1
|
Song Y, Liu S, Liu X, Jia H, Shi H, Liu X, Hao D, Wang H, Xing X. An Integrated Radiopathomics Machine Learning Model to Predict Pathological Response to Preoperative Chemotherapy in Gastric Cancer. Acad Radiol 2025; 32:134-145. [PMID: 39214816 DOI: 10.1016/j.acra.2024.08.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 08/06/2024] [Accepted: 08/08/2024] [Indexed: 09/04/2024]
Abstract
RATIONALE AND OBJECTIVES Accurately predicting the pathological response to chemotherapy before treatment is important for selecting the appropriate treatment groups, formulating individualized treatment plans, and improving the survival rates of patients with gastric cancer (GC). METHODS We retrospectively enrolled 151 patients diagnosed with GC who underwent preoperative chemotherapy and surgical resection at the Affiliated Hospital of Qingdao University between January 2015 and June 2023. Both pretreatment-enhanced computer technology images and whole slide images of pathological hematoxylin and eosin-stained sections were available for each patient. The image features were extracted and used to construct an ensemble radiopathomics machine learning model. In addition, a nomogram was developed by combining the imaging features and clinical characteristics. RESULTS In total, 962 radiomics and 999 pathomics signatures were extracted from 106 patients in the training cohort. A fusion radiopathomics model was constructed using 13 radiomics and 5 pathomics signatures. The fusion model showed favorable performance compared to single-omics models, with an area under the curve (AUC) of 0.789 in the validation cohort. Moreover, a combined radiopathomics nomogram (RPN) was developed based on radiopathomics features and the Borrmann type, which is a classification method for advanced GC according to tumor growth pattern and gross morphology. The RPN showed superior predictive performance in the training (AUC 0.880) and validation cohorts (AUC 0.797). The decision curve analysis showed that RPN could provide favorable clinical benefits to patients with GC. CONCLUSIONS RPN was able to predict the pathological response to preoperative chemotherapy with high accuracy, and therefore provides a novel tool for personalized treatment of GC.
Collapse
Affiliation(s)
- Yaolin Song
- Department of Pathology, The Affiliated Hospital of Qingdao University, No.16 Jiangsu Road, Qingdao, China
| | - Shunli Liu
- Department of Radiology, The Affiliated Hospital of Qingdao University, No.16 Jiangsu Road, Qingdao, China
| | - Xinyu Liu
- Department of Pathology, The Affiliated Hospital of Qingdao University, No.16 Jiangsu Road, Qingdao, China
| | - Huiqing Jia
- Department of Pathology, The Affiliated Hospital of Qingdao University, No.16 Jiangsu Road, Qingdao, China
| | - Hailei Shi
- Department of Pathology, The Affiliated Hospital of Qingdao University, No.16 Jiangsu Road, Qingdao, China
| | - Xianglan Liu
- Department of Pathology, The Affiliated Hospital of Qingdao University, No.16 Jiangsu Road, Qingdao, China
| | - Dapeng Hao
- Department of Radiology, The Affiliated Hospital of Qingdao University, No.16 Jiangsu Road, Qingdao, China
| | - Hexiang Wang
- Department of Radiology, The Affiliated Hospital of Qingdao University, No.16 Jiangsu Road, Qingdao, China
| | - Xiaoming Xing
- Department of Pathology, The Affiliated Hospital of Qingdao University, No.16 Jiangsu Road, Qingdao, 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
|
Li Q, Teodoro G, Jiang Y, Kong J. NACNet: A histology context-aware transformer graph convolution network for predicting treatment response to neoadjuvant chemotherapy in Triple Negative Breast Cancer. Comput Med Imaging Graph 2024; 118:102467. [PMID: 39591709 DOI: 10.1016/j.compmedimag.2024.102467] [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: 06/18/2024] [Revised: 11/07/2024] [Accepted: 11/07/2024] [Indexed: 11/28/2024]
Abstract
Neoadjuvant chemotherapy (NAC) response prediction for triple negative breast cancer (TNBC) patients is a challenging task clinically as it requires understanding complex histology interactions within the tumor microenvironment (TME). Digital whole slide images (WSIs) capture detailed tissue information, but their giga-pixel size necessitates computational methods based on multiple instance learning, which typically analyze small, isolated image tiles without the spatial context of the TME. To address this limitation and incorporate TME spatial histology interactions in predicting NAC response for TNBC patients, we developed a histology context-aware transformer graph convolution network (NACNet). Our deep learning method identifies the histopathological labels on individual image tiles from WSIs, constructs a spatial TME graph, and represents each node with features derived from tissue texture and social network analysis. It predicts NAC response using a transformer graph convolution network model enhanced with graph isomorphism network layers. We evaluate our method with WSIs of a cohort of TNBC patient (N=105) and compared its performance with multiple state-of-the-art machine learning and deep learning models, including both graph and non-graph approaches. Our NACNet achieves 90.0% accuracy, 96.0% sensitivity, 88.0% specificity, and an AUC of 0.82, through eight-fold cross-validation, outperforming baseline models. These comprehensive experimental results suggest that NACNet holds strong potential for stratifying TNBC patients by NAC response, thereby helping to prevent overtreatment, improve patient quality of life, reduce treatment cost, and enhance clinical outcomes, marking an important advancement toward personalized breast cancer treatment.
Collapse
Affiliation(s)
- Qiang Li
- Department of Mathematics and Statistics, Georgia State University, Atlanta, 30303, GA, USA.
| | - George Teodoro
- Department of Computer Science, Federal University of Minas Gerais, Belo Horizonte, 31270, Minas Gerais, Brazil.
| | - Yi Jiang
- Department of Mathematics and Statistics, Georgia State University, Atlanta, 30303, GA, USA; Winship Cancer Institute, Emory University, Atlanta, 30322, GA, USA.
| | - Jun Kong
- Department of Mathematics and Statistics, Georgia State University, Atlanta, 30303, GA, USA; Department of Computer Science, Georgia State University, Atlanta, 30303, GA, USA; Winship Cancer Institute, Emory University, Atlanta, 30322, GA, USA.
| |
Collapse
|
4
|
Patharia P, Sethy PK, Nanthaamornphong A. Advancements and Challenges in the Image-Based Diagnosis of Lung and Colon Cancer: A Comprehensive Review. Cancer Inform 2024; 23:11769351241290608. [PMID: 39483315 PMCID: PMC11526153 DOI: 10.1177/11769351241290608] [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: 05/09/2024] [Accepted: 09/25/2024] [Indexed: 11/03/2024] Open
Abstract
Image-based diagnosis has become a crucial tool in the identification and management of various cancers, particularly lung and colon cancer. This review delves into the latest advancements and ongoing challenges in the field, with a focus on deep learning, machine learning, and image processing techniques applied to X-rays, CT scans, and histopathological images. Significant progress has been made in imaging technologies like computed tomography (CT), magnetic resonance imaging (MRI), and positron emission tomography (PET), which, when combined with machine learning and artificial intelligence (AI) methodologies, have greatly enhanced the accuracy of cancer detection and characterization. These advances have enabled early detection, more precise tumor localization, personalized treatment plans, and overall improved patient outcomes. However, despite these improvements, challenges persist. Variability in image interpretation, the lack of standardized diagnostic protocols, unequal access to advanced imaging technologies, and concerns over data privacy and security within AI-based systems remain major obstacles. Furthermore, integrating imaging data with broader clinical information is crucial to achieving a more comprehensive approach to cancer diagnosis and treatment. This review provides valuable insights into the recent developments and challenges in image-based diagnosis for lung and colon cancers, underscoring both the remarkable progress and the hurdles that still need to be overcome to optimize cancer care.
Collapse
Affiliation(s)
- Pragati Patharia
- Department of Electronics and Communication Engineering, Guru Ghasidas Vishwavidyalaya, Bilaspur, Chhattisgarh, India
| | - Prabira Kumar Sethy
- Department of Electronics and Communication Engineering, Guru Ghasidas Vishwavidyalaya, Bilaspur, Chhattisgarh, India
- Department of Electronics, Sambalpur University, Burla, Odisha, India
| | | |
Collapse
|
5
|
Chen S, Yu J, Chamouni S, Wang Y, Li Y. Integrating machine learning and artificial intelligence in life-course epidemiology: pathways to innovative public health solutions. BMC Med 2024; 22:354. [PMID: 39218895 PMCID: PMC11367811 DOI: 10.1186/s12916-024-03566-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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Accepted: 08/19/2024] [Indexed: 09/04/2024] Open
Abstract
The integration of machine learning (ML) and artificial intelligence (AI) techniques in life-course epidemiology offers remarkable opportunities to advance our understanding of the complex interplay between biological, social, and environmental factors that shape health trajectories across the lifespan. This perspective summarizes the current applications, discusses future potential and challenges, and provides recommendations for harnessing ML and AI technologies to develop innovative public health solutions. ML and AI have been increasingly applied in epidemiological studies, demonstrating their ability to handle large, complex datasets, identify intricate patterns and associations, integrate multiple and multimodal data types, improve predictive accuracy, and enhance causal inference methods. In life-course epidemiology, these techniques can help identify sensitive periods and critical windows for intervention, model complex interactions between risk factors, predict individual and population-level disease risk trajectories, and strengthen causal inference in observational studies. By leveraging the five principles of life-course research proposed by Elder and Shanahan-lifespan development, agency, time and place, timing, and linked lives-we discuss a framework for applying ML and AI to uncover novel insights and inform targeted interventions. However, the successful integration of these technologies faces challenges related to data quality, model interpretability, bias, privacy, and equity. To fully realize the potential of ML and AI in life-course epidemiology, fostering interdisciplinary collaborations, developing standardized guidelines, advocating for their integration in public health decision-making, prioritizing fairness, and investing in training and capacity building are essential. By responsibly harnessing the power of ML and AI, we can take significant steps towards creating healthier and more equitable futures across the life course.
Collapse
Affiliation(s)
- Shanquan Chen
- Faculty of Epidemiology and Population Health, London School of Hygiene & Tropical Medicine, Keppel Street, London, WC1E 7HT, UK.
| | - Jiazhou Yu
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Sarah Chamouni
- Faculty of Epidemiology and Population Health, London School of Hygiene & Tropical Medicine, Keppel Street, London, WC1E 7HT, UK
| | - Yuqi Wang
- Department of Computer Science, University College London, London, WC1E 6BT, UK
| | - Yunfei Li
- Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, 171 64, Sweden.
| |
Collapse
|
6
|
Li Z, Gao J, Zhou H, Li X, Zheng T, Lin F, Wang X, Chu T, Wang Q, Wang S, Cao K, Liang Y, Zhao F, Xie H, Xu C, Zhang H, Niu Q, Ma H, Mao N. Multiregional dynamic contrast-enhanced MRI-based integrated system for predicting pathological complete response of axillary lymph node to neoadjuvant chemotherapy in breast cancer: multicentre study. EBioMedicine 2024; 107:105311. [PMID: 39191174 PMCID: PMC11400626 DOI: 10.1016/j.ebiom.2024.105311] [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: 04/04/2024] [Revised: 08/11/2024] [Accepted: 08/12/2024] [Indexed: 08/29/2024] Open
Abstract
BACKGROUND The accurate evaluation of axillary lymph node (ALN) response to neoadjuvant chemotherapy (NAC) in breast cancer holds great value. This study aimed to develop an artificial intelligence system utilising multiregional dynamic contrast-enhanced MRI (DCE-MRI) and clinicopathological characteristics to predict axillary pathological complete response (pCR) after NAC in breast cancer. METHODS This study included retrospective and prospective datasets from six medical centres in China between May 2018 and December 2023. A fully automated integrated system based on deep learning (FAIS-DL) was built to perform tumour and ALN segmentation and axillary pCR prediction sequentially. The predictive performance of FAIS-DL was assessed using the area under the receiver operating characteristic curve (AUC), accuracy, sensitivity, and specificity. RNA sequencing analysis were conducted on 45 patients to explore the biological basis of FAIS-DL. FINDINGS 1145 patients (mean age, 50 years ±10 [SD]) were evaluated. Among these patients, 506 were in the training and validation sets (axillary pCR rate of 40.3%), 127 in the internal test set (axillary pCR rate of 37.8%), 414 in the pooled external test set (axillary pCR rate of 48.8%), and 98 in the prospective test set (axillary pCR rate of 43.9%). For predicting axillary pCR, FAIS-DL achieved AUCs of 0.95, 0.93, and 0.94 in the internal test set, pooled external test set, and prospective test set, respectively, which were also significantly higher than those of the clinical model and deep learning models based on single-regional DCE-MRI (all P < 0.05, DeLong test). In the pooled external and prospective test sets, the FAIS-DL decreased the unnecessary axillary lymph node dissection rate from 47.9% to 6.8%, and increased the benefit rate from 52.2% to 86.5%. RNA sequencing analysis revealed that high FAIS-DL scores were associated with the upregulation of immune-mediated genes and pathways. INTERPRETATION FAIS-DL has demonstrated satisfactory performance in predicting axillary pCR, which may guide the formulation of personalised treatment regimens for patients with breast cancer in clinical practice. FUNDING This study was supported by the National Natural Science Foundation of China (82371933), National Natural Science Foundation of Shandong Province of China (ZR2021MH120), Mount Taishan Scholars and Young Experts Program (tsqn202211378), Key Projects of China Medicine Education Association (2022KTM030), China Postdoctoral Science Foundation (314730), and Beijing Postdoctoral Research Foundation (2023-zz-012).
