1
|
Liu L, Cui WC, Sun Y, Wang H, Liang ZN, Wu W, Yan K, Ji YL, Dong L, Yang W. Classification of Neoadjuvant Therapy Response in Patients With Colorectal Liver Metastases Using Contrast-Enhanced Ultrasound-With Histological Pathology as the Gold Standard. ULTRASOUND IN MEDICINE & BIOLOGY 2025; 51:102-111. [PMID: 39414406 DOI: 10.1016/j.ultrasmedbio.2024.09.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 08/29/2024] [Accepted: 09/16/2024] [Indexed: 10/18/2024]
Abstract
OBJECTIVE To evaluate the response to neoadjuvant therapy in patients with colorectal liver metastases (CRLMs) using ultrasound(US) and contrast-enhanced ultrasound(CEUS), with correction to the tumor regression grade (TRG) of pathological results. METHODS This study included patients with resectable CRLMs admitted from February to December 2022. After at least 4 cycles neoadjuvant therapy, all the patients received US and CEUS examinations within two weeks before hepatectomy. CEUS clips were postprocessed with color parameter imaging (CPI) and microflow imaging (MFI) analysis. Logistic regression analyses were used to develop an evaluation Nomogram. Ultrasound-based model was constructed to discriminate between the response (TRG1/2/3) and nonresponse (TRG4/5) groups at the lesion level. The model's predictive ability was evaluated using the C index and calibration curve, with decision curve analysis assessing the Nomogram's added value. RESULTS The study analyzed 105 CRLM lesions (the lesion with the highest diameter analyzed for each patient), with 43.8% showing a response to therapy. Univariate analysis identified calcification on US (p = 0.039), CEUS enhancement degree (p < 0.001), CEUS enhancement pattern (p<0.001), CEUS washout type (p < 0.001), CEUS necrosis (p < 0.001), CPI feeding artery (p = 0.003) and MFI pattern (p < 0.001) were significantly associated with TRG. The multivariate analysis showed CEUS enhancement pattern (p = 0.026), CEUS washout type (p = 0.018) and CEUS necrosis (p = 0.005) were independently associated with the neoadjuvant therapy response. A Nomogram with the three independent predictors was developed, with an AUC of 0.898. CONCLUSION The ultrasound-based model provided accurate evaluation of pathological tumor response to preoperative chemotherapy in patients with CRLM, and may help to decide the individualized treatment strategy.
Collapse
Affiliation(s)
- Li Liu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Ultrasound, Peking University Cancer Hospital & Institute, Beijing, China
| | - Wen-Chao Cui
- Department of Ultrasonography, Shengli Oil Field Center Hospital, Dongying, Shandong Province, China
| | - Yu Sun
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Pathology, Peking University Cancer Hospital & Institute, Beijing, China
| | - Hong Wang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Ultrasound, Peking University Cancer Hospital & Institute, Beijing, China
| | - Zi-Nan Liang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Ultrasound, Peking University Cancer Hospital & Institute, Beijing, China
| | - Wei Wu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Ultrasound, Peking University Cancer Hospital & Institute, Beijing, China
| | - Kun Yan
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Ultrasound, Peking University Cancer Hospital & Institute, Beijing, China
| | - Yong-Li Ji
- Department of Ultrasonography, Shengli Oil Field Center Hospital, Dongying, Shandong Province, China
| | - Liang Dong
- Department of Ultrasonography, Shengli Oil Field Center Hospital, Dongying, Shandong Province, China
| | - Wei Yang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Ultrasound, Peking University Cancer Hospital & Institute, Beijing, China.
| |
Collapse
|
2
|
Chiu SH, Li HC, Chang WC, Wu CC, Lin HH, Lo CH, Chang PY. Improving the prediction of patient survival with the aid of residual convolutional neural network (ResNet) in colorectal cancer with unresectable liver metastases treated with bevacizumab-based chemotherapy. Cancer Imaging 2024; 24:165. [PMID: 39696483 DOI: 10.1186/s40644-024-00809-1] [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: 03/13/2024] [Accepted: 11/25/2024] [Indexed: 12/20/2024] Open
Abstract
BACKGROUND To verify overall survival predictions made with residual convolutional neural network-determined morphological response (ResNet-MR) in patients with unresectable synchronous liver-only metastatic colorectal cancer (mCRC) treated with bevacizumab-based chemotherapy (BBC). METHODS A retrospective review of liver-only mCRC patients treated with BBC from December 2011 to Apr 2021 was performed. Patients who had metachronous liver metastases or received locoregional treatment before the initiation of BBC were excluded. The percentage of downstaging to curative treatment and overall survival (OS) were recorded. Two abdominal radiologists evaluated portal venous phase CT images based on the morphological criteria and divided the images into Groups 1, 2, and 3. These images were used to establish the radiologists-determined morphological response (RD-MR), which classified patients into responders and non-responders based on the morphological change 3 months after the initiation of BBC. Then, the Group 1 and 3 images classified by the radiologists were input into ResNet as the training dataset. The trained ResNet then redivided the Group 2 images into Groups 1, 2 and 3. The ResNet-MR was determined on the basis of these redivided images and the initial Group 1 and 3 images classified by the radiologists. RESULTS Eighty-four patients were included in this study (53 males and 31 females, with a median age of 60.0 years). The follow-up time ranged from 10 to 86 months. A total of 407 CT images were input into ResNet as the training dataset. Both RD-MR and ResNet-MR correlated with OS (p value = 0.0167 and 0.0225, respectively). Regarding discriminatory ability for mortality, ResNet-MR had higher area under curve than RD-MR at both 1 year and 2 years and showed a significant difference in discriminatory ability (p-value = 0.0321) at 2 years. RD-MR classified 28 patients (33.3%) as responders, and ResNet-MR classified an additional 16 patients (19.0%) as responders; these 16 patients had longer OS than the remaining non-responders in the RD-MR group (27.49 versus 21.20 months, p value = 0.043) and had a higher percentage of downstaging (37.5% versus 17.5%, p value = 0.1610). CONCLUSIONS In CRC patients with liver metastases treated with BBC, prediction of survival can be improved with the aid of ResNet, enabling optimized individualized treatment.
