.
| |
Collapse
|
9
|
Yap WK, Hsiao IT, Yap WL, Tsai TY, Lu YA, Yang CK, Peng MT, Su EL, Cheng SC. A Radiotherapy Dose Map-Guided Deep Learning Method for Predicting Pathological Complete Response in Esophageal Cancer Patients after Neoadjuvant Chemoradiotherapy Followed by Surgery. Biomedicines 2023; 11:3072. [PMID: 38002072 PMCID: PMC10669191 DOI: 10.3390/biomedicines11113072] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 10/28/2023] [Accepted: 11/03/2023] [Indexed: 11/26/2023] Open
Abstract
Esophageal cancer is a deadly disease, and neoadjuvant chemoradiotherapy can improve patient survival, particularly for patients achieving a pathological complete response (ypCR). However, existing imaging methods struggle to accurately predict ypCR. This study explores computer-aided detection methods, considering both imaging data and radiotherapy dose variations to enhance prediction accuracy. It involved patients with node-positive esophageal squamous cell carcinoma undergoing neoadjuvant chemoradiotherapy and surgery, with data collected from 2014 to 2017, randomly split into five subsets for 5-fold cross-validation. The algorithm DCRNet, an advanced version of OCRNet, integrates RT dose distribution into dose contextual representations (DCR), combining dose and pixel representation with ten soft regions. Among the 80 enrolled patients (mean age 55.68 years, primarily male, with stage III disease and middle-part lesions), the ypCR rate was 28.75%, showing no significant demographic or disease differences between the ypCR and non-ypCR groups. Among the three summarization methods, the maximum value across the CTV method produced the best results with an AUC of 0.928. The HRNetV2p model with DCR performed the best among the four backbone models tested, with an AUC of 0.928 (95% CI, 0.884-0.972) based on 5-fold cross-validation, showing significant improvement compared to other models. This underscores DCR-equipped models' superior AUC outcomes. The study highlights the potential of dose-guided deep learning in ypCR prediction, necessitating larger, multicenter studies to validate the results.
Collapse
Affiliation(s)
- Wing-Keen Yap
- Department of Radiation Oncology, Proton and Radiation Therapy Center, Chang Gung Memorial Hospital—Linkou Medical Center, 5 Fu-Shin Street, Kwei-Shan, Taoyuan 333, Taiwan
| | - Ing-Tsung Hsiao
- Department of Medical Imaging and Radiological Sciences, Healthy Aging Research Center, Chang Gung University, Taoyuan 333, Taiwan
| | - Wing-Lake Yap
- Department of Post Baccalaureate Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Tsung-You Tsai
- Department of Otolaryngology—Head and Neck Surgery, Chang Gung Memorial Hospital—Linkou Medical Center, 5 Fu-Shin Street, Kwei-Shan, Taoyuan 333, Taiwan
| | - Yi-An Lu
- Department of Otolaryngology—Head and Neck Surgery, Chang Gung Memorial Hospital—Linkou Medical Center, 5 Fu-Shin Street, Kwei-Shan, Taoyuan 333, Taiwan
| | - Chan-Keng Yang
- Division of Hematology and Oncology, Department of Internal Medicine, Chang Gung Memorial Hospital at Linkou Branch, Chang Gung University College of Medicine, Taoyuan 333, Taiwan
| | - Meng-Ting Peng
- Division of Hematology and Oncology, Department of Internal Medicine, Chang Gung Memorial Hospital at Linkou Branch, Chang Gung University College of Medicine, Taoyuan 333, Taiwan
| | - En-Lin Su
- Department of School of Medicine, National Yang Ming Chiao Tung University, Taipei 112, Taiwan
| | | |
Collapse
|
10
|
Yang Z, Gong J, Li J, Sun H, Pan Y, Zhao L. The gap before real clinical application of imaging-based machine-learning and radiomic models for chemoradiation outcome prediction in esophageal cancer: a systematic review and meta-analysis. Int J Surg 2023; 109:2451-2466. [PMID: 37463039 PMCID: PMC10442126 DOI: 10.1097/js9.0000000000000441] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Accepted: 05/01/2023] [Indexed: 08/21/2023]
Abstract
BACKGROUND Due to tumoral heterogeneity and the lack of robust biomarkers, the prediction of chemoradiotherapy response and prognosis in patients with esophageal cancer (EC) is challenging. The goal of this study was to assess the study quality and clinical value of machine learning and radiomic-based quantitative imaging studies for predicting the outcomes of EC patients after chemoradiotherapy. MATERIALS AND METHODS PubMed, Embase, and Cochrane were searched for eligible articles. The methodological quality and risk of bias were evaluated using the Radiomics Quality Score (RQS), Image Biomarkers Standardization Initiative (IBSI) Guideline, and Transparent Reporting of a multivariable prediction model for Individual Prognosis or Diagnosis (TRIPOD) statement, as well as the modified Quality Assessment of Diagnostic Accuracy Studies (QUADAS-2) tool. A meta-analysis of the evidence focusing on predicting chemoradiotherapy response and outcome in EC patients was implemented. RESULTS Forty-six studies were eligible for qualitative synthesis. The mean RQS score was 9.07, with an adherence rate of 42.52%. The adherence rates of the TRIPOD and IBSI were 61.70 and 43.17%, respectively. Ultimately, 24 studies were included in the meta-analysis, of which 16 studies had a pooled sensitivity, specificity, and area under the curve (AUC) of 0.83 (0.76-0.89), 0.83 (0.79-0.86), and 0.84 (0.81-0.87) in neoadjuvant chemoradiotherapy datasets, as well as 0.84 (0.75-0.93), 0.89 (0.83-0.93), and 0.93 (0.90-0.95) in definitive chemoradiotherapy datasets, respectively. Moreover, radiomics could distinguish patients from the low-risk and high-risk groups with different disease-free survival (DFS) (pooled hazard ratio: 3.43, 95% CI 2.39-4.92) and overall survival (pooled hazard ratio: 2.49, 95% CI 1.91-3.25). The results of subgroup and regression analyses showed that some of the heterogeneity was explained by the combination with clinical factors, sample size, and usage of the deep learning (DL) signature. CONCLUSIONS Noninvasive radiomics offers promising potential for optimizing treatment decision-making in EC patients. However, it is necessary to make scientific advancements in EC radiomics regarding reproducibility, clinical usefulness analysis, and open science categories. Improved model reporting of study objectives, blind assessment, and image processing steps are required to help promote real clinical applications of radiomics in EC research.
Collapse
Affiliation(s)
- Zhi Yang
- Department of Radiation Oncology, Xijing Hospital
| | - Jie Gong
- Department of Radiation Oncology, Xijing Hospital
| | - Jie Li
- Department of Radiation Oncology, Xijing Hospital
| | - Hongfei Sun
- Department of Radiation Oncology, Xijing Hospital
| | - Yanglin Pan
- State Key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases and Xijing Hospital of Digestive Diseases, Air Force Medical University, Xi’an, People’s Republic of China
| | - Lina Zhao
- Department of Radiation Oncology, Xijing Hospital
| |
Collapse
|
11
|
Zhang YP, Zhang XY, Cheng YT, Li B, Teng XZ, Zhang J, Lam S, Zhou T, Ma ZR, Sheng JB, Tam VCW, Lee SWY, Ge H, Cai J. Artificial intelligence-driven radiomics study in cancer: the role of feature engineering and modeling. Mil Med Res 2023; 10:22. [PMID: 37189155 DOI: 10.1186/s40779-023-00458-8] [Citation(s) in RCA: 43] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Accepted: 05/04/2023] [Indexed: 05/17/2023] Open
Abstract
Modern medicine is reliant on various medical imaging technologies for non-invasively observing patients' anatomy. However, the interpretation of medical images can be highly subjective and dependent on the expertise of clinicians. Moreover, some potentially useful quantitative information in medical images, especially that which is not visible to the naked eye, is often ignored during clinical practice. In contrast, radiomics performs high-throughput feature extraction from medical images, which enables quantitative analysis of medical images and prediction of various clinical endpoints. Studies have reported that radiomics exhibits promising performance in diagnosis and predicting treatment responses and prognosis, demonstrating its potential to be a non-invasive auxiliary tool for personalized medicine. However, radiomics remains in a developmental phase as numerous technical challenges have yet to be solved, especially in feature engineering and statistical modeling. In this review, we introduce the current utility of radiomics by summarizing research on its application in the diagnosis, prognosis, and prediction of treatment responses in patients with cancer. We focus on machine learning approaches, for feature extraction and selection during feature engineering and for imbalanced datasets and multi-modality fusion during statistical modeling. Furthermore, we introduce the stability, reproducibility, and interpretability of features, and the generalizability and interpretability of models. Finally, we offer possible solutions to current challenges in radiomics research.