Collapse
Affiliation(s)
- Ziyin Li
- School of Medical Imaging, Binzhou Medical University, No. 346 Guanhai Road, Yantai, Shandong, 264003, China; Department of Radiology, Yantai Yuhuangding Hospital, Qingdao University, Yantai, Shandong, 264000, China
| | - Jing Gao
- Department of Radiology, Yantai Yuhuangding Hospital, Qingdao University, Yantai, Shandong, 264000, China
| | - Heng Zhou
- School of Information and Electronic Engineering, Shandong Technology and Business University, Yantai, Shandong, 264005, China
| | - Xianglin Li
- School of Medical Imaging, Binzhou Medical University, No. 346 Guanhai Road, Yantai, Shandong, 264003, China
| | - Tiantian Zheng
- School of Medical Imaging, Binzhou Medical University, No. 346 Guanhai Road, Yantai, Shandong, 264003, China; Department of Radiology, Yantai Yuhuangding Hospital, Qingdao University, Yantai, Shandong, 264000, China
| | - Fan Lin
- Department of Radiology, Yantai Yuhuangding Hospital, Qingdao University, Yantai, Shandong, 264000, China
| | - Xiaodong Wang
- School of Medical Imaging, Binzhou Medical University, No. 346 Guanhai Road, Yantai, Shandong, 264003, China; Department of Radiology, Yantai Yuhuangding Hospital, Qingdao University, Yantai, Shandong, 264000, China
| | - Tongpeng Chu
- Big Data and Artificial Intelligence Laboratory, Yantai Yuhuangding Hospital, Qingdao University, Yantai, Shandong, 264000, China; Shandong Provincial Key Medical and Health Laboratory of Intelligent Diagnosis and Treatment for Women's Diseases, Yantai Yuhuangding Hospital, Qingdao University, Yantai, Shandong, 264000, China
| | - Qi Wang
- Big Data and Artificial Intelligence Laboratory, Yantai Yuhuangding Hospital, Qingdao University, Yantai, Shandong, 264000, China; Shandong Provincial Key Medical and Health Laboratory of Intelligent Diagnosis and Treatment for Women's Diseases, Yantai Yuhuangding Hospital, Qingdao University, Yantai, Shandong, 264000, China
| | - Simin Wang
- Department of Radiology, Fudan University Cancer Center, Shanghai, 200433, China
| | - Kun Cao
- Department of Radiology, Beijing Cancer Hospital, Beijing, 100142, China
| | - Yun Liang
- Department of Radiology, Guilin Municipal Hospital of Traditional Chinese Medicine, Guilin, Yunnan, 541002, China
| | - Feng Zhao
- School of Computer Science and Technology, Shandong Technology and Business University, Yantai, Shandong, 264005, China
| | - Haizhu Xie
- Department of Radiology, Yantai Yuhuangding Hospital, Qingdao University, Yantai, Shandong, 264000, China
| | - Cong Xu
- Physical Examination Center, Yantai Yuhuangding Hospital, Qingdao University, Yantai, Shandong, 264000, China
| | - Haicheng Zhang
- Big Data and Artificial Intelligence Laboratory, Yantai Yuhuangding Hospital, Qingdao University, Yantai, Shandong, 264000, China; Shandong Provincial Key Medical and Health Laboratory of Intelligent Diagnosis and Treatment for Women's Diseases, Yantai Yuhuangding Hospital, Qingdao University, Yantai, Shandong, 264000, China
| | - Qingliang Niu
- Weifang NO.2 People's Hospital, Weifang, Shandong, 261041, China.
| | - Heng Ma
- Department of Radiology, Yantai Yuhuangding Hospital, Qingdao University, Yantai, Shandong, 264000, China.
| | - Ning Mao
- Department of Radiology, Yantai Yuhuangding Hospital, Qingdao University, Yantai, Shandong, 264000, China.
| |
Collapse
|
7
|
Jemaa S, Ounadjela S, Wang X, El-Galaly TC, Kostakoglu L, Knapp A, Ku G, Musick L, Sahin D, Wei MC, Yin S, Bengtsson T, De Crespigny A, Carano RA. Automated Lugano Metabolic Response Assessment in 18F-Fluorodeoxyglucose-Avid Non-Hodgkin Lymphoma With Deep Learning on 18F-Fluorodeoxyglucose-Positron Emission Tomography. J Clin Oncol 2024; 42:2966-2977. [PMID: 38843483 PMCID: PMC11361360 DOI: 10.1200/jco.23.01978] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 02/12/2024] [Accepted: 03/14/2024] [Indexed: 08/30/2024] Open
Abstract
PURPOSE Artificial intelligence can reduce the time used by physicians on radiological assessments. For 18F-fluorodeoxyglucose-avid lymphomas, obtaining complete metabolic response (CMR) by end of treatment is prognostic. METHODS Here, we present a deep learning-based algorithm for fully automated treatment response assessments according to the Lugano 2014 classification. The proposed four-stage method, trained on a multicountry clinical trial (ClinicalTrials.gov identifier: NCT01287741) and tested in three independent multicenter and multicountry test sets on different non-Hodgkin lymphoma subtypes and different lines of treatment (ClinicalTrials.gov identifiers NCT02257567, NCT02500407; 20% holdout in ClinicalTrials.gov identifier NCT01287741), outputs the detected lesions at baseline and follow-up to enable focused radiologist review. RESULTS The method's response assessment achieved high agreement with the adjudicated radiologic responses (eg, agreement for overall response assessment of 93%, 87%, and 85% in ClinicalTrials.gov identifiers NCT01287741, NCT02500407, and NCT02257567, respectively) similar to inter-radiologist agreement and was strongly prognostic of outcomes with a trend toward higher accuracy for death risk than adjudicated radiologic responses (hazard ratio for end of treatment by-model CMR of 0.123, 0.054, and 0.205 in ClinicalTrials.gov identifiers NCT01287741, NCT02500407, and NCT02257567, compared with, respectively, 0.226, 0.292, and 0.272 for CMR by the adjudicated responses). Furthermore, a radiologist review of the algorithm's assessments was conducted. The radiologist median review time was 1.38 minutes/assessment, and no statistically significant differences were observed in the level of agreement of the radiologist with the model's response compared with the level of agreement of the radiologist with the adjudicated responses. CONCLUSION These results suggest that the proposed method can be incorporated into radiologic response assessment workflows in cancer imaging for significant time savings and with performance similar to trained medical experts.
Collapse
Affiliation(s)
| | | | | | - Tarec C. El-Galaly
- Department of Hematology, Aalborg University Hospital, Aalborg, Denmark
- Hematology Research Unit, Department of Hematology, Odense University Hospital, Odense, Denmark
- Department of Clinical Research, University of Southern Denmark, Odense, Denmark
- Department of Medicine Solna, Clinical Epidemiology Division, Karolinska Institutet, Stockholm, Sweden
| | - Lale Kostakoglu
- Department of Radiology and Medical Imaging, University of Virginia, Charlottesville, VA
| | | | - Grace Ku
- Genentech, Inc, South San Francisco, CA
| | | | | | | | - Shen Yin
- Genentech, Inc, South San Francisco, CA
| | - Thomas Bengtsson
- Department of Statistics, University of California, Berkeley, CA
| | | | | |
Collapse
|
8
|
Riaz IB, Khan MA, Haddad TC. Potential application of artificial intelligence in cancer therapy. Curr Opin Oncol 2024; 36:437-448. [PMID: 39007164 DOI: 10.1097/cco.0000000000001068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/16/2024]
Abstract
PURPOSE OF REVIEW This review underscores the critical role and challenges associated with the widespread adoption of artificial intelligence in cancer care to enhance disease management, streamline clinical processes, optimize data retrieval of health information, and generate and synthesize evidence. RECENT FINDINGS Advancements in artificial intelligence models and the development of digital biomarkers and diagnostics are applicable across the cancer continuum from early detection to survivorship care. Additionally, generative artificial intelligence has promised to streamline clinical documentation and patient communications, generate structured data for clinical trial matching, automate cancer registries, and facilitate advanced clinical decision support. Widespread adoption of artificial intelligence has been slow because of concerns about data diversity and data shift, model reliability and algorithm bias, legal oversight, and high information technology and infrastructure costs. SUMMARY Artificial intelligence models have significant potential to transform cancer care. Efforts are underway to deploy artificial intelligence models in the cancer practice, evaluate their clinical impact, and enhance their fairness and explainability. Standardized guidelines for the ethical integration of artificial intelligence models in cancer care pathways and clinical operations are needed. Clear governance and oversight will be necessary to gain trust in artificial intelligence-assisted cancer care by clinicians, scientists, and patients.
Collapse
Affiliation(s)
- Irbaz Bin Riaz
- Department of AI and Informatics, Mayo Clinic, Minnesota
- Division of Hematology and Oncology, Mayo Clinic, Phoenix, Arizona
| | | | - Tufia C Haddad
- Department of Oncology, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
9
|
Chen Z, Chen Y, Sun Y, Tang L, Zhang L, Hu Y, He M, Li Z, Cheng S, Yuan J, Wang Z, Wang Y, Zhao J, Gong J, Zhao L, Cao B, Li G, Zhang X, Dong B, Shen L. Predicting gastric cancer response to anti-HER2 therapy or anti-HER2 combined immunotherapy based on multi-modal data. Signal Transduct Target Ther 2024; 9:222. [PMID: 39183247 PMCID: PMC11345439 DOI: 10.1038/s41392-024-01932-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Revised: 07/04/2024] [Accepted: 07/17/2024] [Indexed: 08/27/2024] Open
Abstract
The sole use of single modality data often fails to capture the complex heterogeneity among patients, including the variability in resistance to anti-HER2 therapy and outcomes of combined treatment regimens, for the treatment of HER2-positive gastric cancer (GC). This modality deficit has not been fully considered in many studies. Furthermore, the application of artificial intelligence in predicting the treatment response, particularly in complex diseases such as GC, is still in its infancy. Therefore, this study aimed to use a comprehensive analytic approach to accurately predict treatment responses to anti-HER2 therapy or anti-HER2 combined immunotherapy in patients with HER2-positive GC. We collected multi-modal data, comprising radiology, pathology, and clinical information from a cohort of 429 patients: 310 treated with anti-HER2 therapy and 119 treated with a combination of anti-HER2 and anti-PD-1/PD-L1 inhibitors immunotherapy. We introduced a deep learning model, called the Multi-Modal model (MuMo), that integrates these data to make precise treatment response predictions. MuMo achieved an area under the curve score of 0.821 for anti-HER2 therapy and 0.914 for combined immunotherapy. Moreover, patients classified as low-risk by MuMo exhibited significantly prolonged progression-free survival and overall survival (log-rank test, P < 0.05). These findings not only highlight the significance of multi-modal data analysis in enhancing treatment evaluation and personalized medicine for HER2-positive gastric cancer, but also the potential and clinical value of our model.
Collapse
Affiliation(s)
- Zifan Chen
- Center for Data Science, Peking University, Beijing, China
| | - Yang Chen
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital and Institute, Beijing, China
| | - Yu Sun
- Department of Pathology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital and Institute, Beijing, China
| | - Lei Tang
- Department of Radiology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital and Institute, Beijing, China
| | - Li Zhang
- Center for Data Science, Peking University, Beijing, China
- National Biomedical Imaging Center, Peking University, Beijing, China
| | - Yajie Hu
- Department of Pathology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital and Institute, Beijing, China
| | - Meng He
- Department of Radiology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital and Institute, Beijing, China
| | - Zhiwei Li
- Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Guangzhou, China
| | - Siyuan Cheng
- Department of Medical Oncology and Radiation Sickness, Peking University Third Hospital, Beijing, China
| | - Jiajia Yuan
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital and Institute, Beijing, China
| | - Zhenghang Wang
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital and Institute, Beijing, China
| | - Yakun Wang
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital and Institute, Beijing, China
| | - Jie Zhao
- National Engineering Laboratory for Big Data Analysis and Applications, Peking University, Beijing, China
| | - Jifang Gong
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital and Institute, Beijing, China
| | - Liying Zhao
- Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Guangzhou, China
| | - Baoshan Cao
- Department of Medical Oncology and Radiation Sickness, Peking University Third Hospital, Beijing, China
| | - Guoxin Li
- Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Guangzhou, China
| | - Xiaotian Zhang
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital and Institute, Beijing, China.
| | - Bin Dong
- National Biomedical Imaging Center, Peking University, Beijing, China.
- Beijing International Center for Mathematical Research (BICMR), Peking University, Beijing, China.
- Center for Machine Learning Research, Peking University, Beijing, China.
| | - Lin Shen
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital and Institute, Beijing, China.
| |
Collapse
|
10
|
Karim ME. Can supervised deep learning architecture outperform autoencoders in building propensity score models for matching? BMC Med Res Methodol 2024; 24:167. [PMID: 39095707 PMCID: PMC11295454 DOI: 10.1186/s12874-024-02284-5] [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: 03/05/2024] [Accepted: 07/15/2024] [Indexed: 08/04/2024] Open
Abstract
PURPOSE Propensity score matching is vital in epidemiological studies using observational data, yet its estimates relies on correct model-specification. This study assesses supervised deep learning models and unsupervised autoencoders for propensity score estimation, comparing them with traditional methods for bias and variance accuracy in treatment effect estimations. METHODS Utilizing a plasmode simulation based on the Right Heart Catheterization dataset, under a variety of settings, we evaluated (1) a supervised deep learning architecture and (2) an unsupervised autoencoder, alongside two traditional methods: logistic regression and a spline-based method in estimating propensity scores for matching. Performance metrics included bias, standard errors, and coverage probability. The analysis was also extended to real-world data, with estimates compared to those obtained via a double robust approach. RESULTS The analysis revealed that supervised deep learning models outperformed unsupervised autoencoders in variance estimation while maintaining comparable levels of bias. These results were supported by analyses of real-world data, where the supervised model's estimates closely matched those derived from conventional methods. Additionally, deep learning models performed well compared to traditional methods in settings where exposure was rare. CONCLUSION Supervised deep learning models hold promise in refining propensity score estimations in epidemiological research, offering nuanced confounder adjustment, especially in complex datasets. We endorse integrating supervised deep learning into epidemiological research and share reproducible codes for widespread use and methodological transparency.
Collapse
Affiliation(s)
- Mohammad Ehsanul Karim
- School of Population and Public Health, University of British Columbia, 2206 East Mall, Vancouver, BC, V6T 1Z3, Canada.