Collapse
Affiliation(s)
- Sung-Hua Chiu
- Department of Electrical Engineering, National Taipei University of Technology, Taipei, Taiwan
- Department of Radiology, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Hsiao-Chi Li
- Department of Electrical Engineering, National Taipei University of Technology, Taipei, Taiwan
| | - Wei-Chou Chang
- Department of Radiology, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Chao-Cheng Wu
- Department of Electrical Engineering, National Taipei University of Technology, Taipei, Taiwan
| | - Hsuan-Hwai Lin
- Division of Gastroenterology, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Cheng-Hsiang Lo
- Department of Radiotherapy, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Ping-Ying Chang
- Division of Hematology/Oncology, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan.
| |
Collapse
|
3
|
Xu Z, Jiang G, Dai J. Tumor therapeutics in the era of "RECIST": past, current insights, and future prospects. Oncol Rev 2024; 18:1435922. [PMID: 39493769 PMCID: PMC11527623 DOI: 10.3389/or.2024.1435922] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Accepted: 08/30/2024] [Indexed: 11/05/2024] Open
Abstract
In recent years, advancements in medical treatment and imaging technologies have revolutionized the assessment of tumor response. However, the Response Evaluation Criteria in Solid Tumors (RECIST) has long been established as the gold standard for evaluating tumor treatment. As treatment modalities evolve, the need for continuous refinement and adaptation of RECIST becomes increasingly apparent. This review explores the historical evolution, current applications, limitations, and future directions of RECIST. It discusses the challenges of distinguishing true progression from pseudo-progression in ICIs (immune checkpoint inhibitors), the integration of advanced imaging tools, and the necessity for RECIST criteria tailored to specific therapies like neoadjuvant treatments. The review highlights the ongoing efforts to enhance RECIST's accuracy and reliability in clinical decision-making and the potential for developing new standards to better evaluate treatment efficacy in the rapidly evolving landscape of oncology.
Collapse
Affiliation(s)
| | - Gening Jiang
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Jie Dai
- *Correspondence: Gening Jiang, ; Jie Dai,
| |
Collapse
|
4
|
Bektaş M, Chia CM, Burchell GL, Daams F, Bonjer HJ, van der Peet DL. Artificial intelligence-aided ultrasound imaging in hepatopancreatobiliary surgery: where are we now? Surg Endosc 2024; 38:4869-4879. [PMID: 39160306 PMCID: PMC11362182 DOI: 10.1007/s00464-024-11130-0] [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: 04/02/2024] [Accepted: 07/28/2024] [Indexed: 08/21/2024]
Abstract
BACKGROUND Artificial intelligence (AI) models have been applied in various medical imaging modalities and surgical disciplines, however the current status and progress of ultrasound-based AI models within hepatopancreatobiliary surgery have not been evaluated in literature. Therefore, this review aimed to provide an overview of ultrasound-based AI models used for hepatopancreatobiliary surgery, evaluating current advancements, validation, and predictive accuracies. METHOD Databases PubMed, EMBASE, Cochrane, and Web of Science were searched for studies using AI models on ultrasound for patients undergoing hepatopancreatobiliary surgery. To be eligible for inclusion, studies needed to apply AI methods on ultrasound imaging for patients undergoing hepatopancreatobiliary surgery. The Probast risk of bias tool was used to evaluate the methodological quality of AI methods. RESULTS AI models have been primarily used within hepatopancreatobiliary surgery, to predict tumor recurrence, differentiate between tumoral tissues, and identify lesions during ultrasound imaging. Most studies have combined radiomics with convolutional neural networks, with AUCs up to 0.98. CONCLUSION Ultrasound-based AI models have demonstrated promising accuracies in predicting early tumoral recurrence and even differentiating between tumoral tissue types during and after hepatopancreatobiliary surgery. However, prospective studies are required to evaluate if these results will remain consistent and externally valid.
Collapse
Affiliation(s)
- Mustafa Bektaş
- Amsterdam UMC Location Vrije Universiteit Amsterdam, Surgery, De Boelelaan 1117, Amsterdam, The Netherlands.
| | - Catherine M Chia
- Department of Computer Science, Vrije Universiteit Amsterdam, De Boelelaan 1105, Amsterdam, The Netherlands
| | - George L Burchell
- Amsterdam UMC Location Vrije Universiteit Amsterdam, Medical Library, De Boelelaan 1117, Amsterdam, The Netherlands
| | - Freek Daams
- Amsterdam UMC Location Vrije Universiteit Amsterdam, Surgery, De Boelelaan 1117, Amsterdam, The Netherlands
| | - H Jaap Bonjer
- Amsterdam UMC Location Vrije Universiteit Amsterdam, Surgery, De Boelelaan 1117, Amsterdam, The Netherlands
| | - Donald L van der Peet
- Amsterdam UMC Location Vrije Universiteit Amsterdam, Surgery, De Boelelaan 1117, Amsterdam, The Netherlands
| |
Collapse
|
5
|
Liu M, Bao Q, Zhao T, Huang L, Zhang D, Wang Y, Yan X, Wang H, Jin K, Liu W, Wang K, Xing B. Pre-hepatectomy dynamic circulating tumor DNA to predict pathologic response to preoperative chemotherapy and post-hepatectomy recurrence in patients with colorectal liver metastases. Hepatol Int 2024; 18:1029-1039. [PMID: 38427145 DOI: 10.1007/s12072-023-10628-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Accepted: 12/15/2023] [Indexed: 03/02/2024]
Abstract
OBJECTIVE To evaluate the predictive value of pre-hepatectomy dynamic circulating tumor DNA (ctDNA) on pathologic response to preoperative chemotherapy and recurrence after liver resection for colorectal liver metastases (CRLM). BACKGROUND Pathologic response is a predictor of clinical outcomes for patients undergoing hepatectomy for CRLM. Postoperative ctDNA has been proven to be sensitive for recurrence detection. However, few studies investigate the impact of pre-hepatectomy ctDNA on pathologic response and recurrence. METHODS Patients with potential resectable CRLM underwent preoperative chemotherapy and hepatectomy between 2018 and 2021 was considered for inclusion. Plasma ctDNA was collected before and after preoperative chemotherapy. Pathologic response was analyzed for all patients after liver resection. Recurrence free survival was compared between patients with different ctDNA status and different pathologic response. The relation between ctDNA and pathologic response was also analyzed. RESULTS A total of 114 patients were included. ctDNA was detectable in 108 of 114 patients (94.7%) before chemotherapy, in 56 of 114 patients (49.1%) after chemotherapy. Patients with ctDNA positive at baseline and negative after chemotherapy had significantly longer RFS (median RFS 17 vs 7 months, p = 0.001) and HRFS (median HRFS unreached vs 8 months, p < 0.001) than those with ctDNA persistently positive after chemotherapy. Two patients (1.6%) had a pathologic complete response and 56 patients (45.2%) had a pathologic major response. Post-chemotherapy ctDNA- was associated with improved major pathologic response (53.4% vs 32.1%, p = 0.011). In the multivariable analysis, ctDNA- after chemotherapy (HR 0.51, 95% CI 0.28-0.93), major pathologic response (HR 0.34, 95% CI 0.19-0.62) and surgery combined with radiofrequency ablation (HR 2.62, 95% CI 1.38-5.00) were independently associated with RFS (all p < 0.05). CONCLUSIONS Pre-hepatectomy dynamic monitoring of ctDNA could predict pathologic response to preoperative chemotherapy and post-hepatectomy recurrence in CRLM patients. Negative ctDNA after preoperative chemotherapy was associated with better tumor regression grade and recurrence-free survival, which might be used to guide pre-hepatectomy chemotherapy and predict prognosis.