Collapse
Affiliation(s)
- Yuan-Peng Zhang
- Department of Medical Informatics, Nantong University, Nantong, 226001, Jiangsu, China
- Department of Health Technology and Informatics, the Hong Kong Polytechnic University, Hong Kong, 999077, China
- The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen, 518000, Guangdong, China
| | - Xin-Yun Zhang
- Department of Medical Informatics, Nantong University, Nantong, 226001, Jiangsu, China
| | - Yu-Ting Cheng
- Department of Medical Informatics, Nantong University, Nantong, 226001, Jiangsu, China
| | - Bing Li
- Department of Radiation Oncology, the Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, 450008, Henan, China
| | - Xin-Zhi Teng
- Department of Health Technology and Informatics, the Hong Kong Polytechnic University, Hong Kong, 999077, China
| | - Jiang Zhang
- Department of Health Technology and Informatics, the Hong Kong Polytechnic University, Hong Kong, 999077, China
| | - Saikit Lam
- Department of Health Technology and Informatics, the Hong Kong Polytechnic University, Hong Kong, 999077, China
| | - Ta Zhou
- Department of Health Technology and Informatics, the Hong Kong Polytechnic University, Hong Kong, 999077, China
| | - Zong-Rui Ma
- Department of Health Technology and Informatics, the Hong Kong Polytechnic University, Hong Kong, 999077, China
| | - Jia-Bao Sheng
- Department of Health Technology and Informatics, the Hong Kong Polytechnic University, Hong Kong, 999077, China
| | - Victor C W Tam
- Department of Health Technology and Informatics, the Hong Kong Polytechnic University, Hong Kong, 999077, China
| | - Shara W Y Lee
- Department of Health Technology and Informatics, the Hong Kong Polytechnic University, Hong Kong, 999077, China
| | - Hong Ge
- Department of Radiation Oncology, the Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, 450008, Henan, China
| | - Jing Cai
- Department of Health Technology and Informatics, the Hong Kong Polytechnic University, Hong Kong, 999077, China.
- The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen, 518000, Guangdong, China.
| |
Collapse
|
12
|
Liu J, Yang X, Mao X, Wang T, Zheng X, Feng G, Dai T, Du X. Predicting the efficacy of radiotherapy for esophageal squamous cell carcinoma based on enhanced computed tomography radiomics and combined models. Front Oncol 2023; 13:1089365. [PMID: 37007134 PMCID: PMC10061127 DOI: 10.3389/fonc.2023.1089365] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Accepted: 02/24/2023] [Indexed: 03/18/2023] Open
Abstract
PurposeThis study aimed to investigate the ability of enhanced computed tomography (CT)-based radiomics and dosimetric parameters in predicting response to radiotherapy for esophageal cancer.MethodsA retrospective analysis of 147 patients diagnosed with esophageal cancer was performed, and the patients were divided into a training group (104 patients) and a validation group (43 patients). In total, 851 radiomics features were extracted from the primary lesions for analysis. Maximum correlation minimum redundancy and minimum least absolute shrinkage and selection operator were utilized for feature screening of radiomics features, and logistic regression was applied to construct a radiotherapy radiomics model for esophageal cancer. Finally, univariate and multivariate parameters were used to identify significant clinical and dosimetric characteristics for constructing combination models. The area evaluated the predictive performance under the receiver operating characteristics (AUC) curve and the accuracy, sensitivity, and specificity of the training and validation cohorts.ResultsUnivariate logistic regression analysis revealed statistically significant differences in clinical parameters of sex (p=0.031) and esophageal cancer thickness (p=0.028) on treatment response, whereas dosimetric parameters did not differ significantly in response to treatment. The combined model demonstrated improved discrimination between the training and validation groups, with AUCs of 0.78 (95% confidence interval [CI], 0.69–0.87) and 0.79 (95% CI, 0.65–0.93) in the training and validation groups, respectively.ConclusionThe combined model has potential application value in predicting the treatment response of patients with esophageal cancer after radiotherapy.
Collapse
|
13
|
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
|
14
|
Korte JC, Cardenas C, Hardcastle N, Kron T, Wang J, Bahig H, Elgohari B, Ger R, Court L, Fuller CD, Ng SP. Radiomics feature stability of open-source software evaluated on apparent diffusion coefficient maps in head and neck cancer. Sci Rep 2021; 11:17633. [PMID: 34480036 PMCID: PMC8417253 DOI: 10.1038/s41598-021-96600-4] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Accepted: 08/10/2021] [Indexed: 02/07/2023] Open
Abstract
Radiomics is a promising technique for discovering image based biomarkers of therapy response in cancer. Reproducibility of radiomics features is a known issue that is addressed by the image biomarker standardisation initiative (IBSI), but it remains challenging to interpret previously published radiomics signatures. This study investigates the reproducibility of radiomics features calculated with two widely used radiomics software packages (IBEX, MaZda) in comparison to an IBSI compliant software package (PyRadiomics). Intensity histogram, shape and textural features were extracted from 334 diffusion weighted magnetic resonance images of 59 head and neck cancer (HNC) patients from the PREDICT-HN observational radiotherapy study. Based on name and linear correlation, PyRadiomics shares 83 features with IBEX and 49 features with MaZda, a sub-set of well correlated features are considered reproducible (IBEX: 15 features, MaZda: 18 features). We explore the impact of including non-reproducible radiomics features in a HNC radiotherapy response model. It is possible to classify equivalent patient groups using radiomic features from either software, but only when restricting the model to reliable features using a correlation threshold method. This is relevant for clinical biomarker validation trials as it provides a framework to assess the reproducibility of reported radiomic signatures from existing trials.
Collapse
Affiliation(s)
- James C. Korte
- grid.1055.10000000403978434Department of Physical Science, Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne, VIC 3000 Australia ,grid.1008.90000 0001 2179 088XDepartment of Biomedical Engineering, University of Melbourne, Melbourne, Australia
| | - Carlos Cardenas
- grid.240145.60000 0001 2291 4776Department of Radiation Physics, University of Texas MD Anderson Cancer Center, Houston, USA
| | - Nicholas Hardcastle
- grid.1055.10000000403978434Department of Physical Science, Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne, VIC 3000 Australia ,grid.1007.60000 0004 0486 528XCentre for Medical Radiation Physics, University of Wollongong, Wollongong, Australia
| | - Tomas Kron
- grid.1055.10000000403978434Department of Physical Science, Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne, VIC 3000 Australia ,grid.1008.90000 0001 2179 088XSir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Australia
| | - Jihong Wang
- grid.240145.60000 0001 2291 4776Department of Radiation Physics, University of Texas MD Anderson Cancer Center, Houston, USA
| | - Houda Bahig
- grid.410559.c0000 0001 0743 2111Radiation Oncology Department, Centre Hospitalier de l’Université de Montréal, Montreal, Canada
| | - Baher Elgohari
- grid.240145.60000 0001 2291 4776Department of Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, USA ,grid.10251.370000000103426662Clinical Oncology & Nuclear Medicine Department, Mansoura University, Mansoura, Egypt
| | - Rachel Ger
- grid.470142.40000 0004 0443 9766Department of Radiation Oncology, Mayo Clinic, Phoenix, AZ USA
| | - Laurence Court
- grid.240145.60000 0001 2291 4776Department of Radiation Physics, University of Texas MD Anderson Cancer Center, Houston, USA
| | - Clifton D. Fuller
- grid.240145.60000 0001 2291 4776Department of Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, USA
| | - Sweet Ping Ng
- grid.240145.60000 0001 2291 4776Department of Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, USA ,grid.1055.10000000403978434Department of Radiation Oncology, Peter MacCallum Cancer Centre, Melbourne, Australia ,grid.482637.cDepartment of Radiation Oncology, Olivia Newton-John Cancer Wellness and Research Centre, Melbourne, Australia
| |
Collapse
|
15
|
deSouza NM. Imaging to assist fertility-sparing surgery. Best Pract Res Clin Obstet Gynaecol 2021; 75:23-36. [PMID: 33722497 DOI: 10.1016/j.bpobgyn.2021.01.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2020] [Accepted: 01/31/2021] [Indexed: 11/23/2022]
Abstract
Cytological screening and human papilloma virus testing has led to diagnosis of cervical cancer in young women at an earlier stage. Defining the full extent of the disease within the cervix with imaging aids the decision on feasibility of fertility-sparing surgical options, such as extended cone biopsy or trachelectomy. High spatial resolution images with maximal contrast between tumour and surrounding background are achieved with T2-weighted and diffusion-weighted (DW) magnetic resonance imaging (MRI) obtained using an endovaginal receiver coil. Tumour size and volume demonstrated in this way correlates between observers and with histology and differences between MRI and histology estimates of normal endocervical canal length are not significant. For planning fertility-sparing surgery, this imaging technique facilitates the best oncological outcome while minimising subsequent obstetric risks. Parametrial invasion may be assessed on large field of view T2-weighted MRI. The fat content of the parametrium limits the utility of DW imaging in this context, because fat typically shows diffusion restriction. The use of contrast-enhanced MRI for assessing the parametrium does not provide additional benefits to the T2-weighted images and the need for an extrinsic contrast agent merely adds additional complexity and cost. For nodal assessment, 18fluorodeoxyglucose positron emission tomography-computerised tomography (18FDG PET-CT) remains the gold standard.