- Centre for Advancing Health Outcomes, 588 - 1081 Burrard Street, Vancouver, BC, V6Z 1Y6, Canada.
| |
Collapse
|
11
|
Jang SJ, Driscoll DA, Anderson CG, Sokrab R, Flevas DA, Mayman DJ, Vigdorchik JM, Jerabek SA, Sculco PK. Radiographic Findings Associated With Mild Hip Dysplasia in 3869 Patients Using a Deep Learning Measurement Tool. Arthroplast Today 2024; 28:101398. [PMID: 38993836 PMCID: PMC11237356 DOI: 10.1016/j.artd.2024.101398] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Revised: 03/10/2024] [Accepted: 04/02/2024] [Indexed: 07/13/2024] Open
Abstract
Background Hip dysplasia is considered one of the leading etiologies contributing to hip degeneration and the eventual need for total hip arthroplasty (THA). We validated a deep learning (DL) algorithm to measure angles relevant to hip dysplasia and applied this algorithm to determine the prevalence of dysplasia in a large population based on incremental radiographic cutoffs. Methods Patients from the Osteoarthritis Initiative with anteroposterior pelvis radiographs and without previous THAs were included. A DL algorithm automated 3 angles associated with hip dysplasia: modified lateral center-edge angle (LCEA), Tönnis angle, and modified Sharp angle. The algorithm was validated against manual measurements, and all angles were measured in a cohort of 3869 patients (61.2 ± 9.2 years, 57.1% female). The percentile distributions and prevalence of dysplastic hips were analyzed using each angle. Results The algorithm had no significant difference (P > .05) in measurements (paired difference: 0.3°-0.7°) against readers and had excellent agreement for dysplasia classification (kappa = 0.78-0.88). In 140 minutes, 23,214 measurements were automated for 3869 patients. LCEA and Sharp angles were higher and the Tönnis angle was lower (P < .01) in females. The dysplastic hip prevalence varied from 2.5% to 20% utilizing the following cutoffs: 17.3°-25.5° (LCEA), 9.4°-15.6° (Tönnis), and 41.3°-45.9° (Sharp). Conclusions A DL algorithm was developed to measure and classify hips with mild hip dysplasia. The reported prevalence of dysplasia in a large patient cohort was dependent on both the measurement and threshold, with 12.4% of patients having dysplasia radiographic indices indicative of higher THA risk.
Collapse
Affiliation(s)
- Seong Jun Jang
- Weill Cornell College of Medicine, New York, NY, USA
- Department of Orthopedic Surgery, Hospital for Special Surgery, New York, NY, USA
| | - Daniel A Driscoll
- Department of Orthopedic Surgery, Hospital for Special Surgery, New York, NY, USA
- Adult Reconstruction and Joint Replacement Service, Hospital for Special Surgery, New York, NY, USA
| | - Christopher G Anderson
- Stavros Niarchos Foundation Complex Joint Reconstruction Center, Hospital for Special Surgery, New York, NY, USA
| | - Ruba Sokrab
- Stavros Niarchos Foundation Complex Joint Reconstruction Center, Hospital for Special Surgery, New York, NY, USA
| | - Dimitrios A Flevas
- Stavros Niarchos Foundation Complex Joint Reconstruction Center, Hospital for Special Surgery, New York, NY, USA
| | - David J Mayman
- Adult Reconstruction and Joint Replacement Service, Hospital for Special Surgery, New York, NY, USA
| | - Jonathan M Vigdorchik
- Adult Reconstruction and Joint Replacement Service, Hospital for Special Surgery, New York, NY, USA
| | - Seth A Jerabek
- Adult Reconstruction and Joint Replacement Service, Hospital for Special Surgery, New York, NY, USA
| | - Peter K Sculco
- Adult Reconstruction and Joint Replacement Service, Hospital for Special Surgery, New York, NY, USA
| |
Collapse
|
12
|
Amygdalos I, Truhn D, Vondran FWR. Outcome prediction after resection of colorectal cancer liver metastases: out with the old, in with the new? Hepatobiliary Surg Nutr 2024; 13:732-735. [PMID: 39175724 PMCID: PMC11336544 DOI: 10.21037/hbsn-24-187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Accepted: 06/19/2024] [Indexed: 08/24/2024]
Affiliation(s)
- Iakovos Amygdalos
- Department of General, Visceral, Pediatric and Transplantation Surgery, University Hospital RWTH Aachen, Aachen, Germany
| | - Daniel Truhn
- Department of Diagnostic and Interventional Radiology, University Hospital RWTH Aachen, Aachen, Germany
| | - Florian W. R. Vondran
- Department of General, Visceral, Pediatric and Transplantation Surgery, University Hospital RWTH Aachen, Aachen, Germany
| |
Collapse
|
13
|
Rodríguez-Miguel A, Arruabarrena C, Allendes G, Olivera M, Zarranz-Ventura J, Teus MA. Hybrid deep learning models for the screening of Diabetic Macular Edema in optical coherence tomography volumes. Sci Rep 2024; 14:17633. [PMID: 39085461 PMCID: PMC11291805 DOI: 10.1038/s41598-024-68489-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2024] [Accepted: 07/24/2024] [Indexed: 08/02/2024] Open
Abstract
Several studies published so far used highly selective image datasets from unclear sources to train computer vision models and that may lead to overestimated results, while those studies conducted in real-life remain scarce. To avoid image selection bias, we stacked convolutional and recurrent neural networks (CNN-RNN) to analyze complete optical coherence tomography (OCT) cubes in a row and predict diabetic macular edema (DME), in a real-world diabetic retinopathy screening program. A retrospective cohort study was carried out. Throughout 4-years, 5314 OCT cubes from 4408 subjects who attended to the diabetic retinopathy (DR) screening program were included. We arranged twenty-two (22) pre-trained CNNs in parallel with a bidirectional RNN layer stacked at the bottom, allowing the model to make a prediction for the whole OCT cube. The staff of retina experts built a ground truth of DME later used to train a set of these CNN-RNN models with different configurations. For each trained CNN-RNN model, we performed threshold tuning to find the optimal cut-off point for binary classification of DME. Finally, the best models were selected according to sensitivity, specificity, and area under the receiver operating characteristics curve (AUROC) with their 95% confidence intervals (95%CI). An ensemble of the best models was also explored. 5188 cubes were non-DME and 126 were DME. Three models achieved an AUROC of 0.94. Among these, sensitivity, and specificity (95%CI) ranged from 84.1-90.5 and 89.7-93.3, respectively, at threshold 1, from 89.7-92.1 and 80-83.1 at threshold 2, and from 80.2-81 and 93.8-97, at threshold 3. The ensemble model improved these results, and lower specificity was observed among subjects with sight-threatening DR. Analysis by age, gender, or grade of DME did not vary the performance of the models. CNN-RNN models showed high diagnostic accuracy for detecting DME in a real-world setting. This engine allowed us to detect extra-foveal DMEs commonly overlooked in other studies, and showed potential for application as the first filter of non-referable patients in an outpatient center within a population-based DR screening program, otherwise ended up in specialized care.
Collapse
Affiliation(s)
| | - Carolina Arruabarrena
- Department of Ophthalmology, Retina Unit, University Hospital "Príncipe de Asturias", 28805, Madrid, Spain
| | - Germán Allendes
- Department of Ophthalmology, Retina Unit, University Hospital "Príncipe de Asturias", 28805, Madrid, Spain
| | | | - Javier Zarranz-Ventura
- Hospital Clínic de Barcelona, University of Barcelona, 08036, Barcelona, Spain
- Institut de Investigacions Biomediques August Pi I Sunyer (IDIBAPS), 08036, Barcelona, Spain
| | - Miguel A Teus
- Department of Surgery, Medical and Social Sciences (Ophthalmology), University of Alcalá, 28871, Madrid, Spain
| |
Collapse
|
14
|
Tak D, Garomsa BA, Zapaishchykova A, Ye Z, Vajapeyam S, Mahootiha M, Climent Pardo JC, Smith C, Familiar AM, Chaunzwa T, Liu KX, Prabhu S, Bandopadhayay P, Nabavizadeh A, Mueller S, Aerts HJ, Haas-Kogan D, Poussaint TY, Kann BH. Longitudinal risk prediction for pediatric glioma with temporal deep learning. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.06.04.24308434. [PMID: 38978642 PMCID: PMC11230342 DOI: 10.1101/2024.06.04.24308434] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/10/2024]
Abstract
Pediatric glioma recurrence can cause morbidity and mortality; however, recurrence pattern and severity are heterogeneous and challenging to predict with established clinical and genomic markers. Resultingly, almost all children undergo frequent, long-term, magnetic resonance (MR) brain surveillance regardless of individual recurrence risk. Deep learning analysis of longitudinal MR may be an effective approach for improving individualized recurrence prediction in gliomas and other cancers but has thus far been infeasible with current frameworks. Here, we propose a self-supervised, deep learning approach to longitudinal medical imaging analysis, temporal learning, that models the spatiotemporal information from a patient's current and prior brain MRs to predict future recurrence. We apply temporal learning to pediatric glioma surveillance imaging for 715 patients (3,994 scans) from four distinct clinical settings. We find that longitudinal imaging analysis with temporal learning improves recurrence prediction performance by up to 41% compared to traditional approaches, with improvements in performance in both low- and high-grade glioma. We find that recurrence prediction accuracy increases incrementally with the number of historical scans available per patient. Temporal deep learning may enable point-of-care decision-support for pediatric brain tumors and be adaptable more broadly to patients with other cancers and chronic diseases undergoing surveillance imaging.
Collapse
|
15
|
van Timmeren JE, Bussink J, Koopmans P, Smeenk RJ, Monshouwer R. Longitudinal Image Data for Outcome Modeling. Clin Oncol (R Coll Radiol) 2024:S0936-6555(24)00277-2. [PMID: 39003124 DOI: 10.1016/j.clon.2024.06.053] [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: 10/23/2023] [Revised: 04/15/2024] [Accepted: 06/24/2024] [Indexed: 07/15/2024]
Abstract
In oncology, medical imaging is crucial for diagnosis, treatment planning and therapy execution. Treatment responses can be complex and varied and are known to involve factors of treatment, patient characteristics and tumor microenvironment. Longitudinal image analysis is able to track temporal changes, aiding in disease monitoring, treatment evaluation, and outcome prediction. This allows for the enhancement of personalized medicine. However, analyzing longitudinal 2D and 3D images presents unique challenges, including image registration, reliable segmentation, dealing with variable imaging intervals, and sparse data. This review presents an overview of techniques and methodologies in longitudinal image analysis, with a primary focus on outcome modeling in radiation oncology.
Collapse
Affiliation(s)
- J E van Timmeren
- Department of Radiation Oncology, Radboud University Medical Center, Nijmegen, the Netherlands.
| | - J Bussink
- Department of Radiation Oncology, Radboud University Medical Center, Nijmegen, the Netherlands.
| | - P Koopmans
- Department of Radiation Oncology, Radboud University Medical Center, Nijmegen, the Netherlands.
| | - R J Smeenk
- Department of Radiation Oncology, Radboud University Medical Center, Nijmegen, the Netherlands.
| | - R Monshouwer
- Department of Radiation Oncology, Radboud University Medical Center, Nijmegen, the Netherlands.
| |
Collapse
|
16
|
Reitsam NG, Enke JS, Vu Trung K, Märkl B, Kather JN. Artificial Intelligence in Colorectal Cancer: From Patient Screening over Tailoring Treatment Decisions to Identification of Novel Biomarkers. Digestion 2024; 105:331-344. [PMID: 38865982 PMCID: PMC11457979 DOI: 10.1159/000539678] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Accepted: 06/04/2024] [Indexed: 06/14/2024]
Abstract
BACKGROUND Artificial intelligence (AI) is increasingly entering and transforming not only medical research but also clinical practice. In the last 10 years, new AI methods have enabled computers to perform visual tasks, reaching high performance and thereby potentially supporting and even outperforming human experts. This is in particular relevant for colorectal cancer (CRC), which is the 3rd most common cancer type in general, as along the CRC patient journey many complex visual tasks need to be performed: from endoscopy over imaging to histopathology; the screening, diagnosis, and treatment of CRC involve visual image analysis tasks. SUMMARY In all these clinical areas, AI models have shown promising results by supporting physicians, improving accuracy, and providing new biological insights and biomarkers. By predicting prognostic and predictive biomarkers from routine images/slides, AI models could lead to an improved patient stratification for precision oncology approaches in the near future. Moreover, it is conceivable that AI models, in particular together with innovative techniques such as single-cell or spatial profiling, could help identify novel clinically as well as biologically meaningful biomarkers that could pave the way to new therapeutic approaches. KEY MESSAGES Here, we give a comprehensive overview of AI in colorectal cancer, describing and discussing these developments as well as the next steps which need to be taken to incorporate AI methods more broadly into the clinical care of CRC.