Collapse
Affiliation(s)
- Ming Liu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Hepato-Biliary-Pancreatic Surgery I, Peking University Cancer Hospital and Institute, No. 52, Fucheng Road, Haidian District, Beijing, People's Republic of China
| | - Quan Bao
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Hepato-Biliary-Pancreatic Surgery I, Peking University Cancer Hospital and Institute, No. 52, Fucheng Road, Haidian District, Beijing, People's Republic of China
| | - Tingting Zhao
- GloriousMed Clinical Laboratory (Shanghai) Co., Ltd., Shanghai, People's Republic of China
| | - Longfei Huang
- GloriousMed Clinical Laboratory (Shanghai) Co., Ltd., Shanghai, People's Republic of China
| | - Danhua Zhang
- GloriousMed Clinical Laboratory (Shanghai) Co., Ltd., Shanghai, People's Republic of China
| | - Yanyan Wang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Hepato-Biliary-Pancreatic Surgery I, Peking University Cancer Hospital and Institute, No. 52, Fucheng Road, Haidian District, Beijing, People's Republic of China
| | - Xiaoluan Yan
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Hepato-Biliary-Pancreatic Surgery I, Peking University Cancer Hospital and Institute, No. 52, Fucheng Road, Haidian District, Beijing, People's Republic of China
| | - Hongwei Wang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Hepato-Biliary-Pancreatic Surgery I, Peking University Cancer Hospital and Institute, No. 52, Fucheng Road, Haidian District, Beijing, People's Republic of China
| | - Kemin Jin
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Hepato-Biliary-Pancreatic Surgery I, Peking University Cancer Hospital and Institute, No. 52, Fucheng Road, Haidian District, Beijing, People's Republic of China
| | - Wei Liu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Hepato-Biliary-Pancreatic Surgery I, Peking University Cancer Hospital and Institute, No. 52, Fucheng Road, Haidian District, Beijing, People's Republic of China
| | - Kun Wang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Hepato-Biliary-Pancreatic Surgery I, Peking University Cancer Hospital and Institute, No. 52, Fucheng Road, Haidian District, Beijing, People's Republic of China.
| | - Baocai Xing
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Hepato-Biliary-Pancreatic Surgery I, Peking University Cancer Hospital and Institute, No. 52, Fucheng Road, Haidian District, Beijing, People's Republic of China.
| |
Collapse
|
6
|
Yoon S, Kim YJ, Jeon JS, Ahn SJ, Choi SJ. Radiomics and machine learning analysis of liver magnetic resonance imaging for prediction and early detection of tumor response in colorectal liver metastases. KOREAN JOURNAL OF CLINICAL ONCOLOGY 2024; 20:27-35. [PMID: 38988016 PMCID: PMC11261177 DOI: 10.14216/kjco.24005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 05/31/2024] [Accepted: 06/17/2024] [Indexed: 07/12/2024]
Abstract
PURPOSE The aim of this study was to demonstrate the effectiveness of a machine learning-based radiomics model for distinguishing tumor response and overall survival in patients with unresectable colorectal liver metastases (CRLM) treated with targeted biological therapy. METHODS We prospectively recruited 17 patients with unresectable liver metastases of colorectal cancer, who had been given targeted biological therapy as the first line of treatment. All patients underwent liver magnetic resonance imaging (MRI) three times up until 8 weeks after chemotherapy. We evaluated the diagnostic performance of machine learning-based radiomics model in tumor response of liver MRI compared with the guidelines for the Response Evaluation Criteria in Solid Tumors. Overall survival was evaluated using the Kaplan-Meier analysis and compared to the Cox proportional hazard ratios following univariate and multivariate analyses. RESULTS Performance measurement of the trained model through metrics showed the accuracy of the machine learning model to be 76.5%, and the area under the receiver operating characteristic curve was 0.857 (95% confidence interval [CI], 0.605-0.976; P < 0.001). For the patients classified as non-progressing or progressing by the radiomics model, the median overall survival was 17.5 months (95% CI, 12.8-22.2), and 14.8 months (95% CI, 14.2-15.4), respectively (P = 0.431, log-rank test). CONCLUSION Machine learning-based radiomics models could have the potential to predict tumor response in patients with unresectable CRLM treated with biologic therapy.
Collapse
Affiliation(s)
- Sungjin Yoon
- Department of Radiology, Gachon University Gil Medical Center, Gachon University College of Medicine, Incheon,
Korea
| | - Young Jae Kim
- Biomedical Engineering, Gachon University College of Medicine, Incheon,
Korea
| | - Ji Soo Jeon
- Biomedical Engineering, Gachon University College of Medicine, Incheon,
Korea
| | - Su Joa Ahn
- Department of Radiology, Gachon University Gil Medical Center, Gachon University College of Medicine, Incheon,
Korea
| | - Seung Joon Choi
- Department of Radiology, Gachon University Gil Medical Center, Gachon University College of Medicine, Incheon,
Korea
| |
Collapse
|
7
|
Petinrin OO, Saeed F, Toseef M, Liu Z, Basurra S, Muyide IO, Li X, Lin Q, Wong KC. Machine learning in metastatic cancer research: Potentials, possibilities, and prospects. Comput Struct Biotechnol J 2023; 21:2454-2470. [PMID: 37077177 PMCID: PMC10106342 DOI: 10.1016/j.csbj.2023.03.046] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Revised: 03/26/2023] [Accepted: 03/27/2023] [Indexed: 03/31/2023] Open
Abstract
Cancer has received extensive recognition for its high mortality rate, with metastatic cancer being the top cause of cancer-related deaths. Metastatic cancer involves the spread of the primary tumor to other body organs. As much as the early detection of cancer is essential, the timely detection of metastasis, the identification of biomarkers, and treatment choice are valuable for improving the quality of life for metastatic cancer patients. This study reviews the existing studies on classical machine learning (ML) and deep learning (DL) in metastatic cancer research. Since the majority of metastatic cancer research data are collected in the formats of PET/CT and MRI image data, deep learning techniques are heavily involved. However, its black-box nature and expensive computational cost are notable concerns. Furthermore, existing models could be overestimated for their generality due to the non-diverse population in clinical trial datasets. Therefore, research gaps are itemized; follow-up studies should be carried out on metastatic cancer using machine learning and deep learning tools with data in a symmetric manner.