Collapse
Affiliation(s)
- N M deSouza
- Division of Radiotherapy and Imaging, The Institute of Cancer Research, 15 Cotswold Road, SM2 5NG, UK.
| |
Collapse
|
16
|
Berbís MA, Aneiros-Fernández J, Mendoza Olivares FJ, Nava E, Luna A. Role of artificial intelligence in multidisciplinary imaging diagnosis of gastrointestinal diseases. World J Gastroenterol 2021; 27:4395-4412. [PMID: 34366612 PMCID: PMC8316909 DOI: 10.3748/wjg.v27.i27.4395] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 04/14/2021] [Accepted: 06/07/2021] [Indexed: 02/06/2023] Open
Abstract
The use of artificial intelligence-based tools is regarded as a promising approach to increase clinical efficiency in diagnostic imaging, improve the interpretability of results, and support decision-making for the detection and prevention of diseases. Radiology, endoscopy and pathology images are suitable for deep-learning analysis, potentially changing the way care is delivered in gastroenterology. The aim of this review is to examine the key aspects of different neural network architectures used for the evaluation of gastrointestinal conditions, by discussing how different models behave in critical tasks, such as lesion detection or characterization (i.e. the distinction between benign and malignant lesions of the esophagus, the stomach and the colon). To this end, we provide an overview on recent achievements and future prospects in deep learning methods applied to the analysis of radiology, endoscopy and histologic whole-slide images of the gastrointestinal tract.
Collapse
Affiliation(s)
| | - José Aneiros-Fernández
- Department of Pathology, Hospital Universitario Clínico San Cecilio, Granada 18012, Spain
| | | | - Enrique Nava
- Department of Communications Engineering, University of Málaga, Malaga 29016, Spain
| | - Antonio Luna
- MRI Unit, Department of Radiology, HT Médica, Jaén 23007, Spain
| |
Collapse
|
17
|
Li X, Gao H, Zhu J, Huang Y, Zhu Y, Huang W, Li Z, Sun K, Liu Z, Tian J, Li B. 3D Deep Learning Model for the Pretreatment Evaluation of Treatment Response in Esophageal Carcinoma: A Prospective Study (ChiCTR2000039279). Int J Radiat Oncol Biol Phys 2021; 111:926-935. [PMID: 34229050 DOI: 10.1016/j.ijrobp.2021.06.033] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Revised: 06/11/2021] [Accepted: 06/21/2021] [Indexed: 12/22/2022]
Abstract
PURPOSE To develop and validate a pretreatment computed tomography (CT)-based deep-learning (DL) model for predicting the treatment response to concurrent chemoradiation therapy (CCRT) among patients with locally advanced thoracic esophageal squamous cell carcinoma (TESCC). METHODS AND MATERIALS We conducted a prospective, multicenter study on the therapeutic efficacy of CCRT among TESCC patients across 9 hospitals in China (ChiCTR2000039279). A total of 306 patients with locally advanced TESCC diagnosed by histopathology from August 2015 to May 2020 were included in this study. A 3-dimensional DL radiomics model (3D-DLRM) was developed and validated based on pretreatment CT images to predict the response to CCRT. Furthermore, the prediction performance of the newly developed 3D-DLRM was analyzed according to 3 categories: radiation therapy plan, radiation field, and prescription dose used. RESULTS The 3D-DLRM achieved good prediction performance, with areas under the receiver operating characteristic curve of 0.897 (95% confidence interval, 0.840-0.959) for the training cohort and 0.833 (95% confidence interval, 0.654-1.000) for the validation cohort. Specifically, the 3D-DLRM accurately predicted patients who would not respond to CCRT, with a positive predictive value (PPV) of 100% for the validation cohort. Moreover, the 3D-DLRM performed well in all 3 categories, each with areas under the receiver operating characteristic curve of >0.8 and positive predictive values of approximately 100%. CONCLUSION The proposed pretreatment CT-based 3D-DLRM provides a potential tool for predicting the response to CCRT among patients with locally advanced TESCC. With the help of precise pretreatment prediction, we may guide the individualized treatment of patients and improve survival.
Collapse
Affiliation(s)
- Xiaoqin Li
- Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China; Shandong Medical Imaging and Radiotherapy Engineering Center (SMIREC), Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Han Gao
- Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, School of Medicine and Engineering, Beihang University, Beijing, China; CAS Key Laboratory of Molecular Imaging, Beijing Key Laboratory of Molecular Imaging, State Key Laboratory of Management and Control for Complex Systems, Institute of Automation, Chinese Academy of Sciences, Beijing, China
| | - Jian Zhu
- Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China; Shandong Medical Imaging and Radiotherapy Engineering Center (SMIREC), Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Yong Huang
- Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China; Shandong Medical Imaging and Radiotherapy Engineering Center (SMIREC), Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Yongbei Zhu
- Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, School of Medicine and Engineering, Beihang University, Beijing, China; CAS Key Laboratory of Molecular Imaging, Beijing Key Laboratory of Molecular Imaging, State Key Laboratory of Management and Control for Complex Systems, Institute of Automation, Chinese Academy of Sciences, Beijing, China
| | - Wei Huang
- Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China; Shandong Medical Imaging and Radiotherapy Engineering Center (SMIREC), Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Zhenjiang Li
- Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China; Shandong Medical Imaging and Radiotherapy Engineering Center (SMIREC), Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Kai Sun
- Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, School of Medicine and Engineering, Beihang University, Beijing, China; Engineering Research Center of Molecular and Neuro Imaging of Ministry of Education, School of Life Science and Technology, Xidian University, Xi'an, Shaanxi, China
| | - Zhenyu Liu
- CAS Key Laboratory of Molecular Imaging, Beijing Key Laboratory of Molecular Imaging, State Key Laboratory of Management and Control for Complex Systems, Institute of Automation, Chinese Academy of Sciences, Beijing, China; CAS Center for Excellence in Brain Science and Intelligence Technology, Institute of Automation, Chinese Academy of Sciences, Beijing, China; School of Artificial Intelligence, University of Chinese Academy of Sciences, Beijing, China
| | - Jie Tian
- Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, School of Medicine and Engineering, Beihang University, Beijing, China; CAS Key Laboratory of Molecular Imaging, Beijing Key Laboratory of Molecular Imaging, State Key Laboratory of Management and Control for Complex Systems, Institute of Automation, Chinese Academy of Sciences, Beijing, China; Engineering Research Center of Molecular and Neuro Imaging of Ministry of Education, School of Life Science and Technology, Xidian University, Xi'an, Shaanxi, China; Key Laboratory of Big Data-Based Precision Medicine (Beihang University), Ministry of Industry and Information Technology, Beijing, China
| | - Baosheng Li
- Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China; Shandong Medical Imaging and Radiotherapy Engineering Center (SMIREC), Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China.
| |
Collapse
|
18
|
Wesdorp NJ, Hellingman T, Jansma EP, van Waesberghe JHTM, Boellaard R, Punt CJA, Huiskens J, Kazemier G. Advanced analytics and artificial intelligence in gastrointestinal cancer: a systematic review of radiomics predicting response to treatment. Eur J Nucl Med Mol Imaging 2021; 48:1785-1794. [PMID: 33326049 PMCID: PMC8113210 DOI: 10.1007/s00259-020-05142-w] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Accepted: 11/29/2020] [Indexed: 02/08/2023]
Abstract
PURPOSE Advanced medical image analytics is increasingly used to predict clinical outcome in patients diagnosed with gastrointestinal tumors. This review provides an overview on the value of radiomics in predicting response to treatment in patients with gastrointestinal tumors. METHODS A systematic review was conducted, according to PRISMA guidelines. The protocol was prospectively registered (PROSPERO: CRD42019128408). PubMed, Embase, and Cochrane databases were searched. Original studies reporting on the value of radiomics in predicting response to treatment in patients with a gastrointestinal tumor were included. A narrative synthesis of results was conducted. Results were stratified by tumor type. Quality assessment of included studies was performed, according to the radiomics quality score. RESULTS The comprehensive literature search identified 1360 unique studies, of which 60 articles were included for analysis. In 37 studies, radiomics models and individual radiomic features showed good predictive performance for response to treatment (area under the curve or accuracy > 0.75). Various strategies to construct predictive models were used. Internal validation of predictive models was often performed, while the majority of studies lacked external validation. None of the studies reported predictive models implemented in clinical practice. CONCLUSION Radiomics is increasingly used to predict response to treatment in patients suffering from gastrointestinal cancer. This review demonstrates its great potential to help predict response to treatment and improve patient selection and early adjustment of treatment strategy in a non-invasive manner.