Collapse
Affiliation(s)
- Nic Gabriel Reitsam
- Pathology, Faculty of Medicine, University of Augsburg, Augsburg, Germany,
- Bavarian Cancer Research Center (BZKF), Augsburg, Germany,
| | - Johanna Sophie Enke
- Nuclear Medicine, Faculty of Medicine, University of Augsburg, Augsburg, Germany
| | - Kien Vu Trung
- Division of Gastroenterology, Medical Department II, University of Leipzig Medical Center, Leipzig, Germany
| | - Bruno Märkl
- Pathology, Faculty of Medicine, University of Augsburg, Augsburg, Germany
- Bavarian Cancer Research Center (BZKF), Augsburg, Germany
| | - Jakob Nikolas Kather
- Else Kroener Fresenius Center for Digital Health, Technical University Dresden, Dresden, Germany
- Pathology and Data Analytics, Leeds Institute of Medical Research at St James's, University of Leeds, Leeds, UK
- Department of Medicine I, University Hospital Dresden, Dresden, Germany
- Medical Oncology, National Center for Tumor Diseases (NCT), University Hospital Heidelberg, Heidelberg, Germany
| |
Collapse
|
17
|
Perez-Lopez R, Ghaffari Laleh N, Mahmood F, Kather JN. A guide to artificial intelligence for cancer researchers. Nat Rev Cancer 2024; 24:427-441. [PMID: 38755439 DOI: 10.1038/s41568-024-00694-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/09/2024] [Indexed: 05/18/2024]
Abstract
Artificial intelligence (AI) has been commoditized. It has evolved from a specialty resource to a readily accessible tool for cancer researchers. AI-based tools can boost research productivity in daily workflows, but can also extract hidden information from existing data, thereby enabling new scientific discoveries. Building a basic literacy in these tools is useful for every cancer researcher. Researchers with a traditional biological science focus can use AI-based tools through off-the-shelf software, whereas those who are more computationally inclined can develop their own AI-based software pipelines. In this article, we provide a practical guide for non-computational cancer researchers to understand how AI-based tools can benefit them. We convey general principles of AI for applications in image analysis, natural language processing and drug discovery. In addition, we give examples of how non-computational researchers can get started on the journey to productively use AI in their own work.
Collapse
Affiliation(s)
- Raquel Perez-Lopez
- Radiomics Group, Vall d'Hebron Institute of Oncology, Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain
| | - Narmin Ghaffari Laleh
- Else Kroener Fresenius Center for Digital Health, Technical University Dresden, Dresden, Germany
| | - Faisal Mahmood
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Cancer Program, Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Cancer Data Science Program, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
- Harvard Data Science Initiative, Harvard University, Cambridge, MA, USA
| | - Jakob Nikolas Kather
- Else Kroener Fresenius Center for Digital Health, Technical University Dresden, Dresden, Germany.
- Department of Medicine I, University Hospital Dresden, Dresden, Germany.
- Medical Oncology, National Center for Tumour Diseases (NCT), University Hospital Heidelberg, Heidelberg, Germany.
| |
Collapse
|
18
|
Kummar S, Lu R. Using Radiomics in Cancer Management. JCO Precis Oncol 2024; 8:e2400155. [PMID: 38723232 DOI: 10.1200/po.24.00155] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2024] [Accepted: 03/19/2024] [Indexed: 08/23/2024] Open
Affiliation(s)
- Shivaani Kummar
- Division of Hematology and Medical Oncology, Knight Cancer Institute, Oregon Heath & Science University School of Medicine, Portland, OR
| | | |
Collapse
|
19
|
Nakajima EC, Simpson A, Bogaerts J, de Vries EGE, Do R, Garalda E, Goldmacher G, Kinahan PE, Lambin P, LeStage B, Li Q, Lin F, Litière S, Perez-Lopez R, Petrick N, Schwartz L, Seymour L, Shankar L, Laurie SA. Tumor Size Is Not Everything: Advancing Radiomics as a Precision Medicine Biomarker in Oncology Drug Development and Clinical Care. A Report of a Multidisciplinary Workshop Coordinated by the RECIST Working Group. JCO Precis Oncol 2024; 8:e2300687. [PMID: 38635935 DOI: 10.1200/po.23.00687] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2023] [Revised: 02/08/2024] [Accepted: 03/05/2024] [Indexed: 04/20/2024] Open
Abstract
Radiomics, the science of extracting quantifiable data from routine medical images, is a powerful tool that has many potential applications in oncology. The Response Evaluation Criteria in Solid Tumors Working Group (RWG) held a workshop in May 2022, which brought together various stakeholders to discuss the potential role of radiomics in oncology drug development and clinical trials, particularly with respect to response assessment. This article summarizes the results of that workshop, reviewing radiomics for the practicing oncologist and highlighting the work that needs to be done to move forward the incorporation of radiomics into clinical trials.
Collapse
Affiliation(s)
| | | | | | | | - Richard Do
- Memorial Sloan-Kettering Cancer Center, NY, NY
| | - Elena Garalda
- Vall d'Hebron Institute of Oncology, Barcelona, Spain
| | | | | | | | | | | | - Frank Lin
- University of Sydney, Sydney, Australia
| | | | | | | | | | - Lesley Seymour
- Canadian Cancer Trials Group, Queen's University, Kingston, ON, Canada
| | - Lalitha Shankar
- National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Scott A Laurie
- The Ottawa Hospital Cancer Centre, University of Ottawa, Ottawa, ON, Canada
| |
Collapse
|
20
|
Wei Y, Wang H, Chen Z, Zhu Y, Li Y, Lu B, Pan K, Wen C, Cao G, He Y, Zhou J, Pan Z, Wang M. Deep Learning-Based Multiparametric MRI Model for Preoperative T-Stage in Rectal Cancer. J Magn Reson Imaging 2024; 59:1083-1092. [PMID: 37367938 DOI: 10.1002/jmri.28856] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 05/24/2023] [Accepted: 05/29/2023] [Indexed: 06/28/2023] Open
Abstract
BACKGROUND Conventional MRI staging can be challenging in the preoperative assessment of rectal cancer. Deep learning methods based on MRI have shown promise in cancer diagnosis and prognostication. However, the value of deep learning in rectal cancer T-staging is unclear. PURPOSE To develop a deep learning model based on preoperative multiparametric MRI for evaluation of rectal cancer and to investigate its potential to improve T-staging accuracy. STUDY TYPE Retrospective. POPULATION After cross-validation, 260 patients (123 with T-stage T1-2 and 134 with T-stage T3-4) with histopathologically confirmed rectal cancer were randomly divided to the training (N = 208) and test sets (N = 52). FIELD STRENGTH/SEQUENCE 3.0 T/Dynamic contrast enhanced (DCE), T2-weighted imaging (T2W), and diffusion-weighted imaging (DWI). ASSESSMENT The deep learning (DL) model of multiparametric (DCE, T2W, and DWI) convolutional neural network were constructed for evaluating preoperative diagnosis. The pathological findings served as the reference standard for T-stage. For comparison, the single parameter DL-model, a logistic regression model composed of clinical features and subjective assessment of radiologists were used. STATISTICAL TESTS The receiver operating characteristic curve (ROC) was used to evaluate the models, the Fleiss' kappa for the intercorrelation coefficients, and DeLong test for compare the diagnostic performance of ROCs. P-values less than 0.05 were considered statistically significant. RESULTS The Area Under Curve (AUC) of the multiparametric DL-model was 0.854, which was significantly higher than the radiologist's assessment (AUC = 0.678), clinical model (AUC = 0.747), and the single parameter DL-models including T2W-model (AUC = 0.735), DWI-model (AUC = 0.759), and DCE-model (AUC = 0.789). DATA CONCLUSION In the evaluation of rectal cancer patients, the proposed multiparametric DL-model outperformed the radiologist's assessment, the clinical model as well as the single parameter models. The multiparametric DL-model has the potential to assist clinicians by providing more reliable and precise preoperative T staging diagnosis. EVIDENCE LEVEL 3 TECHNICAL EFFICACY: Stage 2.
Collapse
Affiliation(s)
- Yaru Wei
- Department of Radiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
- School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, China
| | - Haojie Wang
- Cixi Biomedical Research Institute, Wenzhou Medical University, Zhejiang, China
- Key Laboratory of Intelligent Medical Imaging of Wenzhou, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Zhongwei Chen
- Department of Radiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Ying Zhu
- Department of Radiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Yingfa Li
- Key Laboratory of Intelligent Medical Imaging of Wenzhou, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Beichen Lu
- Key Laboratory of Intelligent Medical Imaging of Wenzhou, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Kehua Pan
- Department of Radiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Caiyun Wen
- Department of Radiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Guoquan Cao
- Department of Radiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Yun He
- Department of Radiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Jiejie Zhou
- Department of Radiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Zhifang Pan
- Key Laboratory of Intelligent Medical Imaging of Wenzhou, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Meihao Wang
- Department of Radiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
- School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
21
|
Chantadisai M, Wongwijitsook J, Ritlumlert N, Rakvongthai Y. Combined clinical variable and radiomics of post-treatment total body scan for prediction of successful I-131 ablation in low-risk papillary thyroid carcinoma patients. Sci Rep 2024; 14:5001. [PMID: 38424177 PMCID: PMC10904821 DOI: 10.1038/s41598-024-55755-6] [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: 10/12/2023] [Accepted: 02/27/2024] [Indexed: 03/02/2024] Open
Abstract
To explore the feasibility of combined radiomics of post-treatment I-131 total body scan (TBS) and clinical parameter to predict successful ablation in low-risk papillary thyroid carcinoma (PTC) patients. Data of low-risk PTC patients who underwent total/near total thyroidectomy and I-131 ablation 30 mCi between April 2015 and July 2021 were retrospectively reviewed. The clinical factors studied included age, sex, and pre-ablative serum thyroglobulin (Tg). Radiomic features were extracted via PyRadiomics, and radiomic feature selection was performed. The predictive performance for successful ablation of the clinical parameter, radiomic, and combined models (radiomics combined with clinical parameter) was calculated using the area under the receiver operating characteristic curve (AUC). One hundred and thirty patients were included. Successful ablation was achieved in 77 patients (59.2%). The mean pre-ablative Tg in the unsuccessful group (15.50 ± 18.04 ng/ml) was statistically significantly higher than those in the successful ablation group (7.12 ± 7.15 ng/ml). The clinical parameter, radiomic, and combined models produced AUCs of 0.66, 0.77, and 0.87 in the training sets, and 0.65, 0.69, and 0.78 in the validation sets, respectively. The combined model produced a significantly higher AUC than that of the clinical parameter (p < 0.05). Radiomic analysis of the post-treatment TBS combined with pre-ablative serum Tg showed a significant improvement in the predictive performance of successful ablation in low-risk PTC patients compared to the use of clinical parameter alone.Thai Clinical Trials Registry TCTR identification number is TCTR20230816004 ( https://www.thaiclinicaltrials.org/show/TCTR20230816004 ).
Collapse
Affiliation(s)
- Maythinee Chantadisai
- Division of Nuclear Medicine, Department of Radiology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand.
- Division of Nuclear Medicine, Department of Radiology, King Chulalongkorn Memorial Hospital, The Thai Red Cross Society, Bangkok, Thailand.
| | - Jirarot Wongwijitsook
- Division of Nuclear Medicine, Department of Radiology, King Chulalongkorn Memorial Hospital, The Thai Red Cross Society, Bangkok, Thailand
- Division of Nuclear Medicine, Department of Radiology, Surin Hospital, Surin, Thailand
| | - Napat Ritlumlert
- Chulalongkorn University Biomedical Imaging Group, Department of Radiology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
- Biomedical Engineering Program, Faculty of Engineering, Chulalongkorn University, Bangkok, Thailand
- School of Radiological Technology, Faculty of Health Science Technology, Chulabhorn Royal Academy, Bangkok, Thailand
| | - Yothin Rakvongthai
- Division of Nuclear Medicine, Department of Radiology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
- Chulalongkorn University Biomedical Imaging Group, Department of Radiology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| |
Collapse
|
22
|
He M, Chen ZF, Liu S, Chen Y, Zhang H, Zhang L, Zhao J, Yang J, Zhang XT, Shen L, Gao JB, Dong B, Tang L. Deep learning model based on multi-lesion and time series CT images for predicting the benefits from anti-HER2 targeted therapy in stage IV gastric cancer. Insights Imaging 2024; 15:59. [PMID: 38411839 PMCID: PMC10899559 DOI: 10.1186/s13244-024-01639-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Accepted: 01/31/2024] [Indexed: 02/28/2024] Open
Abstract
OBJECTIVE To develop and validate a deep learning model based on multi-lesion and time series CT images in predicting overall survival (OS) in patients with stage IV gastric cancer (GC) receiving anti-HER2 targeted therapy. METHODS A total of 207 patients were enrolled in this multicenter study, with 137 patients for retrospective training and internal validation, 33 patients for prospective validation, and 37 patients for external validation. All patients received anti-HER2 targeted therapy and underwent pre- and post-treatment CT scans (baseline and at least one follow-up). The proposed deep learning model evaluated the multiple lesions in time series CT images to predict risk probabilities. We further evaluated and validated the risk score of the nomogram combining a two-follow-up lesion-based deep learning model (LDLM-2F), tumor markers, and clinical information for predicting the benefits from treatment (Nomo-LDLM-2F). RESULTS In the internal validation and prospective cohorts, the one-year AUCs for Nomo-LDLM-2F using the time series medical images and tumor markers were 0.894 (0.728-1.000) and 0.809 (0.561-1.000), respectively. In the external validation cohort, the one-year AUC of Nomo-LDLM-2F without tumor markers was 0.771 (0.510-1.000). Patients with a low Nomo-LDLM-2F score derived survival benefits from anti-HER2 targeted therapy significantly compared to those with a high Nomo-LDLM-2F score (all p < 0.05). CONCLUSION The Nomo-LDLM-2F score derived from multi-lesion and time series CT images holds promise for the effective readout of OS probability in patients with HER2-positive stage IV GC receiving anti-HER2 therapy. CRITICAL RELEVANCE STATEMENT The deep learning model using baseline and early follow-up CT images aims to predict OS in patients with stage IV gastric cancer receiving anti-HER2 targeted therapy. This model highlights the spatiotemporal heterogeneity of stage IV GC, assisting clinicians in the early evaluation of the efficacy of anti-HER2 therapy. KEY POINTS • Multi-lesion and time series model revealed the spatiotemporal heterogeneity in anti-HER2 therapy. • The Nomo-LDLM-2F score was a valuable prognostic marker for anti-HER2 therapy. • CT-based deep learning model incorporating time-series tumor markers improved performance.