Collapse
Affiliation(s)
| | - Faisal Saeed
- DAAI Research Group, Department of Computing and Data Science, School of Computing and Digital Technology, Birmingham City University, Birmingham B4 7XG, UK
| | - Muhammad Toseef
- Department of Computer Science, City University of Hong Kong, Kowloon Tong, Kowloon, Hong Kong SAR
| | - Zhe Liu
- Department of Computer Science, City University of Hong Kong, Kowloon Tong, Kowloon, Hong Kong SAR
| | - Shadi Basurra
- DAAI Research Group, Department of Computing and Data Science, School of Computing and Digital Technology, Birmingham City University, Birmingham B4 7XG, UK
| | | | - Xiangtao Li
- School of Artificial Intelligence, Jilin University, Jilin, China
| | - Qiuzhen Lin
- School of Computer Science and Software Engineering, Shenzhen University, Shenzhen, China
| | - Ka-Chun Wong
- Department of Computer Science, City University of Hong Kong, Kowloon Tong, Kowloon, Hong Kong SAR
- Hong Kong Institute for Data Science, City University of Hong Kong, Kowloon Tong, Kowloon, Hong Kong SAR
| |
Collapse
|
8
|
Qu H, Zhai H, Zhang S, Chen W, Zhong H, Cui X. Dynamic radiomics for predicting the efficacy of antiangiogenic therapy in colorectal liver metastases. Front Oncol 2023; 13:992096. [PMID: 36814812 PMCID: PMC9939899 DOI: 10.3389/fonc.2023.992096] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Accepted: 01/12/2023] [Indexed: 02/08/2023] Open
Abstract
Background and objective For patients with advanced colorectal liver metastases (CRLMs) receiving first-line anti-angiogenic therapy, an accurate, rapid and noninvasive indicator is urgently needed to predict its efficacy. In previous studies, dynamic radiomics predicted more accurately than conventional radiomics. Therefore, it is necessary to establish a dynamic radiomics efficacy prediction model for antiangiogenic therapy to provide more accurate guidance for clinical diagnosis and treatment decisions. Methods In this study, we use dynamic radiomics feature extraction method that extracts static features using tomographic images of different sequences of the same patient and then quantifies them into new dynamic features for the prediction of treatmentefficacy. In this retrospective study, we collected 76 patients who were diagnosed with unresectable CRLM between June 2016 and June 2021 in the First Hospital of China Medical University. All patients received standard treatment regimen of bevacizumab combined with chemotherapy in the first-line treatment, and contrast-enhanced abdominal CT (CECT) scans were performed before treatment. Patients with multiple primary lesions as well as missing clinical or imaging information were excluded. Area Under Curve (AUC) and accuracy were used to evaluate model performance. Regions of interest (ROIs) were independently delineated by two radiologists to extract radiomics features. Three machine learning algorithms were used to construct two scores based on the best response and progression-free survival (PFS). Results For the task that predict the best response patients will achieve after treatment, by using ROC curve analysis, it can be seen that the relative change rate (RCR) feature performed best among all features and best in linear discriminantanalysis (AUC: 0.945 and accuracy: 0.855). In terms of predicting PFS, the Kaplan-Meier plots suggested that the score constructed using the RCR features could significantly distinguish patients with good response from those with poor response (Two-sided P<0.0001 for survival analysis). Conclusions This study demonstrates that the application of dynamic radiomics features can better predict the efficacy of CRLM patients receiving antiangiogenic therapy compared with conventional radiomics features. It allows patients to have a more accurate assessment of the effect of medical treatment before receiving treatment, and this assessment method is noninvasive, rapid, and less expensive. Dynamic radiomics model provides stronger guidance for the selection of treatment options and precision medicine.
Collapse
Affiliation(s)
- Hui Qu
- College of Medicine and Biological Information Engineering, Northeastern University, Shenyang, Liaoning, P.R, China
| | - Huan Zhai
- Department of Interventional Radiology, First Affiliated Hospital of China Medical University, Shenyang, Liaoning, China,Key Laboratory of Diagnostic Imaging and Interventional Radiology of Liaoning Province, First Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| | - Shuairan Zhang
- Department of Gastroenterology, First Affiliated Hospital of China Medical University, Shenyang, China
| | - Wenjuan Chen
- Department of Medical Oncology, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| | - Hongshan Zhong
- Department of Interventional Radiology, First Affiliated Hospital of China Medical University, Shenyang, Liaoning, China,Key Laboratory of Diagnostic Imaging and Interventional Radiology of Liaoning Province, First Affiliated Hospital of China Medical University, Shenyang, Liaoning, China,*Correspondence: Xiaoyu Cui, ; Hongshan Zhong,
| | - Xiaoyu Cui
- College of Medicine and Biological Information Engineering, Northeastern University, Shenyang, Liaoning, P.R, China,Key Laboratory of Intelligent Computing in Medical Image, Ministry of Education, Shenyang, China,*Correspondence: Xiaoyu Cui, ; Hongshan Zhong,
| |
Collapse
|
9
|
Moawad AW, Morshid A, Khalaf AM, Elmohr MM, Hazle JD, Fuentes D, Badawy M, Kaseb AO, Hassan M, Mahvash A, Szklaruk J, Qayyum A, Abusaif A, Bennett WC, Nolan TS, Camp B, Elsayes KM. Multimodality annotated hepatocellular carcinoma data set including pre- and post-TACE with imaging segmentation. Sci Data 2023; 10:33. [PMID: 36653372 PMCID: PMC9849450 DOI: 10.1038/s41597-023-01928-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Accepted: 01/03/2023] [Indexed: 01/19/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is the most common primary liver neoplasm, and its incidence has doubled over the past two decades owing to increasing risk factors. Despite surveillance, most HCC cases are diagnosed at advanced stages and can only be treated using transarterial chemo-embolization (TACE) or systemic therapy. TACE failure may occur with incidence reaching up to 60% of cases, leaving patients with a financial and emotional burden. Radiomics has emerged as a new tool capable of predicting tumor response to TACE from pre-procedural computed tomography (CT) studies. This data report defines the HCC-TACE data collection of confirmed HCC patients who underwent TACE and have pre- and post-procedure CT imaging studies and available treatment outcomes (time-to-progression and overall survival). Clinically curated segmentation of pre-procedural CT studies was done for the purpose of algorithm training for prediction and automatic liver tumor segmentation.
Collapse
Affiliation(s)
- Ahmed W Moawad
- Departments of Imaging Physics, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA.
- Department of radiology, Mercy catholic medical center, Darby, PA, 19023, USA.
| | - Ali Morshid
- Departments of Imaging Physics, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA.
| | - Ahmed M Khalaf
- Departments of Imaging Physics, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Mohab M Elmohr
- Departments of Imaging Physics, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA.