Collapse
Affiliation(s)
- Nina J Wesdorp
- Department of Surgery, Cancer Center Amsterdam, Amsterdam University Medical Centers, Vrije Universiteit, Amsterdam, The Netherlands.
| | - Tessa Hellingman
- Department of Surgery, Cancer Center Amsterdam, Amsterdam University Medical Centers, Vrije Universiteit, Amsterdam, The Netherlands
| | - Elise P Jansma
- Department of Epidemiology and Biostatistics, Amsterdam University Medical Centers, Vrije Universiteit, Amsterdam, The Netherlands
| | - Jan-Hein T M van Waesberghe
- Department of Radiology and Molecular Imaging, Cancer Center Amsterdam, Amsterdam University Medical Centers, Vrije Universiteit, Amsterdam, The Netherlands
| | - Ronald Boellaard
- Department of Radiology and Nuclear Medicine, Cancer Center Amsterdam, Amsterdam University Medical Centers, Vrije Universiteit, Amsterdam, The Netherlands
| | - Cornelis J A Punt
- Department of Epidemiology, Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht, The Netherlands
| | | | - Geert Kazemier
- Department of Surgery, Cancer Center Amsterdam, Amsterdam University Medical Centers, Vrije Universiteit, Amsterdam, The Netherlands
| |
Collapse
|
19
|
Xie CY, Pang CL, Chan B, Wong EYY, Dou Q, Vardhanabhuti V. Machine Learning and Radiomics Applications in Esophageal Cancers Using Non-Invasive Imaging Methods-A Critical Review of Literature. Cancers (Basel) 2021; 13:2469. [PMID: 34069367 PMCID: PMC8158761 DOI: 10.3390/cancers13102469] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2021] [Revised: 05/12/2021] [Accepted: 05/15/2021] [Indexed: 11/16/2022] Open
Abstract
Esophageal cancer (EC) is of public health significance as one of the leading causes of cancer death worldwide. Accurate staging, treatment planning and prognostication in EC patients are of vital importance. Recent advances in machine learning (ML) techniques demonstrate their potential to provide novel quantitative imaging markers in medical imaging. Radiomics approaches that could quantify medical images into high-dimensional data have been shown to improve the imaging-based classification system in characterizing the heterogeneity of primary tumors and lymph nodes in EC patients. In this review, we aim to provide a comprehensive summary of the evidence of the most recent developments in ML application in imaging pertinent to EC patient care. According to the published results, ML models evaluating treatment response and lymph node metastasis achieve reliable predictions, ranging from acceptable to outstanding in their validation groups. Patients stratified by ML models in different risk groups have a significant or borderline significant difference in survival outcomes. Prospective large multi-center studies are suggested to improve the generalizability of ML techniques with standardized imaging protocols and harmonization between different centers.
Collapse
Affiliation(s)
- Chen-Yi Xie
- Department of Diagnostic Radiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China;
| | - Chun-Lap Pang
- Department of Radiology, The Christies’ Hospital, Manchester M20 4BX, UK;
- Division of Dentistry, School of Medical Sciences, University of Manchester, Manchester M15 6FH, UK
| | - Benjamin Chan
- Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China; (B.C.); (E.Y.-Y.W.)
| | - Emily Yuen-Yuen Wong
- Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China; (B.C.); (E.Y.-Y.W.)
| | - Qi Dou
- Department of Computer Science and Engineering, The Chinese University of Hong Kong, Hong Kong, China;
| | - Varut Vardhanabhuti
- Department of Diagnostic Radiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China;
| |
Collapse
|
20
|
Luo HS, Huang SF, Xu HY, Li XY, Wu SX, Wu DH. A nomogram based on pretreatment CT radiomics features for predicting complete response to chemoradiotherapy in patients with esophageal squamous cell cancer. Radiat Oncol 2020; 15:249. [PMID: 33121507 PMCID: PMC7597023 DOI: 10.1186/s13014-020-01692-3] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Accepted: 10/20/2020] [Indexed: 12/09/2022] Open
Abstract
Purpose To develop and validate a nomogram model to predict complete response (CR) after concurrent chemoradiotherapy (CCRT) in esophageal squamous cell carcinoma (ESCC) patients using pretreatment CT radiomic features. Methods Data of patients diagnosed as ESCC and treated with CCRT in Shantou Central Hospital during the period from January 2013 to December 2015 were retrospectively collected. Eligible patients were included in this study and randomize divided into a training set and a validation set after successive screening. The least absolute shrinkage and selection operator (LASSO) with logistic regression to select radiomics features calculating Rad-score in the training set. The logistic regression analysis was performed to identify the predictive clinical factors for developing a nomogram model. The area under the receiver operating characteristic curves (AUC) was used to assess the performance of the predictive nomogram model and decision curve was used to analyze the impact of the nomogram model on clinical treatment decisions. Results A total of 226 patients were included and randomly divided into two groups, 160 patients in training set and 66 patients in validation set. After LASSO analysis, seven radiomics features were screened out to develop a radiomics signature Rad-score. The AUC of Rad-score was 0.812 (95% CI 0.742–0.869, p < 0.001) in the training set and 0.744 (95% CI 0.632–0.851, p = 0.003) in the validation set. Multivariate analysis showed that Rad-score and clinical staging were independent predictors of CR status, with p values of 0.035 and 0.023, respectively. A nomogram model incorporating Rad-socre and clinical staging was developed and validated, with an AUC of 0.844 (95% CI 0.779–0.897) in the training set and 0.807 (95% CI 0.691–0.894) in the validation set. Delong test showed that the nomogram model was significantly superior to the clinical staging, with p < 0.001 in the training set and p = 0.026 in the validation set. The decision curve showed that the nomogram model was superior to the clinical staging when the risk threshold was greater than 25%. Conclusion We developed and validated a nomogram model for predicting CR status of ESCC patients after CCRT. The nomogram model was combined radiomics signature Rad-score and clinical staging. This model provided us with an economical and simple method for evaluating the response of chemoradiotherapy for patients with ESCC.
Collapse
Affiliation(s)
- He-San Luo
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong Province, China.,Department of Radiotherapy, Shantou Central Hospital, Shantou, Guangdong, China
| | - Shao-Fu Huang
- Department of Radiotherapy, Shantou Central Hospital, Shantou, Guangdong, China
| | - Hong-Yao Xu
- Department of Radiotherapy, Shantou Central Hospital, Shantou, Guangdong, China
| | - Xu-Yuan Li
- Department of Medical Oncology, Shantou Central Hospital, Shantou, Guangdong, China
| | - Sheng-Xi Wu
- Department of Radiotherapy, Shantou Central Hospital, Shantou, Guangdong, China
| | - De-Hua Wu
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong Province, China.
| |
Collapse
|
21
|
Cao JM, Yang JQ, Ming ZQ, Wu JL, Yang LQ, Chen TW, Li R, Ou J, Zhang XM, Mu QW, Li HJ, Hu J. A radiomics model of liver CT to predict risk of hepatic encephalopathy secondary to hepatitis B related cirrhosis. Eur J Radiol 2020; 130:109201. [PMID: 32738462 DOI: 10.1016/j.ejrad.2020.109201] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 07/18/2020] [Accepted: 07/24/2020] [Indexed: 12/12/2022]
Abstract
PURPOSE To build a radiomics model of liver contrast-enhanced computed tomography (CT) to predict hepatic encephalopathy secondary to Hepatitis B related cirrhosis. MATERIALS AND METHODS This study consisted of 304 consecutive patients with first-diagnosed hepatitis B related cirrhosis. 212 and 92 patients were randomly computer-generated into training and testing cohorts, among which 38 and 21 patients endured HE, respectively. 356 radiomics features of liver were extracted from portal venous-phase CT data, and 3 clinical features were collected from medical record. After data were standardized by Z-score, we used least absolute shrinkage and selection operator to choose useful radiomics features. Ultimately, three predictive models including a radiomics model, a clinical model and an integrated model of radiomics and clinical features were built by analysis of R-software. Predictive performance was tested by multivariable logistic regression, and evaluated by area under receiver-operating characteristic curve (AUC), and accuracy. RESULTS 19 radiomics features of liver CT were selected. The selected radiomics features and 3 relevant clinical features were applied to develop a radiomics model, a clinical model, and an integrated model of both radiomics and clinical features. The integrated model showed better performance than the radiomics model or clinical model to predict HE (AUC = 0.94 vs. 0.91 or 0.76, and 0.87 vs. 0.86 or 0.73; accuracy = 0.93 vs. 0.89 or 0.83, and 0.83 vs. 0.84 or 0.77) in the training and testing cohorts, respectively. CONCLUSION The integrated model of radiomics and clinical features could well predict HE secondary to hepatitis B related cirrhosis.