Collapse
Affiliation(s)
- Meng He
- Department of Radiology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital and Institute, Beijing, China
| | - Zi-Fan Chen
- Center for Data Science, Peking University, Beijing, China
| | - Song Liu
- Department of Radiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, No. 321, Zhongshan Road, Nanjing CityJiangsu Province, 210008, China
| | - Yang Chen
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital & Institute, Beijing, China
| | - Huan Zhang
- Department of Radiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Li Zhang
- Center for Data Science, Peking University, Beijing, China
| | - Jie Zhao
- National Engineering Laboratory for Big Data Analysis and Applications, Peking University, Beijing, China
- Peking University Changsha Institute for Computing and Digital Economy, Changsha, China
| | - Jie Yang
- Department of Radiology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital and Institute, Beijing, China
| | - Xiao-Tian Zhang
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital & Institute, Beijing, China
| | - Lin Shen
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital & Institute, Beijing, China.
| | - Jian-Bo Gao
- Department of Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
| | - Bin Dong
- Peking University Changsha Institute for Computing and Digital Economy, Changsha, China.
- Beijing International Center for Mathematical Research, Peking University, Beijing, China.
- Center for Machine Learning Research, Peking University, Beijing, China.
| | - Lei Tang
- Department of Radiology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital and Institute, Beijing, China.
| |
Collapse
|
23
|
Kato S, Miyoshi N, Fujino S, Minami S, Nagae A, Hayashi R, Sekido Y, Hata T, Hamabe A, Ogino T, Tei M, Kagawa Y, Takahashi H, Uemura M, Yamamoto H, Doki Y, Eguchi H. Treatment response prediction of neoadjuvant chemotherapy for rectal cancer by deep learning of colonoscopy images. Oncol Lett 2023; 26:474. [PMID: 37809043 PMCID: PMC10551859 DOI: 10.3892/ol.2023.14062] [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: 01/11/2023] [Accepted: 08/04/2023] [Indexed: 10/10/2023] Open
Abstract
In current clinical practice, several treatment methods, including neoadjuvant therapy, are being developed to improve overall survival or local recurrence rates for locally advanced rectal cancer. The response to neoadjuvant therapy is usually evaluated using imaging data collected before and after preoperative treatment or postsurgical pathological diagnosis. However, there is a need to accurately predict the response to preoperative treatment before treatment is administered. The present study used a deep learning network to examine colonoscopy images and construct a model to predict the response of rectal cancer to neoadjuvant chemotherapy. A total of 53 patients who underwent preoperative chemotherapy followed by radical resection for advanced rectal cancer at the Osaka University Hospital between January 2011 and August 2019 were retrospectively analyzed. A convolutional neural network model was constructed using 403 images from 43 patients as the learning set. The diagnostic accuracy of the deep learning model was evaluated using 84 images from 10 patients as the validation set. The model demonstrated a sensitivity, specificity, accuracy, positive predictive value and area under the curve of 77.6% (38/49), 62.9% (22/33), 71.4% (60/84), 74.5% (38/51) and 0.713, respectively, in predicting a poor response to neoadjuvant therapy. Overall, deep learning of colonoscopy images may contribute to an accurate prediction of the response of rectal cancer to neoadjuvant chemotherapy.
Collapse
Affiliation(s)
- Shinya Kato
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Suita, Osaka 565-0871, Japan
- Department of Innovative Oncology Research and Regenerative Medicine, Osaka International Cancer Institute, Osaka 541-8567, Japan
| | - Norikatsu Miyoshi
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Suita, Osaka 565-0871, Japan
- Department of Innovative Oncology Research and Regenerative Medicine, Osaka International Cancer Institute, Osaka 541-8567, Japan
| | - Shiki Fujino
- Department of Innovative Oncology Research and Regenerative Medicine, Osaka International Cancer Institute, Osaka 541-8567, Japan
| | - Soichiro Minami
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Suita, Osaka 565-0871, Japan
- Department of Innovative Oncology Research and Regenerative Medicine, Osaka International Cancer Institute, Osaka 541-8567, Japan
| | - Ayumi Nagae
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Suita, Osaka 565-0871, Japan
- Department of Innovative Oncology Research and Regenerative Medicine, Osaka International Cancer Institute, Osaka 541-8567, Japan
| | - Rie Hayashi
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Suita, Osaka 565-0871, Japan
- Department of Innovative Oncology Research and Regenerative Medicine, Osaka International Cancer Institute, Osaka 541-8567, Japan
| | - Yuki Sekido
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Suita, Osaka 565-0871, Japan
| | - Tsuyoshi Hata
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Suita, Osaka 565-0871, Japan
| | - Atsushi Hamabe
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Suita, Osaka 565-0871, Japan
| | - Takayuki Ogino
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Suita, Osaka 565-0871, Japan
| | - Mitsuyoshi Tei
- Department of Surgery, Osaka Rosai Hospital, Sakai, Osaka 591-8025, Japan
| | - Yoshinori Kagawa
- Department of Gastroenterological Surgery, Osaka General Medical Center, Osaka 558-8588, Japan
| | - Hidekazu Takahashi
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Suita, Osaka 565-0871, Japan
| | - Mamoru Uemura
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Suita, Osaka 565-0871, Japan
| | - Hirofumi Yamamoto
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Suita, Osaka 565-0871, Japan
| | - Yuichiro Doki
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Suita, Osaka 565-0871, Japan
| | - Hidetoshi Eguchi
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Suita, Osaka 565-0871, Japan
| |
Collapse
|
24
|
Doleschal B, Kirchweger P, Schwendinger S, Kupferthaler A, Burghofer J, Webersinke G, Jukic E, Wundsam H, Biebl M, Petzer A, Rumpold H. Response prediction by mutation- or methylation-specific detection of ctDNA dynamics in pretreated metastatic colorectal cancer. Ther Adv Med Oncol 2023; 15:17588359231200462. [PMID: 37786537 PMCID: PMC10541738 DOI: 10.1177/17588359231200462] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Accepted: 08/25/2023] [Indexed: 10/04/2023] Open
Abstract
Background Serial analysis of circulating tumor DNA (ctDNA) levels is a promising tool for both relapse prediction in the curative setting, as well as predicting clinical benefit from systemic treatment in metastasic colorectal cancer (mCRC). Most data in this context are derived from treatment naive patients. Objective To predict progressive disease (PD) as early as possible through monitoring of changes in ctDNA levels during systemic treatment in pretreated patients with mCRC. Design A prospective, single-center, observational study. Methods Patients treated beyond first-line were prospectively included between February 2020 and September 2021. Blood for ctDNA detection was taken before every treatment cycle from start of treatment until first restaging by CT-scan. ctDNA was detected by mutation- (mut-ctDNA) and methylation-specific ddPCR. Receiver Operating Characteristic (ROC)-analysis was used to describe sensitivity and specificity for prediction of PD at restaging for all time points. Results A total of 42 patients were included who all carried a mutation in tumor tissue. Detection rate of mut-ctDNA was 88.1% and 74.4% for meth-ctDNA. Absolute ctDNA levels before treatment were prognostic in terms of overall survival. Levels of ctDNA were significantly higher in patients with PD at restaging. Median time from start of treatment to restaging was 93 days (95% CI 88.8-96). After a median of 19 days of treatment (95% CI 16.1-20.2), a decline of either mutation- or methylation-specific ctDNA levels of ⩽58% predicted PD at restaging with a sensitivity/specificity of 92.9/85.7% and 85.7/100%, respectively. Median time to restaging was 66 days (95% CI 56.8-75.2). There was no significant increase of sensitivity/specificity at later time points of ctDNA measurements. Conclusion Monitoring early changes of ctDNA levels either by mut- or meth-ctDNA allows for early prediction of PD in pretreated patients with mCRC. This has the potential to complement RECIST-based treatment assessment with the aim to switch potentially insufficient treatments as early as possible, which is of particular interest in higher treatment lines.
Collapse
Affiliation(s)
- Bernhard Doleschal
- Department of Internal Medicine I for Hematology With Stem Cell Transplantation, Hemostaseology and Medical Oncology, Ordensklinikum Linz, Linz, Oberösterreich, Austria
| | - Patrick Kirchweger
- Department of Surgery, Ordensklinikum Linz, Linz, Oberösterreich, Austria
- Medical Faculty, Johannes Kepler University Linz, Linz, Austria
| | | | - Alexander Kupferthaler
- Medical Faculty, Johannes Kepler University Linz, Linz, Austria
- Department of Diagnostic and Interventional Radiology, Ordensklinikum Linz, Linz, Austria
| | - Jonathan Burghofer
- Laboratory for Molecular Genetic Diagnostics, Ordensklinikum Linz, Linz, Austria
| | - Gerald Webersinke
- Laboratory for Molecular Genetic Diagnostics, Ordensklinikum Linz, Linz, Austria
| | - Emina Jukic
- Institute of Human Genetics, Medical University of Innsbruck, Austria
| | - Helwig Wundsam
- Department of Surgery, Ordensklinikum Linz, Linz, Austria
| | - Matthias Biebl
- Department of Surgery, Ordensklinikum Linz, Linz, Austria
| | - Andreas Petzer
- Department of Internal Medicine I for Hematology With Stem Cell Transplantation, Hemostaseology and Medical Oncology, Ordensklinikum Linz, Linz, Oberösterreich, Austria
| | - Holger Rumpold
- Medical Faculty, Johannes Kepler University Linz, Linz, Austria
- Gastrointestinal Cancer Center, Ordensklinikum Linz, Seilerstaette 4, Linz 4010, Austria
| |
Collapse
|
25
|
Yin R, Guo Y, Wang Y, Zhang Q, Dou Z, Wang Y, Qi L, Chen Y, Zhang C, Li H, Jian X, Ma W. Predicting Neoadjuvant Chemotherapy Response and High-Grade Serous Ovarian Cancer From CT Images in Ovarian Cancer with Multitask Deep Learning: A Multicenter Study. Acad Radiol 2023; 30 Suppl 2:S192-S201. [PMID: 37336707 DOI: 10.1016/j.acra.2023.04.036] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Revised: 04/25/2023] [Accepted: 04/27/2023] [Indexed: 06/21/2023]
Abstract
RATIONALE AND OBJECTIVES Accurate prediction neoadjuvant chemotherapy (NACT) response in ovarian cancer (OC) is essential for personalized medicine. We aimed to develop and validate a deep learning (DL) model based on pretreatment contrast-enhanced CT (CECT) images for predicting NACT responses and classifying high-grade serous ovarian cancer (HGSOC) to identify patients who may benefit from NACT. MATERIALS AND METHODS This multicenter study, which contained both retrospective and prospective studies, included consecutive OC patients (n = 757) from three hospitals. Using WHO RECIST 1.1 for the reference standard, a total of 587 women with 1761 images were included in the training and validation sets, 67 women with 201 images were included in the prospective sets, and 103 women with 309 images were included in the external sets. A multitask DL model based on the multiperiod CT image was developed to predict NACT response and HGSOC. RESULTS Logistic regression analysis showed that peritoneal invasion, retinal invasion, and inguinal lymph node metastasis were independent predictors. The DL achieved promising segmentation performances with DICEmean= 0.83 (range: 0.78-0.87). For predicting NACT response, the DL model combined with clinical risk factors obtained area under the receiver operating characteristic curve (AUCs) of 0.87 (0.83-0.89), 0.88 (0.86-0.91), 0.86 (0.82-0.89), and 0.79 (0.75-0.82) in the training, validation, prospective, and external sets, respectively. The AUCs were 0.91 (0.87-0.94), 0.89 (0.86-0.91), 0.80 (0.76-0.84), and 0.80 (0.75-0.85) in four sets in HGSOC classification. CONCLUSION The multitask DL model developed using multiperiod CT images exhibited a promising performance for predicting NACT response and HGSOC with OC, which could provide valuable information for individualized treatment.
Collapse
Affiliation(s)
- Rui Yin
- School of Biomedical Engineering & Technology, Tianjin Medical University, Tianjin, China (R.Y., X.J.)
| | - Yijun Guo
- Department of Breast Imaging, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin 300060, China (Y.G., Z.D., W.M.)
| | - Yanyan Wang
- Department of CT and MRI, Shanxi Tumor Hospital, Taiyuan, China (Y.W.)
| | - Qian Zhang
- Department of Radiology, Baoding No. 1 Central Hospital, Baoding, China (Q.Z.)
| | - Zhaoxiang Dou
- Department of Breast Imaging, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin 300060, China (Y.G., Z.D., W.M.)
| | - Yigeng Wang
- Department of Radiology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China (Y.W.)
| | - Lisha Qi
- Department of Pathology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China (L.Q.)
| | - Ying Chen
- Department of Gynecologic Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China (Y.C.)
| | - Chao Zhang
- Department of Bone Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China (C.Z.)
| | - Huiyang Li
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin, China (H.L.)
| | - Xiqi Jian
- School of Biomedical Engineering & Technology, Tianjin Medical University, Tianjin, China (R.Y., X.J.)
| | - Wenjuan Ma
- Department of Breast Imaging, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin 300060, China (Y.G., Z.D., W.M.).