- Department of radiology, Baylor college of medicine, TX, 77030, Houston, USA.
| | - John D Hazle
- Departments of Imaging Physics, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA.
| | - David Fuentes
- Departments of Imaging Physics, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA.
| | - Mohamed Badawy
- Departments of Imaging Physics, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Ahmed O Kaseb
- Departments of Gastrointestinal Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA.
| | - Manal Hassan
- Departments of Gastrointestinal Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA.
| | - Armeen Mahvash
- Departments of Interventional Radiology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Janio Szklaruk
- Departments of Body Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA.
| | - Aliyya Qayyum
- Departments of Body Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Abdelrahman Abusaif
- Departments of Body Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - William C Bennett
- Department of Biomedical Informatics, University of Arkansas for Medical Sciences, Little Rock, AR, 72205, USA.
| | - Tracy S Nolan
- Department of Biomedical Informatics, University of Arkansas for Medical Sciences, Little Rock, AR, 72205, USA.
| | - Brittney Camp
- Department of Biomedical Informatics, University of Arkansas for Medical Sciences, Little Rock, AR, 72205, USA.
| | - Khaled M Elsayes
- Departments of Body Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA.
| |
Collapse
|
10
|
Su X, Zhang H, Wang Y. A predictive model for early therapeutic efficacy of colorectal liver metastases using multimodal MRI data. JOURNAL OF X-RAY SCIENCE AND TECHNOLOGY 2023; 31:357-372. [PMID: 36591694 DOI: 10.3233/xst-221317] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
BACKGROUND Liver metastases is a pivotal factor of death in patients with colorectal cancer. The longitudinal data of colorectal liver metastases (CRLM) during treatment can monitor and reflect treatment efficacy and outcomes. OBJECTIVE The objective of this study is to establish a radiomic model based on longitudinal magnetic resonance imaging (MRI) to predict chemotherapy response in patients with CRLM. METHODS This study retrospectively enrolled longitudinal MRI data of five modalities on 100 patients. According to Response Evaluation Criteria in Solid Tumors (RECIST 1.1), 42 and 58 patients were identified as responders and non-responders, respectively. First, radiomic features were computed from different modalities of image data acquired pre-treatment and early-treatment, as well as their differences (Δ). Next, the features were screened by a two-sample t-test, max-relevance and min-redundancy (mRMR), and least absolute shrinkage and selection operator (LASSO). Then, several ensemble radiomic models that integrate support vector machine (SVM), k-nearest neighbor (KNN), gradient boost decision tree (GBDT) and multi-layer perceptron (MLP) were established based on voting method to predict chemotherapy response. Data samples were divided into training and verification queues using a ratio of 8:2. Finally, we used the area under ROC curve (AUC) to evaluate model performance. RESULTS Using the ensemble model developed using featue differences (Δ) computed from the longitudinal apparent diffusion coefficient (ADC) images, AUC is 0.9007±0.0436 for the training cohort. Applying to the testing cohort, AUC is 0.8958 and overall accuracy is 0.9. CONCLUSIONS Study results demonstrate advantages and high performance of the ensemble radiomic model based on the radiomics feature difference of the longitudinal ADC images in predicting chemotherapy response, which has potential to assist treatment decision-making and improve clinical outcome.
Collapse
Affiliation(s)
- Xuan Su
- Institute of Medical Imaging Engineering, University of Shanghai for Science and Technology, Shanghai, China
| | - Huan Zhang
- Department of Radiology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yuanjun Wang
- Institute of Medical Imaging Engineering, University of Shanghai for Science and Technology, Shanghai, China
| |
Collapse
|
11
|
Horvat N, Miranda J, El Homsi M, Peoples JJ, Long NM, Simpson AL, Do RKG. A primer on texture analysis in abdominal radiology. Abdom Radiol (NY) 2022; 47:2972-2985. [PMID: 34825946 DOI: 10.1007/s00261-021-03359-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 11/16/2021] [Accepted: 11/17/2021] [Indexed: 01/18/2023]
Abstract
The number of publications on texture analysis (TA), radiomics, and radiogenomics has been growing exponentially, with abdominal radiologists aiming to build new prognostic or predictive biomarkers for a wide range of clinical applications including the use of oncological imaging to advance the field of precision medicine. TA is specifically concerned with the study of the variation of pixel intensity values in radiological images. Radiologists aim to capture pixel variation in radiological images to deliver new insights into tumor biology that cannot be derived from visual inspection alone. TA remains an active area of investigation and requires further standardization prior to its clinical acceptance and applicability. This review is for radiologists interested in this rapidly evolving field, who are thinking of performing research or want to better interpret results in this arena. We will review the main concepts in TA, workflow processes, and existing challenges and steps to overcome them, as well as look at publications in body imaging with external validation.
Collapse
Affiliation(s)
- Natally Horvat
- Department of Radiology, Memorial Sloan Kettering Cancer Center, 1275 York Ave, New York, NY, 10065, USA
| | - Joao Miranda
- Department of Radiology, University of Sao Paulo, Sao Paulo, SP, Brazil
| | - Maria El Homsi
- Department of Radiology, Memorial Sloan Kettering Cancer Center, 1275 York Ave, New York, NY, 10065, USA
| | - Jacob J Peoples
- School of Computing, Queen's University, Kingston, ON, Canada
| | - Niamh M Long
- Department of Radiology, Memorial Sloan Kettering Cancer Center, 1275 York Ave, New York, NY, 10065, USA
| | - Amber L Simpson
- School of Computing, Queen's University, Kingston, ON, Canada.,Department of Biomedical and Molecular Sciences, Queen's University, Kingston, ON, Canada
| | - Richard K G Do
- Department of Radiology, Memorial Sloan Kettering Cancer Center, 1275 York Ave, New York, NY, 10065, USA.
| |
Collapse
|
12
|
Russo V, Lallo E, Munnia A, Spedicato M, Messerini L, D’Aurizio R, Ceroni EG, Brunelli G, Galvano A, Russo A, Landini I, Nobili S, Ceppi M, Bruzzone M, Cianchi F, Staderini F, Roselli M, Riondino S, Ferroni P, Guadagni F, Mini E, Peluso M. Artificial Intelligence Predictive Models of Response to Cytotoxic Chemotherapy Alone or Combined to Targeted Therapy for Metastatic Colorectal Cancer Patients: A Systematic Review and Meta-Analysis. Cancers (Basel) 2022; 14:4012. [PMID: 36011003 PMCID: PMC9406544 DOI: 10.3390/cancers14164012] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 07/26/2022] [Accepted: 08/12/2022] [Indexed: 12/24/2022] Open
Abstract
Tailored treatments for metastatic colorectal cancer (mCRC) have not yet completely evolved due to the variety in response to drugs. Therefore, artificial intelligence has been recently used to develop prognostic and predictive models of treatment response (either activity/efficacy or toxicity) to aid in clinical decision making. In this systematic review, we have examined the ability of learning methods to predict response to chemotherapy alone or combined with targeted therapy in mCRC patients by targeting specific narrative publications in Medline up to April 2022 to identify appropriate original scientific articles. After the literature search, 26 original articles met inclusion and exclusion criteria and were included in the study. Our results show that all investigations conducted on this field have provided generally promising results in predicting the response to therapy or toxic side-effects. By a meta-analytic approach we found that the overall weighted means of the area under the receiver operating characteristic (ROC) curve (AUC) were 0.90, 95% C.I. 0.80-0.95 and 0.83, 95% C.I. 0.74-0.89 in training and validation sets, respectively, indicating a good classification performance in discriminating response vs. non-response. The calculation of overall HR indicates that learning models have strong ability to predict improved survival. Lastly, the delta-radiomics and the 74 gene signatures were able to discriminate response vs. non-response by correctly identifying up to 99% of mCRC patients who were responders and up to 100% of patients who were non-responders. Specifically, when we evaluated the predictive models with tests reaching 80% sensitivity (SE) and 90% specificity (SP), the delta radiomics showed an SE of 99% and an SP of 94% in the training set and an SE of 85% and SP of 92 in the test set, whereas for the 74 gene signatures the SE was 97.6% and the SP 100% in the training set.