Collapse
Affiliation(s)
- Jin-Ming Cao
- Sichuan Key Laboratory of Medical Imaging, and Department of Radiology, Affiliated Hospital of North Sichuan Medical College, Nanchong 637000, Sichuan, China; Department of Radiology, Nanchong Central Hospital/Second School of Clinical Medicine, North Sichuan Medical College, Nanchong 637000, Sichuan, China
| | - Jian-Qiong Yang
- Sichuan Key Laboratory of Medical Imaging, and Department of Radiology, Affiliated Hospital of North Sichuan Medical College, Nanchong 637000, Sichuan, China; Department of Radiology, Nanchong Central Hospital/Second School of Clinical Medicine, North Sichuan Medical College, Nanchong 637000, Sichuan, China
| | - Zhi-Qiang Ming
- Sichuan Key Laboratory of Medical Imaging, and Department of Radiology, Affiliated Hospital of North Sichuan Medical College, Nanchong 637000, Sichuan, China; Department of Radiology, Zigong First People's Hospital, Zigong 643000, Sichuan, China
| | - Jia-Long Wu
- Sichuan Key Laboratory of Medical Imaging, and Department of Radiology, Affiliated Hospital of North Sichuan Medical College, Nanchong 637000, Sichuan, China; Department of Radiology, Nanchong Central Hospital/Second School of Clinical Medicine, North Sichuan Medical College, Nanchong 637000, Sichuan, China
| | - Li-Qin Yang
- Sichuan Key Laboratory of Medical Imaging, and Department of Radiology, Affiliated Hospital of North Sichuan Medical College, Nanchong 637000, Sichuan, China; Department of Radiology, Nanchong Central Hospital/Second School of Clinical Medicine, North Sichuan Medical College, Nanchong 637000, Sichuan, China
| | - Tian-Wu Chen
- Sichuan Key Laboratory of Medical Imaging, and Department of Radiology, Affiliated Hospital of North Sichuan Medical College, Nanchong 637000, Sichuan, China.
| | - Rui Li
- Sichuan Key Laboratory of Medical Imaging, and Department of Radiology, Affiliated Hospital of North Sichuan Medical College, Nanchong 637000, Sichuan, China
| | - Jing Ou
- Sichuan Key Laboratory of Medical Imaging, and Department of Radiology, Affiliated Hospital of North Sichuan Medical College, Nanchong 637000, Sichuan, China
| | - Xiao-Ming Zhang
- Sichuan Key Laboratory of Medical Imaging, and Department of Radiology, Affiliated Hospital of North Sichuan Medical College, Nanchong 637000, Sichuan, China
| | - Qi-Wen Mu
- Department of Radiology, Nanchong Central Hospital/Second School of Clinical Medicine, North Sichuan Medical College, Nanchong 637000, Sichuan, China
| | - Hong-Jun Li
- Department of Radiology, Beijing YouAn Hospital, Capital Medical University, Beijing 100069, China.
| | - Jiani Hu
- Department of Radiology, Wayne State University, Detroit, MI, USA
| |
Collapse
|
22
|
Feng P, Wang ZD, Fan W, Liu H, Pan JJ. Diagnostic advances of artificial intelligence and radiomics in gastroenterology. Artif Intell Gastroenterol 2020; 1:37-50. [DOI: 10.35712/aig.v1.i2.37] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Revised: 08/22/2020] [Accepted: 08/27/2020] [Indexed: 02/06/2023] Open
Abstract
Traditional medical imaging, including ultrasound, computed tomography, magnetic resonance imaging, or positron emission tomography, remains widely used diagnostic modalities for gastrointestinal diseases at present. These modalities are used to assess changes in morphology, attenuation, signal intensity, and enhancement characteristics. Gastrointestinal tumors, especially malignant tumors, are commonly seen in clinical practice with an increasing number of deaths each year. Because the imaging manifestations of different diseases usually overlap, accurate early diagnosis of tumor lesions, noninvasive and effective evaluation of tumor staging, and prediction of prognosis remain challenging. Fortunately, traditional medical images contain a great deal of important information that cannot be recognized by human eyes but can be extracted by artificial intelligence (AI) technology, which can quantitatively assess the heterogeneity of lesions and provide valuable information, including therapeutic effects and patient prognosis. With the development of computer technology, the combination of medical imaging and AI technology is considered to represent a promising field in medical image analysis. This new emerging field is called “radiomics”, which makes big data mining and extraction from medical imagery possible and can help clinicians make effective decisions and develop personalized treatment plans. Recently, AI and radiomics have been gradually applied to lesion detection, qualitative and quantitative diagnosis, histopathological grading and staging of tumors, therapeutic efficacy assessment, and prognosis evaluation. In this minireview, we briefly introduce the basic principles and technology of radiomics. Then, we review the research and application of AI and radiomics in gastrointestinal diseases, especially diagnostic advancements of radiomics in the differential diagnosis, treatment option, assessment of therapeutic efficacy, and prognosis evaluation of esophageal, gastric, hepatic, pancreatic, and colorectal diseases.
Collapse
Affiliation(s)
- Pei Feng
- Department of Radiology, PLA Rocket Force Characteristic Medical Center, Beijing 100088, China
| | - Zhen-Dong Wang
- Department of Ultrasound, Beijing Sihui Hospital of Traditional Chinese Medicine, Beijing 100022, China
| | - Wei Fan
- Department of Radiology, PLA Rocket Force Characteristic Medical Center, Beijing 100088, China
| | - Heng Liu
- Department of Radiology, PLA Rocket Force Characteristic Medical Center, Beijing 100088, China
| | - Jing-Jing Pan
- Department of Radiology, PLA Rocket Force Characteristic Medical Center, Beijing 100088, China
| |
Collapse
|
23
|
Song L, Yin J. Application of Texture Analysis Based on Sagittal Fat-Suppression and Oblique Axial T2-Weighted Magnetic Resonance Imaging to Identify Lymph Node Invasion Status of Rectal Cancer. Front Oncol 2020; 10:1364. [PMID: 32850437 PMCID: PMC7426518 DOI: 10.3389/fonc.2020.01364] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Accepted: 06/29/2020] [Indexed: 12/18/2022] Open
Abstract
Objective: To investigate the value of texture features derived from T2-weighted magnetic resonance imaging (T2WI) for predicting preoperative lymph node invasion (N stage) in rectal cancer. Materials and Methods: One hundred and eighty-two patients with histopathologically confirmed rectal cancer and preoperative magnetic resonance imaging were retrospectively analyzed, who were divided into high (N1-2) and low N stage (N0). Texture features were calculated from histogram, gray-level co-occurrence matrix, and gray-level run-length matrix from sagittal fat-suppression and oblique axial T2WI. Independent sample t-test or Mann-Whitney U-test were used for statistical analysis. Multivariate logistic regression analysis was conducted to build the predictive models. Predictive performance was evaluated by receiver operating characteristic (ROC) analysis. Results: Energy (ENE), entropy (ENT), information correlation (INC), long-run emphasis (LRE), and short-run low gray-level emphasis (SRLGLE) extracted from sagittal fat-suppression T2WI, and ENE, ENT, INC, low gray-level run emphasis (LGLRE), and SRLGLE from oblique axial T2WI were significantly different between stage N0 and stage N1-2 tumors. The multivariate analysis for features from sagittal fat-suppression T2WI showed that higher SRLGLE and lower ENE were independent predictors of lymph node invasion. The model reached an area under ROC curve (AUC) of 0.759. The analysis for features from oblique axial T2WI showed that higher INC and SRLGLE were independent predictors. The model achieved an AUC of 0.747. The analysis for all extracted features showed that lower ENE from sagittal fat-suppression T2WI and higher INC and SRLGLE from oblique axial T2WI were independent predictors. The model showed an AUC of 0.772. Conclusions: Texture features derived from T2WI could provide valuable information for identifying the status of lymph node invasion in rectal cancer.