| |
Collapse
|
26
|
Zidane M, Makky A, Bruhns M, Rochwarger A, Babaei S, Claassen M, Schürch CM. A review on deep learning applications in highly multiplexed tissue imaging data analysis. FRONTIERS IN BIOINFORMATICS 2023; 3:1159381. [PMID: 37564726 PMCID: PMC10410935 DOI: 10.3389/fbinf.2023.1159381] [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: 02/05/2023] [Accepted: 07/12/2023] [Indexed: 08/12/2023] Open
Abstract
Since its introduction into the field of oncology, deep learning (DL) has impacted clinical discoveries and biomarker predictions. DL-driven discoveries and predictions in oncology are based on a variety of biological data such as genomics, proteomics, and imaging data. DL-based computational frameworks can predict genetic variant effects on gene expression, as well as protein structures based on amino acid sequences. Furthermore, DL algorithms can capture valuable mechanistic biological information from several spatial "omics" technologies, such as spatial transcriptomics and spatial proteomics. Here, we review the impact that the combination of artificial intelligence (AI) with spatial omics technologies has had on oncology, focusing on DL and its applications in biomedical image analysis, encompassing cell segmentation, cell phenotype identification, cancer prognostication, and therapy prediction. We highlight the advantages of using highly multiplexed images (spatial proteomics data) compared to single-stained, conventional histopathological ("simple") images, as the former can provide deep mechanistic insights that cannot be obtained by the latter, even with the aid of explainable AI. Furthermore, we provide the reader with the advantages/disadvantages of DL-based pipelines used in preprocessing highly multiplexed images (cell segmentation, cell type annotation). Therefore, this review also guides the reader to choose the DL-based pipeline that best fits their data. In conclusion, DL continues to be established as an essential tool in discovering novel biological mechanisms when combined with technologies such as highly multiplexed tissue imaging data. In balance with conventional medical data, its role in clinical routine will become more important, supporting diagnosis and prognosis in oncology, enhancing clinical decision-making, and improving the quality of care for patients. Since its introduction into the field of oncology, deep learning (DL) has impacted clinical discoveries and biomarker predictions. DL-driven discoveries and predictions in oncology are based on a variety of biological data such as genomics, proteomics, and imaging data. DL-based computational frameworks can predict genetic variant effects on gene expression, as well as protein structures based on amino acid sequences. Furthermore, DL algorithms can capture valuable mechanistic biological information from several spatial "omics" technologies, such as spatial transcriptomics and spatial proteomics. Here, we review the impact that the combination of artificial intelligence (AI) with spatial omics technologies has had on oncology, focusing on DL and its applications in biomedical image analysis, encompassing cell segmentation, cell phenotype identification, cancer prognostication, and therapy prediction. We highlight the advantages of using highly multiplexed images (spatial proteomics data) compared to single-stained, conventional histopathological ("simple") images, as the former can provide deep mechanistic insights that cannot be obtained by the latter, even with the aid of explainable AI. Furthermore, we provide the reader with the advantages/disadvantages of the DL-based pipelines used in preprocessing the highly multiplexed images (cell segmentation, cell type annotation). Therefore, this review also guides the reader to choose the DL-based pipeline that best fits their data. In conclusion, DL continues to be established as an essential tool in discovering novel biological mechanisms when combined with technologies such as highly multiplexed tissue imaging data. In balance with conventional medical data, its role in clinical routine will become more important, supporting diagnosis and prognosis in oncology, enhancing clinical decision-making, and improving the quality of care for patients.
Collapse
Affiliation(s)
- Mohammed Zidane
- Department of Pathology and Neuropathology, University Hospital and Comprehensive Cancer Center Tübingen, Tübingen, Germany
| | - Ahmad Makky
- Department of Pathology and Neuropathology, University Hospital and Comprehensive Cancer Center Tübingen, Tübingen, Germany
| | - Matthias Bruhns
- Department of Internal Medicine I, University Hospital Tübingen, Tübingen, Germany
- Department of Computer Science, University of Tübingen, Tübingen, Germany
| | - Alexander Rochwarger
- Department of Pathology and Neuropathology, University Hospital and Comprehensive Cancer Center Tübingen, Tübingen, Germany
| | - Sepideh Babaei
- Department of Internal Medicine I, University Hospital Tübingen, Tübingen, Germany
| | - Manfred Claassen
- Department of Internal Medicine I, University Hospital Tübingen, Tübingen, Germany
- Department of Computer Science, University of Tübingen, Tübingen, Germany
| | - Christian M. Schürch
- Department of Pathology and Neuropathology, University Hospital and Comprehensive Cancer Center Tübingen, Tübingen, Germany
| |
Collapse
|
27
|
Deep learning-enabled fully automated pipeline system for segmentation and classification of single-mass breast lesions using contrast-enhanced mammography: a prospective, multicentre study. EClinicalMedicine 2023; 58:101913. [PMID: 36969336 PMCID: PMC10034267 DOI: 10.1016/j.eclinm.2023.101913] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Revised: 03/01/2023] [Accepted: 03/02/2023] [Indexed: 03/19/2023] Open
Abstract
Background Breast cancer is the leading cause of cancer-related deaths in women. However, accurate diagnosis of breast cancer using medical images heavily relies on the experience of radiologists. This study aimed to develop an artificial intelligence model that diagnosed single-mass breast lesions on contrast-enhanced mammography (CEM) for assisting the diagnostic workflow. Methods A total of 1912 women with single-mass breast lesions on CEM images before biopsy or surgery were included from June 2017 to October 2022 at three centres in China. Samples were divided into training and validation sets, internal testing set, pooled external testing set, and prospective testing set. A fully automated pipeline system (FAPS) using RefineNet and the Xception + Pyramid pooling module (PPM) was developed to perform the segmentation and classification of breast lesions. The performances of six radiologists and adjustments in Breast Imaging Reporting and Data System (BI-RADS) category 4 under the FAPS-assisted strategy were explored in pooled external and prospective testing sets. The segmentation performance was assessed using the Dice similarity coefficient (DSC), and the classification was assessed using heatmaps, area under the receiver operating characteristic curve (AUC), sensitivity, and specificity. The radiologists' reading time was recorded for comparison with the FAPS. This trial is registered with China Clinical Trial Registration Centre (ChiCTR2200063444). Findings The FAPS-based segmentation task achieved DSCs of 0.888 ± 0.101, 0.820 ± 0.148 and 0.837 ± 0.132 in the internal, pooled external and prospective testing sets, respectively. For the classification task, the FAPS achieved AUCs of 0.947 (95% confidence interval [CI]: 0.916-0.978), 0.940 (95% [CI]: 0.894-0.987) and 0.891 (95% [CI]: 0.816-0.945). It outperformed radiologists in terms of classification efficiency based on single lesions (6 s vs 3 min). Moreover, the FAPS-assisted strategy improved the performance of radiologists. BI-RADS category 4 in 12.4% and 13.3% of patients was adjusted in two testing sets with the assistance of FAPS, which may play an important guiding role in the selection of clinical management strategies. Interpretation The FAPS based on CEM demonstrated the potential for the segmentation and classification of breast lesions, and had good generalisation ability and clinical applicability. Funding This study was supported by the Taishan Scholar Foundation of Shandong Province of China (tsqn202211378), National Natural Science Foundation of China (82001775), Natural Science Foundation of Shandong Province of China (ZR2021MH120), and Special Fund for Breast Disease Research of Shandong Medical Association (YXH2021ZX055).
Collapse
|
28
|
Yin M, Liu L, Gao J, Lin J, Qu S, Xu W, Liu X, Xu C, Zhu J. Deep learning for pancreatic diseases based on endoscopic ultrasound: A systematic review. Int J Med Inform 2023; 174:105044. [PMID: 36948061 DOI: 10.1016/j.ijmedinf.2023.105044] [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: 09/30/2022] [Revised: 03/06/2023] [Accepted: 03/10/2023] [Indexed: 03/19/2023]
Abstract
BACKGROUND AND AIMS Endoscopic ultrasonography (EUS) is one of the main examinations in pancreatic diseases. A series of the studies reported the application of deep learning (DL)-assisted EUS in the diagnosis of pancreatic diseases. This systematic review is to evaluate the role of DL algorithms in assisting EUS diagnosis of pancreatic diseases. METHODS Literature search were conducted in PubMed and Semantic Scholar databases. Studies that developed DL models for pancreatic diseases based on EUS were eligible for inclusion. This review was conducted in accordance with the Preferred Reporting Items for Systematic Reviews and Meta-Analyses guidelines and quality assessment of the included studies was performed according to the IJMEDI checklist. RESULTS A total of 23 studies were enrolled into this systematic review, which could be categorized into three groups according to computer vision tasks: classification, detection and segmentation. Seventeen studies focused on the classification task, among which five studies developed simple neural network (NN) models while twelve studies constructed convolutional NN (CNN) models. Three studies were concerned the detection task and five studies were the segmentation task, all based on CNN architectures. All models presented in the studies performed well based on EUS images, videos or voice. According to the IJMEDI checklist, six studies were recognized as high-grade quality, with scores beyond 35 points. CONCLUSIONS DL algorithms show great potential in EUS images/videos/voice for pancreatic diseases. However, there is room for improvement such as sample sizes, multi-center cooperation, data preprocessing, model interpretability, and code sharing.
Collapse
Affiliation(s)
- Minyue Yin
- Department of Gastroenterology, The First Affiliated Hospital of Soochow University, Suzhou 215000, China; Suzhou Clinical Center of Digestive Diseases, Suzhou 215000, China
| | - Lu Liu
- Department of Gastroenterology, The First Affiliated Hospital of Soochow University, Suzhou 215000, China; Suzhou Clinical Center of Digestive Diseases, Suzhou 215000, China
| | - Jingwen Gao
- Department of Gastroenterology, The First Affiliated Hospital of Soochow University, Suzhou 215000, China; Suzhou Clinical Center of Digestive Diseases, Suzhou 215000, China
| | - Jiaxi Lin
- Department of Gastroenterology, The First Affiliated Hospital of Soochow University, Suzhou 215000, China; Suzhou Clinical Center of Digestive Diseases, Suzhou 215000, China
| | - Shuting Qu
- Department of Gastroenterology, The First Affiliated Hospital of Soochow University, Suzhou 215000, China; Suzhou Clinical Center of Digestive Diseases, Suzhou 215000, China
| | - Wei Xu
- Department of Gastroenterology, The First Affiliated Hospital of Soochow University, Suzhou 215000, China; Suzhou Clinical Center of Digestive Diseases, Suzhou 215000, China
| | - Xiaolin Liu
- Department of Gastroenterology, The First Affiliated Hospital of Soochow University, Suzhou 215000, China; Suzhou Clinical Center of Digestive Diseases, Suzhou 215000, China
| | - Chunfang Xu
- Department of Gastroenterology, The First Affiliated Hospital of Soochow University, Suzhou 215000, China; Suzhou Clinical Center of Digestive Diseases, Suzhou 215000, China.
| | - Jinzhou Zhu
- Department of Gastroenterology, The First Affiliated Hospital of Soochow University, Suzhou 215000, China; Suzhou Clinical Center of Digestive Diseases, Suzhou 215000, China.
| |
Collapse
|
29
|
From patterns to patients: Advances in clinical machine learning for cancer diagnosis, prognosis, and treatment. Cell 2023; 186:1772-1791. [PMID: 36905928 DOI: 10.1016/j.cell.2023.01.035] [Citation(s) in RCA: 154] [Impact Index Per Article: 77.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Revised: 01/10/2023] [Accepted: 01/26/2023] [Indexed: 03/12/2023]
Abstract
Machine learning (ML) is increasingly used in clinical oncology to diagnose cancers, predict patient outcomes, and inform treatment planning. Here, we review recent applications of ML across the clinical oncology workflow. We review how these techniques are applied to medical imaging and to molecular data obtained from liquid and solid tumor biopsies for cancer diagnosis, prognosis, and treatment design. We discuss key considerations in developing ML for the distinct challenges posed by imaging and molecular data. Finally, we examine ML models approved for cancer-related patient usage by regulatory agencies and discuss approaches to improve the clinical usefulness of ML.
Collapse
|
30
|
A comprehensive regulatory and industry review of modeling and simulation practices in oncology clinical drug development. J Pharmacokinet Pharmacodyn 2023; 50:147-172. [PMID: 36870005 PMCID: PMC10169901 DOI: 10.1007/s10928-023-09850-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2022] [Accepted: 02/16/2023] [Indexed: 03/05/2023]
Abstract
Exposure-response (E-R) analyses are an integral component in the development of oncology products. Characterizing the relationship between drug exposure metrics and response allows the sponsor to use modeling and simulation to address both internal and external drug development questions (e.g., optimal dose, frequency of administration, dose adjustments for special populations). This white paper is the output of an industry-government collaboration among scientists with broad experience in E-R modeling as part of regulatory submissions. The goal of this white paper is to provide guidance on what the preferred methods for E-R analysis in oncology clinical drug development are and what metrics of exposure should be considered.
Collapse
|
31
|
Lin B, Tan Z, Mo Y, Yang X, Liu Y, Xu B. Intelligent oncology: The convergence of artificial intelligence and oncology. JOURNAL OF THE NATIONAL CANCER CENTER 2023; 3:83-91. [PMID: 39036310 PMCID: PMC11256531 DOI: 10.1016/j.jncc.2022.11.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Revised: 10/07/2022] [Accepted: 11/13/2022] [Indexed: 12/12/2022] Open
Abstract
With increasingly explored ideologies and technologies for potential applications of artificial intelligence (AI) in oncology, we here describe a holistic and structured concept termed intelligent oncology. Intelligent oncology is defined as a cross-disciplinary specialty which integrates oncology, radiology, pathology, molecular biology, multi-omics and computer sciences, aiming to promote cancer prevention, screening, early diagnosis and precision treatment. The development of intelligent oncology has been facilitated by fast AI technology development such as natural language processing, machine/deep learning, computer vision, and robotic process automation. While the concept and applications of intelligent oncology is still in its infancy, and there are still many hurdles and challenges, we are optimistic that it will play a pivotal role for the future of basic, translational and clinical oncology.