Collapse
Affiliation(s)
- Valentina Russo
- Research and Development Branch, Regional Cancer Prevention Laboratory, ISPRO-Study, Prevention and Oncology Network Institute, 50139 Florence, Italy
| | - Eleonora Lallo
- Research and Development Branch, Regional Cancer Prevention Laboratory, ISPRO-Study, Prevention and Oncology Network Institute, 50139 Florence, Italy
| | - Armelle Munnia
- Research and Development Branch, Regional Cancer Prevention Laboratory, ISPRO-Study, Prevention and Oncology Network Institute, 50139 Florence, Italy
| | - Miriana Spedicato
- Research and Development Branch, Regional Cancer Prevention Laboratory, ISPRO-Study, Prevention and Oncology Network Institute, 50139 Florence, Italy
| | - Luca Messerini
- Department of Experimental and Clinical Medicine, University of Florence, 50134 Florence, Italy
| | - Romina D’Aurizio
- Institute of Informatics and Telematics, National Research Council, 56124 Pisa, Italy
| | - Elia Giuseppe Ceroni
- Institute of Informatics and Telematics, National Research Council, 56124 Pisa, Italy
| | - Giulia Brunelli
- Institute of Informatics and Telematics, National Research Council, 56124 Pisa, Italy
| | - Antonio Galvano
- Department of Surgical, Oncological and Oral Sciences, University of Palermo, 90127 Palermo, Italy
| | - Antonio Russo
- Department of Surgical, Oncological and Oral Sciences, University of Palermo, 90127 Palermo, Italy
| | - Ida Landini
- Department of Health Sciences, University of Florence, 50139 Florence, Italy
| | - Stefania Nobili
- Department of Neurosciences, Imaging and Clinical Sciences, “G. D’Annunzio” Chieti-Pescara, 66100 Chieti, Italy
| | - Marcello Ceppi
- Clinical Epidemiology Unit, IRCCS-Ospedale Policlinico San Martino, 16131 Genova, Italy
| | - Marco Bruzzone
- Clinical Epidemiology Unit, IRCCS-Ospedale Policlinico San Martino, 16131 Genova, Italy
| | - Fabio Cianchi
- Department of Experimental and Clinical Medicine, University of Florence, 50134 Florence, Italy
| | - Fabio Staderini
- Department of Experimental and Clinical Medicine, University of Florence, 50134 Florence, Italy
| | - Mario Roselli
- Medical Oncology Unit, Department of Systems Medicine, Tor Vergata University, 00133 Rome, Italy
| | - Silvia Riondino
- Medical Oncology Unit, Department of Systems Medicine, Tor Vergata University, 00133 Rome, Italy
| | - Patrizia Ferroni
- BioBIM (InterInstitutional Multidisciplinary Biobank), IRCCS San Raffaele Roma, 00166 Rome, Italy
- Department of Human Sciences & Quality of Life Promotion, San Raffaele Roma Open University, 00166 Rome, Italy
| | - Fiorella Guadagni
- BioBIM (InterInstitutional Multidisciplinary Biobank), IRCCS San Raffaele Roma, 00166 Rome, Italy
- Department of Human Sciences & Quality of Life Promotion, San Raffaele Roma Open University, 00166 Rome, Italy
| | - Enrico Mini
- Department of Health Sciences, University of Florence, 50139 Florence, Italy
| | - Marco Peluso
- Research and Development Branch, Regional Cancer Prevention Laboratory, ISPRO-Study, Prevention and Oncology Network Institute, 50139 Florence, Italy
| |
Collapse
|
13
|
Al Duhayyim M, Mengash HA, Marzouk R, Nour MK, Mahgoub H, Althukair F, Mohamed A. Hybrid Rider Optimization with Deep Learning Driven Biomedical Liver Cancer Detection and Classification. COMPUTATIONAL INTELLIGENCE AND NEUROSCIENCE 2022; 2022:6162445. [PMID: 35814569 PMCID: PMC9262480 DOI: 10.1155/2022/6162445] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Revised: 06/02/2022] [Accepted: 06/15/2022] [Indexed: 11/17/2022]
Abstract
Biomedical engineering is the application of the principles and problem-solving methods of engineering to biology along with medicine. Computation intelligence is the study of design of intelligent agents which are systems acting perceptively. The computation intelligence paradigm offers more advantages to the enhancement and maintenance of the field of biomedical engineering. Liver cancer is the major reason of mortality worldwide. Earlier-stage diagnosis and treatment might increase the survival rate of liver cancer patients. Manual recognition of the cancer tissue is a time-consuming and difficult task. Hence, a computer-aided diagnosis (CAD) is employed in decision making procedures for accurate diagnosis and effective treatment. In contrast to classical image-dependent "semantic" feature evaluation from human expertise, deep learning techniques could learn feature representation automatically from sample images using convolutional neural network (CNN). This study introduces a Hybrid Rider Optimization with Deep Learning Driven Biomedical Liver Cancer Detection and Classification (HRO-DLBLCC) model. The proposed HRO-DLBLCC model majorly focuses on the identification of liver cancer in the medical images. To do so, the proposed HRO-DLBLCC model employs preprocessing in two stages, namely, Gabor filtering (GF) based noise removal and watershed transform based segmentation. In addition, the proposed HRO-DLBLCC model involves NAdam optimizer with DenseNet-201 based feature extractor to generate an optimal set of feature vectors. Finally, the HRO algorithm with recurrent neural network-long short-term memory (RNN-LSTM) model is applied for liver cancer classification, in which the hyperparameters of the RNN-LSTM model are tuned by the use of HRO algorithm. The HRO-DLBLCC model is experimentally validated and compared with existing models. The experimental results assured the promising performance of the HRO-DLBLCC model over recent approaches.