Collapse
Affiliation(s)
- Lirong Song
- Department of Radiology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Jiandong Yin
- Department of Radiology, Shengjing Hospital of China Medical University, Shenyang, China
| |
Collapse
|
24
|
Scalco E, Belfatto A, Mastropietro A, Rancati T, Avuzzi B, Messina A, Valdagni R, Rizzo G. T2w‐MRI signal normalization affects radiomics features reproducibility. Med Phys 2020; 47:1680-1691. [DOI: 10.1002/mp.14038] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Revised: 01/13/2020] [Accepted: 01/14/2020] [Indexed: 01/01/2023] Open
Affiliation(s)
- Elisa Scalco
- CNR, Institute of Biomedical Technologies (ITB) Segrate Italy
- CNR, Institute of Molecular Bioimaging and Physiology (IBFM) Segrate Italy
| | - Antonella Belfatto
- CNR, Institute of Molecular Bioimaging and Physiology (IBFM) Segrate Italy
| | - Alfonso Mastropietro
- CNR, Institute of Biomedical Technologies (ITB) Segrate Italy
- CNR, Institute of Molecular Bioimaging and Physiology (IBFM) Segrate Italy
| | - Tiziana Rancati
- Fondazione IRCCS Istituto Nazionale dei Tumori Prostate Cancer Program Milano Italy
| | - Barbara Avuzzi
- Radiation Oncology 1 Fondazione IRCCS Istituto Nazionale dei Tumori Milano Italy
| | - Antonella Messina
- Radiology Fondazione IRCCS Istituto Nazionale dei Tumori Milano Italy
| | - Riccardo Valdagni
- Fondazione IRCCS Istituto Nazionale dei Tumori Prostate Cancer Program Milano Italy
- Radiation Oncology 1 Fondazione IRCCS Istituto Nazionale dei Tumori Milano Italy
- Department of Oncology and Hemato‐oncology Università degli Studi di Milano Milano Italy
| | - Giovanna Rizzo
- CNR, Institute of Biomedical Technologies (ITB) Segrate Italy
- CNR, Institute of Molecular Bioimaging and Physiology (IBFM) Segrate Italy
| |
Collapse
|
25
|
Cao Q, Li Y, Li Z, An D, Li B, Lin Q. Development and validation of a radiomics signature on differentially expressed features of 18F-FDG PET to predict treatment response of concurrent chemoradiotherapy in thoracic esophagus squamous cell carcinoma. Radiother Oncol 2020; 146:9-15. [PMID: 32065875 DOI: 10.1016/j.radonc.2020.01.027] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2019] [Revised: 01/06/2020] [Accepted: 01/30/2020] [Indexed: 12/29/2022]
Abstract
BACKGROUND AND PURPOSE To investigate potential image markers for early prediction of treatment response on thoracic esophagus squamous cell carcinoma (ESCC) treated with concurrent chemoradiotherapy (CCRT). MATERIALS AND METHODS 159 thoracic ESCC patients enrolled from two institutions were divided into training and validation sets. A total of 944 radiomics features were extracted from pretreatment 18F-FDG PET images. We first performed the inter-observer reproducibility test in 10 pairs of patients (responders vs. nonresponders), and the limma package was used to identify differentially expressed features (DEFs). Then the least absolute shrinkage and selection operator (LASSO) logistic regression model with 10-fold cross-validation was used to construct a treatment response related radiomics signature. Finally, the performance was assessed in both sets with receiver operating characteristic (ROC) curves and Kaplan-Meier analysis. RESULTS After the inter-observer test, 691 features were considered reproducible and been retained (ICC > 0.9). 61 DEFs were selected from limma and entered into the LASSO logistic regression model. The radiomics signature was significantly associated with treatment response in the training (p < 0.001) and validation set (p = 0.026), which achieved area under curve (AUC) values of 0.844 and 0.835, respectively. Delong test results of two ROCs showed no significant difference (p = 0.918). The cut-off value of the radiomics signature could successfully divide patients into high-risk and low-risk groups in both sets. CONCLUSION This study indicated that the proposed radiomics signature could be a useful image marker to predict the therapeutic response of thoracic ESCC patients treated with CCRT.
Collapse
Affiliation(s)
- Qiang Cao
- Laboratory of Image Science and Technology, School of Computer Science and Engineering, Southeast University, Nanjing, PR China; Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, PR China; Shandong Medical Imaging and Radiotherapy Engineering Center (SMIREC), Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, PR China
| | - Yimin Li
- Department of Radiation Oncology, Xiamen Cancer Center, The First Affiliated Hospital of Xiamen University, Xiamen, PR China
| | - Zhe Li
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, PR China; Shandong Medical Imaging and Radiotherapy Engineering Center (SMIREC), Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, PR China
| | - Dianzheng An
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, PR China; Shandong Medical Imaging and Radiotherapy Engineering Center (SMIREC), Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, PR China
| | - Baosheng Li
- Laboratory of Image Science and Technology, School of Computer Science and Engineering, Southeast University, Nanjing, PR China; Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, PR China; Shandong Medical Imaging and Radiotherapy Engineering Center (SMIREC), Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, PR China.
| | - Qin Lin
- Department of Radiation Oncology, Xiamen Cancer Center, The First Affiliated Hospital of Xiamen University, Xiamen, PR China.
| |
Collapse
|
26
|
Wormald BW, Doran SJ, Ind TE, D'Arcy J, Petts J, deSouza NM. Radiomic features of cervical cancer on T2-and diffusion-weighted MRI: Prognostic value in low-volume tumors suitable for trachelectomy. Gynecol Oncol 2020; 156:107-114. [PMID: 31685232 PMCID: PMC7001101 DOI: 10.1016/j.ygyno.2019.10.010] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2019] [Revised: 10/07/2019] [Accepted: 10/08/2019] [Indexed: 12/19/2022]
Abstract
BACKGROUND Textural features extracted from MRI potentially provide prognostic information additional to volume for influencing surgical management of cervical cancer. PURPOSE To identify textural features that differ between cervical tumors above and below the volume threshold of eligibility for trachelectomy and determine their value in predicting recurrence in patients with low-volume tumors. METHODS Of 378 patients with Stage1-2 cervical cancer imaged prospectively (3T, endovaginal coil), 125 had well-defined, histologically-confirmed squamous or adenocarcinomas with >100 voxels (>0.07 cm3) suitable for radiomic analysis. Regions-of-interest outlined the whole tumor on T2-W images and apparent diffusion coefficient (ADC) maps. Textural features based on grey-level co-occurrence matrices were compared (Mann-Whitney test with Bonferroni correction) between tumors greater (n = 46) or less (n = 79) than 4.19 cm3. Clustering eliminated correlated variables. Significantly different features were used to predict recurrence (regression modelling) in surgically-treated patients with low-volume tumors and compared with a model using clinico-pathological features. RESULTS Textural features (Dissimilarity, Energy, ClusterProminence, ClusterShade, InverseVariance, Autocorrelation) in 6 of 10 clusters from T2-W and ADC data differed between high-volume (mean ± SD 15.3 ± 11.7 cm3) and low-volume (mean ± SD 1.3 ± 1.2 cm3) tumors. (p < 0.02). In low-volume tumors, predicting recurrence was indicated by: Dissimilarity, Energy (ADC-radiomics, AUC = 0.864); Dissimilarity, ClusterProminence, InverseVariance (T2-W-radiomics, AUC = 0.808); Volume, Depth of Invasion, LymphoVascular Space Invasion (clinico-pathological features, AUC = 0.794). Combining ADC-radiomic (but not T2-radiomic) and clinico-pathological features improved prediction of recurrence compared to the clinico-pathological model (AUC = 0.916, p = 0.006). Findings were supported by bootstrap re-sampling (n = 1000). CONCLUSION Textural features from ADC maps and T2-W images differ between high- and low-volume tumors and potentially predict recurrence in low-volume tumors.
Collapse
Affiliation(s)
- Benjamin W Wormald
- MRI Unit, Division of Radiotherapy and Imaging, The Institute of Cancer Research and the Royal Marsden NHS Foundation Trust, Sutton, UK
| | - Simon J Doran
- MRI Unit, Division of Radiotherapy and Imaging, The Institute of Cancer Research and the Royal Marsden NHS Foundation Trust, Sutton, UK
| | - Thomas Ej Ind
- Department of Gynaecological Oncology, The Royal Marsden NHS Foundation Trust, London, UK; St George's University of London, Tooting, London, UK
| | - James D'Arcy
- MRI Unit, Division of Radiotherapy and Imaging, The Institute of Cancer Research and the Royal Marsden NHS Foundation Trust, Sutton, UK
| | - James Petts
- MRI Unit, Division of Radiotherapy and Imaging, The Institute of Cancer Research and the Royal Marsden NHS Foundation Trust, Sutton, UK
| | - Nandita M deSouza
- MRI Unit, Division of Radiotherapy and Imaging, The Institute of Cancer Research and the Royal Marsden NHS Foundation Trust, Sutton, UK.
| |
Collapse
|
27
|
Yang JQ, Zeng R, Cao JM, Wu CQ, Chen TW, Li R, Zhang XM, Ou J, Li HJ, Mu QW. Predicting gastro-oesophageal variceal bleeding in hepatitis B-related cirrhosis by CT radiomics signature. Clin Radiol 2019; 74:976.e1-976.e9. [PMID: 31604574 DOI: 10.1016/j.crad.2019.08.028] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2019] [Accepted: 08/30/2019] [Indexed: 11/20/2022]
Abstract
AIM To develop liver a computed tomography (CT) radiomics model to predict gastro-oesophageal variceal bleeding (GVB) secondary to hepatitis B-related cirrhosis. MATERIALS AND METHODS Electronic medical records and image data of liver triple-phase contrast-enhanced CT examinations of 295 patients with hepatitis B-related cirrhosis were collected retrospectively from two hospitals. Two hundred and thirty-six and 59 patients were enrolled randomly into the training and validation cohorts, respectively; and 75 in the training cohort and 16 in the validation cohort endured GVB while the others did not during follow-up period. Radiomics features of the liver were extracted from the portal venous phase images, and clinical features came from medical records. The tree-based method and univariate feature selection were used to select useful features. The radiomics model, clinical model, and integration of radiomics and clinical models were built using the useful image features and/or clinical features. Predicting performance of three models was evaluated with the area under receiver-operating characteristic curve (AUC), accuracy, and F-1 score. RESULTS Twenty-one useful radiomics features and/or three clinical features were selected to build prediction models that correlated with GVB. AUC of integration of radiomics and clinical models was larger than of clinical or radiomics models for the training cohort (0.83±0.09 versus 0.64±0.08 or 0.82±0.10) and the validation cohort (0.64 versus 0.61 or 0.61). Integration of radiomics and clinical models obtained good performance in predicting GVB for both the training and validation cohorts (accuracy: 0.76±0.07 and 0.73, and F-1 score: 0.77±0.09 and 0.72, respectively). CONCLUSION Integration of the radiomics and clinical models may be a non-invasive method to predict GVB.