Collapse
Affiliation(s)
- Bo Lin
- Chongqing Key Laboratory of Intelligent Oncology for Breast Cancer, Chongqing University Cancer Hospital and Chongqing University School of Medicine, Institute of Intelligent Oncology, Chongqing University, China
| | - Zhibo Tan
- Department of Radiation Oncology, Peking University Shenzhen Hospital, Department of Radiation Oncology, Peking University Shenzhen Hospital, Shenzhen, China
| | - Yaqi Mo
- Chongqing Key Laboratory of Intelligent Oncology for Breast Cancer, Chongqing University Cancer Hospital and Chongqing University School of Medicine, Institute of Intelligent Oncology, Chongqing University, China
| | - Xue Yang
- Department of Biochemistry and Molecular Biology, Key Laboratory of Breast Cancer Prevention and Therapy, Ministry of Education, National Cancer Research Center, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Yajie Liu
- Department of Radiation Oncology, Peking University Shenzhen Hospital, Department of Radiation Oncology, Peking University Shenzhen Hospital, Shenzhen, China
| | - Bo Xu
- Chongqing Key Laboratory of Intelligent Oncology for Breast Cancer, Chongqing University Cancer Hospital and Chongqing University School of Medicine, Institute of Intelligent Oncology, Chongqing University, China
- Department of Biochemistry and Molecular Biology, Key Laboratory of Breast Cancer Prevention and Therapy, Ministry of Education, National Cancer Research Center, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| |
Collapse
|
32
|
Accurate tumor segmentation and treatment outcome prediction with DeepTOP. Radiother Oncol 2023; 183:109550. [PMID: 36813177 DOI: 10.1016/j.radonc.2023.109550] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 01/15/2023] [Accepted: 02/09/2023] [Indexed: 02/24/2023]
Abstract
BACKGROUND Accurate outcome prediction prior to treatment can facilitate trial design and clinical decision making to achieve better treatment outcome. METHOD We developed the DeepTOP tool with deep learning approach for region-of-interest segmentation and clinical outcome prediction using magnetic resonance imaging (MRI). DeepTOP was constructed with an automatic pipeline from tumor segmentation to outcome prediction. In DeepTOP, the segmentation model used U-Net with a codec structure, and the prediction model was built with a three-layer convolutional neural network. In addition, the weight distribution algorithm was developed and applied in the prediction model to optimize the performance of DeepTOP. RESULTS A total of 1889 MRI slices from 99 patients in the phase III multicenter randomized clinical trial (NCT01211210) on neoadjuvant treatment for rectal cancer was used to train and validate DeepTOP. We systematically optimized and validated DeepTOP with multiple devised pipelines in the clinical trial, demonstrating a better performance than other competitive algorithms in accurate tumor segmentation (Dice coefficient: 0.79; IoU: 0.75; slice-specific sensitivity: 0.98) and predicting pathological complete response to chemo/radiotherapy (accuracy: 0.789; specificity: 0.725; and sensitivity: 0.812). DeepTOP is a deep learning tool that could avoid manual labeling and feature extraction and realize automatic tumor segmentation and treatment outcome prediction by using the original MRI images. CONCLUSION DeepTOP is open to provide a tractable framework for the development of other segmentation and predicting tools in clinical settings. DeepTOP-based tumor assessment can provide a reference for clinical decision making and facilitate imaging marker-driven trial design.
Collapse
|
33
|
Zhang S, Mu W, Dong D, Wei J, Fang M, Shao L, Zhou Y, He B, Zhang S, Liu Z, Liu J, Tian J. The Applications of Artificial Intelligence in Digestive System Neoplasms: A Review. HEALTH DATA SCIENCE 2023; 3:0005. [PMID: 38487199 PMCID: PMC10877701 DOI: 10.34133/hds.0005] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Accepted: 12/05/2022] [Indexed: 03/17/2024]
Abstract
Importance Digestive system neoplasms (DSNs) are the leading cause of cancer-related mortality with a 5-year survival rate of less than 20%. Subjective evaluation of medical images including endoscopic images, whole slide images, computed tomography images, and magnetic resonance images plays a vital role in the clinical practice of DSNs, but with limited performance and increased workload of radiologists or pathologists. The application of artificial intelligence (AI) in medical image analysis holds promise to augment the visual interpretation of medical images, which could not only automate the complicated evaluation process but also convert medical images into quantitative imaging features that associated with tumor heterogeneity. Highlights We briefly introduce the methodology of AI for medical image analysis and then review its clinical applications including clinical auxiliary diagnosis, assessment of treatment response, and prognosis prediction on 4 typical DSNs including esophageal cancer, gastric cancer, colorectal cancer, and hepatocellular carcinoma. Conclusion AI technology has great potential in supporting the clinical diagnosis and treatment decision-making of DSNs. Several technical issues should be overcome before its application into clinical practice of DSNs.
Collapse
Affiliation(s)
- Shuaitong Zhang
- School of Engineering Medicine, Beihang University, Beijing, China
- Key Laboratory of Big Data-Based Precision Medicine, Beihang University, Ministry of Industry and Information Technology, Beijing, China
| | - Wei Mu
- School of Engineering Medicine, Beihang University, Beijing, China
- Key Laboratory of Big Data-Based Precision Medicine, Beihang University, Ministry of Industry and Information Technology, Beijing, China
| | - Di Dong
- CAS Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing, China
| | - Jingwei Wei
- CAS Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing, China
| | - Mengjie Fang
- School of Engineering Medicine, Beihang University, Beijing, China
- Key Laboratory of Big Data-Based Precision Medicine, Beihang University, Ministry of Industry and Information Technology, Beijing, China
| | - Lizhi Shao
- CAS Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing, China
| | - Yu Zhou
- CAS Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing, China
| | - Bingxi He
- School of Engineering Medicine, Beihang University, Beijing, China
- Key Laboratory of Big Data-Based Precision Medicine, Beihang University, Ministry of Industry and Information Technology, Beijing, China
| | - Song Zhang
- CAS Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing, China
| | - Zhenyu Liu
- CAS Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing, China
| | - Jianhua Liu
- Department of Oncology, Guangdong Provincial People's Hospital/Second Clinical Medical College of Southern Medical University/Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, China
| | - Jie Tian
- School of Engineering Medicine, Beihang University, Beijing, China
- Key Laboratory of Big Data-Based Precision Medicine, Beihang University, Ministry of Industry and Information Technology, Beijing, China
- CAS Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
34
|
Li J, Chen Z, Chen Y, Zhao J, He M, Li X, Zhang L, Dong B, Zhang X, Tang L, Shen L. CT-based delta radiomics in predicting the prognosis of stage IV gastric cancer to immune checkpoint inhibitors. Front Oncol 2023; 12:1059874. [PMID: 36686828 PMCID: PMC9847891 DOI: 10.3389/fonc.2022.1059874] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2022] [Accepted: 11/29/2022] [Indexed: 01/06/2023] Open
Abstract
Introduction To explore the prognostic value of CT-based delta radiomics in predicting the prognosis of patients with stage IV gastric cancer treated with immune checkpoint inhibitors (ICI). Materials and methods Forty-two patients with stage IV gastric cancer, who had received ICI monotherapy, were enrolled in this retrospective study. Baseline and first follow-up CT scans were analyzed. Intratumoral and peritumoral regions of interest (ROI) were contoured, enabling the extraction of 192 features from each ROI. The intraclass correlation coefficients were used to select features with high stability. The least absolute shrinkage and selection operator was used to select features with high weights for predicting patient prognosis. Kaplan-Meier analysis and log-rank test were performed to explore the association between features and progression free survival (PFS). Cox regression analyses were used to identify predictors for PFS. The C-index was used to assess the prediction performance of features. Results Two radiomics features of ΔVintra_ZV and postVperi_Sphericity were identified from intratumoral and peritumoral regions, respectively. The Kaplan-Meier analysis revealed significant differences in PFS between patients with low and high feature value (ΔVintra_ZV: P=0.000; postVperi_Sphericity: P=0.012), and the multivariable cox analysis demonstrated that ΔVintra_ZV was independent predictor for PFS (HR, 1.911; 95% CI: 1.163-3.142; P=0.011), with C-index of 0.705. Conclusions Based on CT scans at baseline and first follow-up, the delta radiomics features could efficiently predict the PFS of gastric cancer patients treated with ICI therapy.
Collapse
Affiliation(s)
- Jiazheng Li
- Department of Radiology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital and Institute, Beijing, China
| | - Zifan Chen
- Center for Data Science, Peking University, Beijing, China
| | - Yang Chen
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital and Institute, Beijing, China
| | - Jie Zhao
- National Engineering Laboratory for Big Data Analysis and Applications, Peking University, Beijing, China
| | - Meng He
- Department of Radiology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital and Institute, Beijing, China
| | - Xiaoting Li
- Department of Radiology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital and Institute, Beijing, China
| | - Li Zhang
- Center for Data Science, Peking University, Beijing, China
| | - Bin Dong
- Beijing International Center for Mathematical Research (BICMR), Peking University, Beijing, China,*Correspondence: Bin Dong, ; Xiaotian Zhang, ; Lei Tang, ; Lin Shen,
| | - Xiaotian Zhang
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital and Institute, Beijing, China,*Correspondence: Bin Dong, ; Xiaotian Zhang, ; Lei Tang, ; Lin Shen,
| | - Lei Tang
- Department of Radiology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital and Institute, Beijing, China,*Correspondence: Bin Dong, ; Xiaotian Zhang, ; Lei Tang, ; Lin Shen,
| | - Lin Shen
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital and Institute, Beijing, China,*Correspondence: Bin Dong, ; Xiaotian Zhang, ; Lei Tang, ; Lin Shen,
| |
Collapse
|
35
|
Nakanishi R, Morooka K, Omori K, Toyota S, Tanaka Y, Hasuda H, Koga N, Nonaka K, Hu Q, Nakaji Y, Nakanoko T, Ando K, Ota M, Kimura Y, Oki E, Oda Y, Yoshizumi T. Artificial Intelligence-Based Prediction of Recurrence after Curative Resection for Colorectal Cancer from Digital Pathological Images. Ann Surg Oncol 2022; 30:3506-3514. [PMID: 36512260 DOI: 10.1245/s10434-022-12926-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Accepted: 10/19/2022] [Indexed: 12/15/2022]
Abstract
BACKGROUND To develop an artificial intelligence-based model to predict recurrence after curative resection for stage I-III colorectal cancer from digitized pathological slides. PATIENTS AND METHODS In this retrospective study, 471 consecutive patients who underwent curative resection for stage I-III colorectal cancer at our institution from 2004 to 2015 were enrolled, and 512 randomly selected tiles from digitally scanned images of hematoxylin and eosin-stained tumor tissue sections were used to train a convolutional neural network. Five-fold cross-validation was used to validate the model. The association between recurrence and the model's output scores were analyzed in the test cohorts. RESULTS The area under the receiver operating characteristic curve of the cross-validation was 0.7245 [95% confidence interval (CI) 0.6707-0.7783; P < 0.0001]. The score successfully classified patients into those with better and worse recurrence free survival (P < 0.0001). Multivariate analysis revealed that a high score was significantly associated with worse recurrence free survival [odds ratio (OR) 1.857; 95% CI 1.248-2.805; P = 0.0021], which was independent from other predictive factors: male sex (P = 0.0238), rectal cancer (P = 0.0396), preoperative abnormal carcinoembryonic antigen (CEA) level (P = 0.0216), pathological T3/T4 stage (P = 0.0162), and pathological positive lymph node metastasis (P < 0.0001). CONCLUSIONS The artificial intelligence-based prediction model discriminated patients with a high risk of recurrence. This approach could help decision-makers consider the benefits of adjuvant chemotherapy.
Collapse
|
36
|
Wong PK, Chan IN, Yan HM, Gao S, Wong CH, Yan T, Yao L, Hu Y, Wang ZR, Yu HH. Deep learning based radiomics for gastrointestinal cancer diagnosis and treatment: A minireview. World J Gastroenterol 2022; 28:6363-6379. [PMID: 36533112 PMCID: PMC9753055 DOI: 10.3748/wjg.v28.i45.6363] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 10/25/2022] [Accepted: 11/16/2022] [Indexed: 12/02/2022] Open
Abstract
Gastrointestinal (GI) cancers are the major cause of cancer-related mortality globally. Medical imaging is an important auxiliary means for the diagnosis, assessment and prognostic prediction of GI cancers. Radiomics is an emerging and effective technology to decipher the encoded information within medical images, and traditional machine learning is the most commonly used tool. Recent advances in deep learning technology have further promoted the development of radiomics. In the field of GI cancer, although there are several surveys on radiomics, there is no specific review on the application of deep-learning-based radiomics (DLR). In this review, a search was conducted on Web of Science, PubMed, and Google Scholar with an emphasis on the application of DLR for GI cancers, including esophageal, gastric, liver, pancreatic, and colorectal cancers. Besides, the challenges and recommendations based on the findings of the review are comprehensively analyzed to advance DLR.
Collapse
Affiliation(s)
- Pak Kin Wong
- Department of Electromechanical Engineering, University of Macau, Taipa 999078, Macau, China
| | - In Neng Chan
- Department of Electromechanical Engineering, University of Macau, Taipa 999078, Macau, China
| | - Hao-Ming Yan
- School of Clinical Medicine, China Medical University, Shenyang 110013, Liaoning Province, China
| | - Shan Gao
- Department of Gastroenterology, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang 441021, Hubei Province, China
| | - Chi Hong Wong
- Faculty of Medicine, Macau University of Science and Technology, Taipa 999078, Macau, China
| | - Tao Yan
- School of Mechanical Engineering, Hubei University of Arts and Science, Xiangyang 441053, Hubei Province, China
| | - Liang Yao
- Department of Electromechanical Engineering, University of Macau, Taipa 999078, Macau, China
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, Guangdong Province, China
| | - Ying Hu
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, Guangdong Province, China
| | - Zhong-Ren Wang
- School of Mechanical Engineering, Hubei University of Arts and Science, Xiangyang 441053, Hubei Province, China
| | - Hon Ho Yu
- Department of Gastroenterology, Kiang Wu Hospital, Macau 999078, China
| |
Collapse
|
37
|
Imaging standardisation in metastatic colorectal cancer: A joint EORTC-ESOI-ESGAR expert consensus recommendation. Eur J Cancer 2022; 176:193-206. [PMID: 36274570 DOI: 10.1016/j.ejca.2022.09.012] [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: 07/19/2022] [Revised: 09/13/2022] [Accepted: 09/14/2022] [Indexed: 12/15/2022]
Abstract
BACKGROUND Treatment monitoring in metastatic colorectal cancer (mCRC) relies on imaging to evaluate the tumour burden. Response Evaluation Criteria in Solid Tumors provide a framework on reporting and interpretation of imaging findings yet offer no guidance on a standardised imaging protocol tailored to patients with mCRC. Imaging protocol heterogeneity remains a challenge for the reproducibility of conventional imaging end-points and is an obstacle for research on novel imaging end-points. PATIENTS AND METHODS Acknowledging the recently highlighted potential of radiomics and artificial intelligence tools as decision support for patient care in mCRC, a multidisciplinary, international and expert panel of imaging specialists was formed to find consensus on mCRC imaging protocols using the Delphi method. RESULTS Under the guidance of the European Organisation for Research and Treatment of Cancer (EORTC) Imaging and Gastrointestinal Tract Cancer Groups, the European Society of Oncologic Imaging (ESOI) and the European Society of Gastrointestinal and Abdominal Radiology (ESGAR), the EORTC-ESOI-ESGAR core imaging protocol was identified. CONCLUSION This consensus protocol attempts to promote standardisation and to diminish variations in patient preparation, scan acquisition and scan reconstruction. We anticipate that this standardisation will increase reproducibility of radiomics and artificial intelligence studies and serve as a catalyst for future research on imaging end-points. For ongoing and future mCRC trials, we encourage principal investigators to support the dissemination of these imaging standards across recruiting centres.