Collapse
Affiliation(s)
- Mesfer Al Duhayyim
- Department of Computer Science, College of Sciences and Humanities-Aflaj, Prince Sattam Bin Abdulaziz University, Al-Kharj, Saudi Arabia
| | - Hanan Abdullah Mengash
- Department of Information Systems, College of Computer and Information Sciences, Princess Nourah Bint Abdulrahman University, P.O. Box 84428, Riyadh 11671, Saudi Arabia
| | - Radwa Marzouk
- Department of Information Systems, College of Computer and Information Sciences, Princess Nourah Bint Abdulrahman University, P.O. Box 84428, Riyadh 11671, Saudi Arabia
| | - Mohamed K Nour
- Department of Computer Sciences, College of Computing and Information System, Umm Al-Qura University, Mecca, Saudi Arabia
| | - Hany Mahgoub
- Department of Computer Science, College of Science & Art at Mahayel, King Khalid University, Abha, Saudi Arabia
- Department of Computer Science, Faculty of Computers and Information, Menoufia University, Shibin Al Kawm, Egypt
| | - Fahd Althukair
- Department of Electrical Engineering and Computer Sciences, College of Engineering, University of CA, Berkeley, USA
| | - Abdullah Mohamed
- Research Center, Future University in Egypt, New Cairo 11845, Egypt
| |
Collapse
|
14
|
Mazaheri Y, Thakur SB, Bitencourt AGV, Lo Gullo R, Hötker AM, Bates DDB, Akin O. Evaluation of cancer outcome assessment using MRI: A review of deep-learning methods. BJR Open 2022; 4:20210072. [PMID: 36105425 PMCID: PMC9459949 DOI: 10.1259/bjro.20210072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 06/01/2022] [Accepted: 06/06/2022] [Indexed: 11/13/2022] Open
Abstract
Accurate evaluation of tumor response to treatment is critical to allow personalized treatment regimens according to the predicted response and to support clinical trials investigating new therapeutic agents by providing them with an accurate response indicator. Recent advances in medical imaging, computer hardware, and machine-learning algorithms have resulted in the increased use of these tools in the field of medicine as a whole and specifically in cancer imaging for detection and characterization of malignant lesions, prognosis, and assessment of treatment response. Among the currently available imaging techniques, magnetic resonance imaging (MRI) plays an important role in the evaluation of treatment assessment of many cancers, given its superior soft-tissue contrast and its ability to allow multiplanar imaging and functional evaluation. In recent years, deep learning (DL) has become an active area of research, paving the way for computer-assisted clinical and radiological decision support. DL can uncover associations between imaging features that cannot be visually identified by the naked eye and pertinent clinical outcomes. The aim of this review is to highlight the use of DL in the evaluation of tumor response assessed on MRI. In this review, we will first provide an overview of common DL architectures used in medical imaging research in general. Then, we will review the studies to date that have applied DL to magnetic resonance imaging for the task of treatment response assessment. Finally, we will discuss the challenges and opportunities of using DL within the clinical workflow.
Collapse
Affiliation(s)
| | | | | | - Roberto Lo Gullo
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, United States
| | - Andreas M. Hötker
- Institute of Diagnostic and Interventional Radiology, University Hospital Zurich, Zurich, Switzerland
| | - David D B Bates
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, United States
| | - Oguz Akin
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, United States
| |
Collapse
|
15
|
A Survey on Deep Learning for Precision Oncology. Diagnostics (Basel) 2022; 12:diagnostics12061489. [PMID: 35741298 PMCID: PMC9222056 DOI: 10.3390/diagnostics12061489] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 06/14/2022] [Accepted: 06/14/2022] [Indexed: 12/27/2022] Open
Abstract
Precision oncology, which ensures optimized cancer treatment tailored to the unique biology of a patient’s disease, has rapidly developed and is of great clinical importance. Deep learning has become the main method for precision oncology. This paper summarizes the recent deep-learning approaches relevant to precision oncology and reviews over 150 articles within the last six years. First, we survey the deep-learning approaches categorized by various precision oncology tasks, including the estimation of dose distribution for treatment planning, survival analysis and risk estimation after treatment, prediction of treatment response, and patient selection for treatment planning. Secondly, we provide an overview of the studies per anatomical area, including the brain, bladder, breast, bone, cervix, esophagus, gastric, head and neck, kidneys, liver, lung, pancreas, pelvis, prostate, and rectum. Finally, we highlight the challenges and discuss potential solutions for future research directions.
Collapse
|
16
|
Predicting survival for hepatic arterial infusion chemotherapy of unresectable colorectal liver metastases: Radiomics analysis of pretreatment computed tomography. J Transl Int Med 2022; 10:56-64. [PMID: 35702189 PMCID: PMC8997799 DOI: 10.2478/jtim-2022-0004] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
Objective Hepatic arterial infusion chemotherapy (HAIC) is an effective treatment for advanced unresectable colorectal cancer liver metastases (CRLM). This study was conducted to predict the efficacy of HAIC in patients with unresectable CRLM by radiomics methods based on pretreatment computed tomography (CT) examinations and clinical data. Materials and Methods A total of 63 patients were included in this study (41 in the training group and 22 in the validation group). All these patients underwent CT examination before HAIC. During the follow-up period, CT scans and laboratory examinations were performed regularly. Eighty-five radiological features were extracted from the regions of interest (ROIs) of CT images using the PyRadiomics program. The t-test and correlation were applied to select features. These features were analyzed using LASSO-Cox regression, and a linear model was developed to predict overall survival (OS). Results After reducing features by t-test and correlation test, seven features remained. After LASSO-Cox cross-validation, four features remained at λ = 0.232. They were gray level co-occurrence matrix (GLCM), gray level run length matrix (GLRLM), neighborhood gray tone difference matrix (NGTDM), and the location of the primary tumor. The C-index was 0.758 in the training group and 0.743 in the test group. Nomograms predicting 1-, 2-, and 3-year survival were established. Conclusion Our study demonstrates that a radiomics approach based on pretreatment CT texture analysis has the ability to predict early the outcome of HAIC in patients with advanced unresectable colorectal cancer with a high degree of accuracy and feasibility.
Collapse
|
17
|
Lu L, Dercle L, Zhao B, Schwartz LH. Deep learning for the prediction of early on-treatment response in metastatic colorectal cancer from serial medical imaging. Nat Commun 2021; 12:6654. [PMID: 34789774 PMCID: PMC8599694 DOI: 10.1038/s41467-021-26990-6] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2021] [Accepted: 10/21/2021] [Indexed: 12/23/2022] Open
Abstract
In current clinical practice, tumor response assessment is usually based on tumor size change on serial computerized tomography (CT) scan images. However, evaluation of tumor response to anti-vascular endothelial growth factor therapies in metastatic colorectal cancer (mCRC) is limited because morphological change in tumor may occur earlier than tumor size change. Here we present an analysis utilizing a deep learning (DL) network to characterize tumor morphological change for response assessment in mCRC patients. We retrospectively analyzed 1,028 mCRC patients who were prospectively included in the VELOUR trial (NCT00561470). We found that DL network was able to predict early on-treatment response in mCRC and showed better performance than its size-based counterpart with C-Index: 0.649 (95% CI: 0.619,0.679) vs. 0.627 (95% CI: 0.567,0.638), p = 0.009, z-test. The integration of DL network with size-based methodology could further improve the prediction performance to C-Index: 0.694 (95% CI: 0.661,0.720), which was superior to size/DL-based-only models (all p < 0.001, z-test). Our study suggests that DL network could provide a noninvasive mean for quantitative and comprehensive characterization of tumor morphological change, which may potentially benefit personalized early on-treatment decision making.