Collapse
Affiliation(s)
- J Q Yang
- Sichuan Key Laboratory of Medical Imaging, Department of Radiology, Affiliated Hospital of North Sichuan Medical College, Nanchong 637000, Sichuan, China; Department of Radiology, Nanchong Central Hospital/Second School of Clinical Medicine, North Sichuan Medical College, Nanchong 637000, Sichuan, China
| | - R Zeng
- Sichuan Key Laboratory of Medical Imaging, Department of Radiology, Affiliated Hospital of North Sichuan Medical College, Nanchong 637000, Sichuan, China
| | - J M Cao
- Sichuan Key Laboratory of Medical Imaging, Department of Radiology, Affiliated Hospital of North Sichuan Medical College, Nanchong 637000, Sichuan, China; Department of Radiology, Nanchong Central Hospital/Second School of Clinical Medicine, North Sichuan Medical College, Nanchong 637000, Sichuan, China
| | - C Q Wu
- Sichuan Key Laboratory of Medical Imaging, Department of Radiology, Affiliated Hospital of North Sichuan Medical College, Nanchong 637000, Sichuan, China
| | - T W Chen
- Sichuan Key Laboratory of Medical Imaging, Department of Radiology, Affiliated Hospital of North Sichuan Medical College, Nanchong 637000, Sichuan, China.
| | - R Li
- Sichuan Key Laboratory of Medical Imaging, Department of Radiology, Affiliated Hospital of North Sichuan Medical College, Nanchong 637000, Sichuan, China.
| | - X M Zhang
- Sichuan Key Laboratory of Medical Imaging, Department of Radiology, Affiliated Hospital of North Sichuan Medical College, Nanchong 637000, Sichuan, China
| | - J Ou
- Sichuan Key Laboratory of Medical Imaging, Department of Radiology, Affiliated Hospital of North Sichuan Medical College, Nanchong 637000, Sichuan, China
| | - H J Li
- Department of Radiology, Beijing YouAn Hospital, Capital Medical University, Beijing 100069, China
| | - Q W Mu
- Department of Radiology, Nanchong Central Hospital/Second School of Clinical Medicine, North Sichuan Medical College, Nanchong 637000, Sichuan, China
| |
Collapse
|
28
|
Sollini M, Antunovic L, Chiti A, Kirienko M. Towards clinical application of image mining: a systematic review on artificial intelligence and radiomics. Eur J Nucl Med Mol Imaging 2019; 46:2656-2672. [PMID: 31214791 PMCID: PMC6879445 DOI: 10.1007/s00259-019-04372-x] [Citation(s) in RCA: 167] [Impact Index Per Article: 27.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Accepted: 05/23/2019] [Indexed: 02/06/2023]
Abstract
PURPOSE The aim of this systematic review was to analyse literature on artificial intelligence (AI) and radiomics, including all medical imaging modalities, for oncological and non-oncological applications, in order to assess how far the image mining research stands from routine medical application. To do this, we applied a trial phases classification inspired from the drug development process. METHODS Among the articles we considered for inclusion from PubMed were multimodality AI and radiomics investigations, with a validation analysis aimed at relevant clinical objectives. Quality assessment of selected papers was performed according to the QUADAS-2 criteria. We developed the phases classification criteria for image mining studies. RESULTS Overall 34,626 articles were retrieved, 300 were selected applying the inclusion/exclusion criteria, and 171 high-quality papers (QUADAS-2 ≥ 7) were identified and analysed. In 27/171 (16%), 141/171 (82%), and 3/171 (2%) studies the development of an AI-based algorithm, radiomics model, and a combined radiomics/AI approach, respectively, was described. A total of 26/27(96%) and 1/27 (4%) AI studies were classified as phase II and III, respectively. Consequently, 13/141 (9%), 10/141 (7%), 111/141 (79%), and 7/141 (5%) radiomics studies were classified as phase 0, I, II, and III, respectively. All three radiomics/AI studies were categorised as phase II trials. CONCLUSIONS The results of the studies are promising but still not mature enough for image mining tools to be implemented in the clinical setting and be widely used. The transfer learning from the well-known drug development process, with some specific adaptations to the image mining discipline could represent the most effective way for radiomics and AI algorithms to become the standard of care tools.
Collapse
Affiliation(s)
- Martina Sollini
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, 20090, Pieve Emanuele, Milan, Italy
| | - Lidija Antunovic
- Nuclear Medicine, Humanitas Clinical and Research Center IRCCS, Rozzano, Milan, Italy
| | - Arturo Chiti
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, 20090, Pieve Emanuele, Milan, Italy
- Nuclear Medicine, Humanitas Clinical and Research Center IRCCS, Rozzano, Milan, Italy
| | - Margarita Kirienko
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, 20090, Pieve Emanuele, Milan, Italy.
| |
Collapse
|
29
|
Gabelloni M, Faggioni L, Neri E. Imaging biomarkers in upper gastrointestinal cancers. BJR Open 2019; 1:20190001. [PMID: 33178936 PMCID: PMC7592483 DOI: 10.1259/bjro.20190001] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2018] [Revised: 02/23/2019] [Accepted: 03/29/2019] [Indexed: 12/02/2022] Open
Abstract
In parallel with the increasingly widespread availability of high performance imaging platforms and recent progresses in pathobiological characterisation and treatment of gastrointestinal malignancies, imaging biomarkers have become a major research topic due to their potential to provide additional quantitative information to conventional imaging modalities that can improve accuracy at staging and follow-up, predict outcome, and guide treatment planning in an individualised manner. The aim of this review is to briefly examine the status of current knowledge about imaging biomarkers in the field of upper gastrointestinal cancers, highlighting their potential applications and future perspectives in patient management from diagnosis onwards.
Collapse
Affiliation(s)
- Michela Gabelloni
- Department of Translational Research, Diagnostic and Interventional Radiology, University of Pisa, Pisa, Italy
| | - Lorenzo Faggioni
- Department of Translational Research, Diagnostic and Interventional Radiology, University of Pisa, Pisa, Italy
| | - Emanuele Neri
- Department of Translational Research, Diagnostic and Interventional Radiology, University of Pisa, Pisa, Italy
| |
Collapse
|
30
|
Liu H, Zhang C, Wang L, Luo R, Li J, Zheng H, Yin Q, Zhang Z, Duan S, Li X, Wang D. MRI radiomics analysis for predicting preoperative synchronous distant metastasis in patients with rectal cancer. Eur Radiol 2018; 29:4418-4426. [PMID: 30413955 DOI: 10.1007/s00330-018-5802-7] [Citation(s) in RCA: 65] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2018] [Revised: 09/12/2018] [Accepted: 09/25/2018] [Indexed: 12/13/2022]
Abstract
OBJECTIVES To investigate the value of MRI radiomics based on T2-weighted (T2W) images in predicting preoperative synchronous distant metastasis (SDM) in patients with rectal cancer. METHODS This retrospective study enrolled 177 patients with histopathology-confirmed rectal adenocarcinoma (123 patients in the training cohort and 54 in the validation cohort). A total of 385 radiomics features were extracted from pretreatment T2W images. Five steps, including univariate statistical tests and a random forest algorithm, were performed to select the best preforming features for predicting SDM. Multivariate logistic regression analysis was conducted to build the clinical and clinical-radiomics combined models in the training cohort. The predictive performance was validated by receiver operating characteristics curve (ROC) analysis and clinical utility implementing a nomogram and decision curve analysis. RESULTS Fifty-nine patients (33.3%) were confirmed to have SDM. Six radiomics features and four clinical characteristics were selected for predicting SDM. The clinical-radiomics combined model performed better than the clinical model in both the training and validation datasets. A threshold of 0.44 yielded an area under the ROC (AUC) value of 0.827 (95% confidence interval (CI), 0.6963-0.9580), a sensitivity of 72.2%, a specificity of 94.4%, and an accuracy of 87.0% in the validation cohort for the combined model. A clinical-radiomics nomogram and decision curve analysis confirmed the clinical utility of the combined model. CONCLUSIONS Our proposed clinical-radiomics combined model could be utilized as a noninvasive biomarker for identifying patients at high risk of SDM, which could aid in tailoring treatment strategies. KEY POINTS • T2WI-based radiomics analysis helps predict synchronous distant metastasis (SDM) of rectal cancer. • The clinical-radiomics combined model could be utilized as a noninvasive biomarker for predicting SDM. • Personalized treatment can be carried out with greater confidence based on the risk stratification for SDM in rectal cancer.