Collapse
|
38
|
Radiomic and Volumetric Measurements as Clinical Trial Endpoints—A Comprehensive Review. Cancers (Basel) 2022; 14:cancers14205076. [PMID: 36291865 PMCID: PMC9599928 DOI: 10.3390/cancers14205076] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 10/12/2022] [Accepted: 10/14/2022] [Indexed: 11/23/2022] Open
Abstract
Simple Summary The extraction of quantitative data from standard-of-care imaging modalities offers opportunities to improve the relevance and salience of imaging biomarkers used in drug development. This review aims to identify the challenges and opportunities for discovering new imaging-based biomarkers based on radiomic and volumetric assessment in the single-site solid tumor sites: breast cancer, rectal cancer, lung cancer and glioblastoma. Developing approaches to harmonize three essential areas: segmentation, validation and data sharing may expedite regulatory approval and adoption of novel cancer imaging biomarkers. Abstract Clinical trials for oncology drug development have long relied on surrogate outcome biomarkers that assess changes in tumor burden to accelerate drug registration (i.e., Response Evaluation Criteria in Solid Tumors version 1.1 (RECIST v1.1) criteria). Drug-induced reduction in tumor size represents an imperfect surrogate marker for drug activity and yet a radiologically determined objective response rate is a widely used endpoint for Phase 2 trials. With the addition of therapies targeting complex biological systems such as immune system and DNA damage repair pathways, incorporation of integrative response and outcome biomarkers may add more predictive value. We performed a review of the relevant literature in four representative tumor types (breast cancer, rectal cancer, lung cancer and glioblastoma) to assess the preparedness of volumetric and radiomics metrics as clinical trial endpoints. We identified three key areas—segmentation, validation and data sharing strategies—where concerted efforts are required to enable progress of volumetric- and radiomics-based clinical trial endpoints for wider clinical implementation.
Collapse
|
39
|
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
|
40
|
Jang SJ, Kunze KN, Vigdorchik JM, Jerabek SA, Mayman DJ, Sculco PK. John Charnley Award: Deep Learning Prediction of Hip Joint Center on Standard Pelvis Radiographs. J Arthroplasty 2022; 37:S400-S407.e1. [PMID: 35304298 DOI: 10.1016/j.arth.2022.03.033] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 03/04/2022] [Accepted: 03/08/2022] [Indexed: 02/02/2023] Open
Abstract
BACKGROUND Accurate hip joint center (HJC) determination is critical for preoperative planning, intraoperative execution, clinical outcomes after total hip arthroplasty, and commonly used classification systems in primary and revision hip replacement. However, current methods of preoperative HJC estimation are prone to subjectivity and human error. The purpose of the study was to leverage deep learning (DL) to develop a rapid and objective HJC estimation tool on anteroposterior (AP) pelvis radiographs. METHODS Radiographs from 3,965 patients (7,930 hips) were included. A DL model workflow was created to detect bony landmarks and estimate HJC based on a pelvic height ratio method. The workflow was utilized to conduct a grid-search for optimal nonspecific, sex-specific, and patient-specific (using contralateral hip) pelvic height ratios on the training/validation cohort (6,344 hips). Algorithm performance was assessed on an independent testing cohort for HJC estimation comparison. RESULTS The algorithm estimated HJC for the testing cohort at a rate of 0.65 seconds/hip based on features in AP radiographs alone. The model predicted HJC within 5 mm of error for 80% of hips using nonspecific ratios, which increased to 83% with sex-specific and 91% with patient-specific pelvic height ratio models. Mean error decreased utilizing the patient-specific model (3.09 ± 1.69 mm, P < .001). CONCLUSION Using DL, we developed nonspecific, sex-specific, and patient-specific models capable of estimating native HJC on AP pelvis radiographs. This tool may provide clinical value when considering preoperative component position in patients planned to undergo THA and in reducing the subjective variability in HJC estimation. LEVEL OF EVIDENCE Diagnostic, level IV.
Collapse
Affiliation(s)
- Seong Jun Jang
- Weill Cornell College of Medicine, New York, New York; Department of Orthopedic Surgery, Hospital for Special Surgery, New York, New York
| | - Kyle N Kunze
- Department of Orthopedic Surgery, Hospital for Special Surgery, New York, New York
| | - Jonathan M Vigdorchik
- Department of Orthopedic Surgery, Hospital for Special Surgery, New York, New York; Adult Reconstruction and Joint Replacement Service, Hospital for Special Surgery, New York, New York
| | - Seth A Jerabek
- Department of Orthopedic Surgery, Hospital for Special Surgery, New York, New York; Adult Reconstruction and Joint Replacement Service, Hospital for Special Surgery, New York, New York
| | - David J Mayman
- Department of Orthopedic Surgery, Hospital for Special Surgery, New York, New York; Adult Reconstruction and Joint Replacement Service, Hospital for Special Surgery, New York, New York
| | - Peter K Sculco
- Department of Orthopedic Surgery, Hospital for Special Surgery, New York, New York; Adult Reconstruction and Joint Replacement Service, Hospital for Special Surgery, New York, New York
| |
Collapse
|
41
|
Li CH, Cai D, Zhong ME, Lv MY, Huang ZP, Zhu Q, Hu C, Qi H, Wu X, Gao F. Multi-Size Deep Learning Based Preoperative Computed Tomography Signature for Prognosis Prediction of Colorectal Cancer. Front Genet 2022; 13:880093. [PMID: 35646105 PMCID: PMC9133721 DOI: 10.3389/fgene.2022.880093] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Accepted: 04/25/2022] [Indexed: 11/13/2022] Open
Abstract
Background: Preoperative and postoperative evaluation of colorectal cancer (CRC) patients is crucial for subsequent treatment guidance. Our study aims to provide a timely and rapid assessment of the prognosis of CRC patients with deep learning according to non-invasive preoperative computed tomography (CT) and explore the underlying biological explanations.Methods: A total of 808 CRC patients with preoperative CT (development cohort: n = 426, validation cohort: n = 382) were enrolled in our study. We proposed a novel end-to-end Multi-Size Convolutional Neural Network (MSCNN) to predict the risk of CRC recurrence with CT images (CT signature). The prognostic performance of CT signature was evaluated by Kaplan-Meier curve. An integrated nomogram was constructed to improve the clinical utility of CT signature by combining with other clinicopathologic factors. Further visualization and correlation analysis for CT deep features with paired gene expression profiles were performed to reveal the molecular characteristics of CRC tumors learned by MSCNN in radiographic imaging.Results: The Kaplan-Meier analysis showed that CT signature was a significant prognostic factor for CRC disease-free survival (DFS) prediction [development cohort: hazard ratio (HR): 50.7, 95% CI: 28.4–90.6, p < 0.001; validation cohort: HR: 2.04, 95% CI: 1.44–2.89, p < 0.001]. Multivariable analysis confirmed the independence prognostic value of CT signature (development cohort: HR: 30.7, 95% CI: 19.8–69.3, p < 0.001; validation cohort: HR: 1.83, 95% CI: 1.19–2.83, p = 0.006). Dimension reduction and visualization of CT deep features demonstrated a high correlation with the prognosis of CRC patients. Functional pathway analysis further indicated that CRC patients with high CT signature presented down-regulation of several immunology pathways. Correlation analysis found that CT deep features were mainly associated with activation of metabolic and proliferative pathways.Conclusions: Our deep learning based preoperative CT signature can effectively predict prognosis of CRC patients. Integration analysis of multi-omic data revealed that some molecular characteristics of CRC tumor can be captured by deep learning in CT images.
Collapse
Affiliation(s)
- Cheng-Hang Li
- Department of Colorectal Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Institute of Gastroenterology, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Disease, Supported by National Key Clinical Discipline, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- School of Computer Science and Engineering, Guangzhou Higher Education Mega Center, Sun Yat-sen University, Guangzhou, China
| | - Du Cai
- Department of Colorectal Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Institute of Gastroenterology, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Disease, Supported by National Key Clinical Discipline, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Min-Er Zhong
- Department of Colorectal Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Institute of Gastroenterology, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Disease, Supported by National Key Clinical Discipline, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Min-Yi Lv
- Department of Colorectal Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Institute of Gastroenterology, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Disease, Supported by National Key Clinical Discipline, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Ze-Ping Huang
- Department of Colorectal Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Institute of Gastroenterology, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Disease, Supported by National Key Clinical Discipline, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Qiqi Zhu
- Ningbo Medical Center Lihuili Hospital, Ningbo, China
| | - Chuling Hu
- Department of Colorectal Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Institute of Gastroenterology, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Disease, Supported by National Key Clinical Discipline, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Haoning Qi
- Department of Colorectal Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Institute of Gastroenterology, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Disease, Supported by National Key Clinical Discipline, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xiaojian Wu
- Department of Colorectal Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Institute of Gastroenterology, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Disease, Supported by National Key Clinical Discipline, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- *Correspondence: Xiaojian Wu, ; Feng Gao,
| | - Feng Gao
- Department of Colorectal Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Institute of Gastroenterology, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Disease, Supported by National Key Clinical Discipline, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- *Correspondence: Xiaojian Wu, ; Feng Gao,
| |
Collapse
|
42
|
Stadlbauer A, Marhold F, Oberndorfer S, Heinz G, Buchfelder M, Kinfe TM, Meyer-Bäse A. Radiophysiomics: Brain Tumors Classification by Machine Learning and Physiological MRI Data. Cancers (Basel) 2022; 14:2363. [PMID: 35625967 PMCID: PMC9139355 DOI: 10.3390/cancers14102363] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 05/04/2022] [Accepted: 05/09/2022] [Indexed: 01/06/2023] Open
Abstract
The precise initial characterization of contrast-enhancing brain tumors has significant consequences for clinical outcomes. Various novel neuroimaging methods have been developed to increase the specificity of conventional magnetic resonance imaging (cMRI) but also the increased complexity of data analysis. Artificial intelligence offers new options to manage this challenge in clinical settings. Here, we investigated whether multiclass machine learning (ML) algorithms applied to a high-dimensional panel of radiomic features from advanced MRI (advMRI) and physiological MRI (phyMRI; thus, radiophysiomics) could reliably classify contrast-enhancing brain tumors. The recently developed phyMRI technique enables the quantitative assessment of microvascular architecture, neovascularization, oxygen metabolism, and tissue hypoxia. A training cohort of 167 patients suffering from one of the five most common brain tumor entities (glioblastoma, anaplastic glioma, meningioma, primary CNS lymphoma, or brain metastasis), combined with nine common ML algorithms, was used to develop overall 135 classifiers. Multiclass classification performance was investigated using tenfold cross-validation and an independent test cohort. Adaptive boosting and random forest in combination with advMRI and phyMRI data were superior to human reading in accuracy (0.875 vs. 0.850), precision (0.862 vs. 0.798), F-score (0.774 vs. 0.740), AUROC (0.886 vs. 0.813), and classification error (5 vs. 6). The radiologists, however, showed a higher sensitivity (0.767 vs. 0.750) and specificity (0.925 vs. 0.902). We demonstrated that ML-based radiophysiomics could be helpful in the clinical routine diagnosis of contrast-enhancing brain tumors; however, a high expenditure of time and work for data preprocessing requires the inclusion of deep neural networks.
Collapse
Affiliation(s)
- Andreas Stadlbauer
- Institute of Medical Radiology, University Clinic St. Pölten, Karl Landsteiner University of Health Sciences, A-3100 St. Pölten, Austria;
- Department of Neurosurgery, Friedrich-Alexander University (FAU) Erlangen-Nürnberg, D-91054 Erlangen, Germany; (M.B.); (T.M.K.)
| | - Franz Marhold
- Department of Neurosurgery, University Clinic of St. Pölten, Karl Landsteiner University of Health Sciences, A-3100 St. Pölten, Austria;
| | - Stefan Oberndorfer
- Department of Neurology, University Clinic of St. Pölten, Karl Landsteiner University of Health Sciences, A-3100 St. Pölten, Austria;
| | - Gertraud Heinz
- Institute of Medical Radiology, University Clinic St. Pölten, Karl Landsteiner University of Health Sciences, A-3100 St. Pölten, Austria;
| | - Michael Buchfelder
- Department of Neurosurgery, Friedrich-Alexander University (FAU) Erlangen-Nürnberg, D-91054 Erlangen, Germany; (M.B.); (T.M.K.)
| | - Thomas M. Kinfe
- Department of Neurosurgery, Friedrich-Alexander University (FAU) Erlangen-Nürnberg, D-91054 Erlangen, Germany; (M.B.); (T.M.K.)
- Division of Functional Neurosurgery and Stereotaxy, Friedrich-Alexander University (FAU) Erlangen-Nürnberg, D-91054 Erlangen, Germany
| | - Anke Meyer-Bäse
- Department of Scientific Computing, Florida State University, 400 Dirac Science Library, Tallahassee, FL 32306-4120, USA;
| |
Collapse
|