Collapse
Affiliation(s)
- Lin Lu
- Department of Radiology, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - Laurent Dercle
- Department of Radiology, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - Binsheng Zhao
- Department of Radiology, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - Lawrence H Schwartz
- Department of Radiology, Columbia University Irving Medical Center, New York, NY, 10032, USA.
| |
Collapse
|
18
|
Javaid A, Shahab O, Adorno W, Fernandes P, May E, Syed S. Machine Learning Predictive Outcomes Modeling in Inflammatory Bowel Diseases. Inflamm Bowel Dis 2021; 28:819-829. [PMID: 34417815 PMCID: PMC9165557 DOI: 10.1093/ibd/izab187] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Indexed: 12/14/2022]
Abstract
There is a rising interest in use of big data approaches to personalize treatment of inflammatory bowel diseases (IBDs) and to predict and prevent outcomes such as disease flares and therapeutic nonresponse. Machine learning (ML) provides an avenue to identify and quantify features across vast quantities of data to produce novel insights in disease management. In this review, we cover current approaches in ML-driven predictive outcomes modeling for IBD and relate how advances in other fields of medicine may be applied to improve future IBD predictive models. Numerous studies have incorporated clinical, laboratory, or omics data to predict significant outcomes in IBD, including hospitalizations, outpatient corticosteroid use, biologic response, and refractory disease after colectomy, among others, with considerable health care dollars saved as a result. Encouraging results in other fields of medicine support efforts to use ML image analysis-including analysis of histopathology, endoscopy, and radiology-to further advance outcome predictions in IBD. Though obstacles to clinical implementation include technical barriers, bias within data sets, and incongruence between limited data sets preventing model validation in larger cohorts, ML-predictive analytics have the potential to transform the clinical management of IBD. Future directions include the development of models that synthesize all aforementioned approaches to produce more robust predictive metrics.
Collapse
Affiliation(s)
- Aamir Javaid
- Division of Pediatric Gastroenterology and Hepatology, Department of Pediatrics, University of Virginia, Charlottesville, VA, USA
| | - Omer Shahab
- Division of Gastroenterology and Hepatology, Department of Medicine, Virginia Commonwealth University, Richmond, VA, USA
| | - William Adorno
- School of Data Science, University of Virginia, Charlottesville, VA, USA
| | - Philip Fernandes
- Division of Pediatric Gastroenterology and Hepatology, Department of Pediatrics, University of Virginia, Charlottesville, VA, USA
| | - Eve May
- Division of Gastroenterology and Hepatology, Department of Pediatrics, Children’s National Hospital, Washington, DC, USA
| | - Sana Syed
- Division of Pediatric Gastroenterology and Hepatology, Department of Pediatrics, University of Virginia, Charlottesville, VA, USA,School of Data Science, University of Virginia, Charlottesville, VA, USA,Address Correspondence to: Sana Syed, MD, MSCR, MSDS, Division of Pediatric Gastroenterology and Hepatology, Department of Pediatrics, University of Virginia, 409 Lane Rd, Room 2035B, Charlottesville, VA, 22908, USA ()
| |
Collapse
|
19
|
Zhang Y, Lv X, Qiu J, Zhang B, Zhang L, Fang J, Li M, Chen L, Wang F, Liu S, Zhang S. Deep Learning With 3D Convolutional Neural Network for Noninvasive Prediction of Microvascular Invasion in Hepatocellular Carcinoma. J Magn Reson Imaging 2021; 54:134-143. [PMID: 33559293 DOI: 10.1002/jmri.27538] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Revised: 01/13/2021] [Accepted: 01/16/2021] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Microvascular invasion (MVI) is a critical prognostic factor of hepatocellular carcinoma (HCC). However, it could only be obtained by postoperative histological examination. PURPOSE To develop an end-to-end deep-learning models based on MRI images for preoperative prediction of MVI in HCC patients who underwent surgical resection. STUDY TYPE Retrospective. POPULATION Two hundred and thirty-seven patients with histologically confirmed HCC. FIELD STRENGTH 1.5 T and 3.0 T. SEQUENCE Axial T2 -weighted (T2 -w) with turbo spin echo sequence, T2 -Spectral Presaturation with Inversion Recovery (T2 -SPIR), and dynamic contrast-enhanced (DCE) imaging with fat suppressed enhanced T1 high-resolution isotropic volume examination. ASSESSMENT The patients were randomly divided into training (N = 158) and validation (N = 79) sets. Data augmentation by random rotation was performed on the training set and the sample size increased to 1940 for each MR sequence. A three-dimensional convolutional neural network (3D CNN) was used to develop four deep-learning models, including three single-layer models based on single-sequence, and fusion model combining three sequences. MVI status was obtained from the postoperative pathology reports. STATISTICAL TESTS The dice similarity coefficient (DSC) and Hausdorff distance (HD) were applied to assess the similarity and reproducibility between the manual segmentations of tumor from two radiologists. Receiver operating characteristic curve analysis was used to evaluate model performance. MVI was identified in 92 (38.8%) patients. Good reproducibility with interobserver DSCs of 0.90, 0.89, and 0.89 and HDs of 4.09, 3.67, and 3.60 was observed for PVP, T2 WI, and T2 -SPIR, respectively. The fusion model achieved an area under the curve (AUC) of 0.81, sensitivity of 69%, and specificity of 79% in the training set and 0.72, sensitivity of 55%, and specificity of 81% in the validation set. DATA CONCLUSION 3D CNN model may serve as a noninvasive tool to predict MVI in HCC, whereas its accuracy needs to be enhanced with larger cohort. LEVEL OF EVIDENCE 3 TECHNICAL EFFICACY: Stage 2.
Collapse
Affiliation(s)
- Yongxin Zhang
- Department of Radiology, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China.,Department of MR, Zhongshan City People's Hospital Affiliated to Sun Yat-sen University, Zhongshan, Guangdong, China
| | - Xiaofei Lv
- Department of Medical Imaging, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Jiliang Qiu
- Department of Liver Surgery, State Key Laboratory of Oncology in South China, Collaborative Innovation Centre for Cancer Medicine, Sun Yat-sen University Cancer Centre, Guangzhou, China
| | - Bin Zhang
- Department of Radiology, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China
| | - Lu Zhang
- Department of Radiology, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China
| | - Jin Fang
- Department of Radiology, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China
| | - Minmin Li
- Department of Radiology, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China
| | - Luyan Chen
- Department of Radiology, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China
| | - Fei Wang
- Department of Radiology, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China
| | - Shuyi Liu
- Department of Radiology, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China
| | - Shuixing Zhang
- Department of Radiology, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China
| |
Collapse
|