Collapse
Affiliation(s)
- Huanhuan Liu
- Department of Radiology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, No. 1665 Kongjiang Road, Shanghai, 200092, China
| | - Caiyuan Zhang
- Department of Radiology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, No. 1665 Kongjiang Road, Shanghai, 200092, China
| | - Lijun Wang
- Department of Radiology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, No. 1665 Kongjiang Road, Shanghai, 200092, China
| | - Ran Luo
- Department of Radiology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, No. 1665 Kongjiang Road, Shanghai, 200092, China
| | - Jinning Li
- Department of Radiology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, No. 1665 Kongjiang Road, Shanghai, 200092, China
| | - Hui Zheng
- Department of Radiology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, No. 1665 Kongjiang Road, Shanghai, 200092, China
| | - Qiufeng Yin
- Department of Radiology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, No. 1665 Kongjiang Road, Shanghai, 200092, China
| | - Zhongyang Zhang
- Department of Radiology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, No. 1665 Kongjiang Road, Shanghai, 200092, China
| | - Shaofeng Duan
- GE Healthcare, Pudong New Town, No.1, Huatuo Road, Shanghai, 210000, China
| | - Xin Li
- GE Healthcare, Pudong New Town, No.1, Huatuo Road, Shanghai, 210000, China
| | - Dengbin Wang
- Department of Radiology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, No. 1665 Kongjiang Road, Shanghai, 200092, China.
| |
Collapse
|
31
|
Arimura H, Soufi M, Ninomiya K, Kamezawa H, Yamada M. Potentials of radiomics for cancer diagnosis and treatment in comparison with computer-aided diagnosis. Radiol Phys Technol 2018; 11:365-374. [PMID: 30374837 DOI: 10.1007/s12194-018-0486-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Revised: 10/14/2018] [Accepted: 10/17/2018] [Indexed: 12/22/2022]
Abstract
Computer-aided diagnosis (CAD) is a field that is essentially based on pattern recognition that improves the accuracy of a diagnosis made by a physician who takes into account the computer's "opinion" derived from the quantitative analysis of radiological images. Radiomics is a field based on data science that massively and comprehensively analyzes a large number of medical images to extract a large number of phenotypic features reflecting disease traits, and explores the associations between the features and patients' prognoses for precision medicine. According to the definitions for both, you may think that radiomics is not a paraphrase of CAD, but you may also think that these definitions are "image manipulation". However, there are common and different features between the two fields. This review paper elaborates on these common and different features and introduces the potential of radiomics for cancer diagnosis and treatment by comparing it with CAD.
Collapse
Affiliation(s)
- Hidetaka Arimura
- Division of Medical Quantum Science, Department of Health Sciences, Faculty of Medical Sciences, Kyushu University, 3-1-1, Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan.
| | - Mazen Soufi
- Division of Information Science, Nara Institute of Science and Technology, Ikoma, Japan
| | - Kenta Ninomiya
- Division of Medical Quantum Science, Department of Health Sciences, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Hidemi Kamezawa
- Department of Radiological Technology, Faculty of Fukuoka Medical Technology, Teikyo University, Omuta, Japan
| | - Masahiro Yamada
- Division of Medical Quantum Science, Department of Health Sciences, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| |
Collapse
|
32
|
Filograna L, Lenkowicz J, Cellini F, Dinapoli N, Manfrida S, Magarelli N, Leone A, Colosimo C, Valentini V. Identification of the most significant magnetic resonance imaging (MRI) radiomic features in oncological patients with vertebral bone marrow metastatic disease: a feasibility study. Radiol Med 2018; 124:50-57. [PMID: 30191445 DOI: 10.1007/s11547-018-0935-y] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2017] [Accepted: 08/20/2018] [Indexed: 02/07/2023]
Abstract
OBJECTIVES Recently, radiomic analysis has gained attention as a valuable instrument for the management of oncological patients. The aim of the study is to isolate which features of magnetic resonance imaging (MRI)-based radiomic analysis have to be considered the most significant predictors of metastasis in oncological patients with spinal bone marrow metastatic disease. MATERIALS AND METHODS Eight oncological patients (3 lung cancer; 1 prostatic cancer; 1 esophageal cancer; 1 nasopharyngeal cancer; 1 hepatocarcinoma; 1 breast cancer) with pre-radiotherapy MR imaging for a total of 58 dorsal vertebral bodies, 29 metastatic and 29 non-metastatic were included. Each vertebral body was contoured in T1 and T2 weighted images at a radiotherapy delineation console. The obtained data were transferred to an automated data extraction system for morphological, statistical and textural analysis. Eighty-nine features for each lesion in both T1 and T2 images were computed as the median of by-slice values. A Wilcoxon test was applied to the 89 features and the most statistically significant of them underwent to a stepwise feature selection, to find the best performing predictors of metastasis in a logistic regression model. An internal cross-validation via bootstrap was conducted for estimating the model performance in terms of the area under the curve (AUC) of the receiver operating characteristic. RESULTS Of the 89 textural features tested, 16 were found to differ with statistical significance in the metastatic vs non-metastatic group. The best performing model was constituted by two predictors for T1 and T2 images, namely one morphological feature (center of mass shift) (p value < 0.01) for both datasets and one histogram feature minimum grey level (p value < 0.01) for T1 images and one textural feature (grey-level co-occurrence matrix joint variance (p value < 0.01) for T2 images. The internal cross-validation showed an AUC of 0.8141 (95% CI 0.6854-0.9427) in T1 images and 0.9116 (95% CI 0.8294-0.9937) in T2 images. CONCLUSIONS The results suggest that MRI-based radiomic analysis on oncological patients with bone marrow metastatic disease is able to differentiate between metastatic and non-metastatic vertebral bodies. The most significant predictors of metastasis were found to be based on T2 sequence and were one morphological and one textural feature.
Collapse
Affiliation(s)
- Laura Filograna
- Department of Radiation Oncology - Gemelli-ART, Catholic University of Rome, School of Medicine, Foundation University Hospital "A. Gemelli", Largo A. Gemelli 8, 00168, Rome, Italy. .,Department of Radiological Sciences, Catholic University of Rome, School of Medicine, Foundation University Hospital "A. Gemelli", Largo A. Gemelli 8, 00168, Rome, Italy. .,Department of Radiological Sciences, PhD Training Program in Oncological Sciences, Catholic University of Rome, School of Medicine, University Hospital "A. Gemelli", Largo A. Gemelli 8, 00168, Rome, Italy. .,Department of Diagnostic and Interventional Radiology, Molecular Imaging and Radiotherapy, PTV Foundation, "Tor Vergata" University of Rome, Viale Oxford 81, 00133, Rome, Italy.
| | - Jacopo Lenkowicz
- Department of Radiation Oncology - Gemelli-ART, Catholic University of Rome, School of Medicine, Foundation University Hospital "A. Gemelli", Largo A. Gemelli 8, 00168, Rome, Italy
| | - Francesco Cellini
- Department of Radiation Oncology - Gemelli-ART, Catholic University of Rome, School of Medicine, Foundation University Hospital "A. Gemelli", Largo A. Gemelli 8, 00168, Rome, Italy
| | - Nicola Dinapoli
- Department of Radiation Oncology - Gemelli-ART, Catholic University of Rome, School of Medicine, Foundation University Hospital "A. Gemelli", Largo A. Gemelli 8, 00168, Rome, Italy
| | - Stefania Manfrida
- Department of Radiation Oncology - Gemelli-ART, Catholic University of Rome, School of Medicine, Foundation University Hospital "A. Gemelli", Largo A. Gemelli 8, 00168, Rome, Italy
| | - Nicola Magarelli
- Department of Radiological Sciences, Catholic University of Rome, School of Medicine, Foundation University Hospital "A. Gemelli", Largo A. Gemelli 8, 00168, Rome, Italy
| | - Antonio Leone
- Department of Radiological Sciences, Catholic University of Rome, School of Medicine, Foundation University Hospital "A. Gemelli", Largo A. Gemelli 8, 00168, Rome, Italy
| | - Cesare Colosimo
- Department of Radiological Sciences, Catholic University of Rome, School of Medicine, Foundation University Hospital "A. Gemelli", Largo A. Gemelli 8, 00168, Rome, Italy
| | - Vincenzo Valentini
- Department of Radiation Oncology - Gemelli-ART, Catholic University of Rome, School of Medicine, Foundation University Hospital "A. Gemelli", Largo A. Gemelli 8, 00168, Rome, Italy
| |
Collapse
|