1
|
Ahn S, Hong Y, Park S, Cho Y, Hwang I, Na JM, Lee H, Min BH, Lee JH, Kim JJ, Kim KM. Development and application of deep learning-based diagnostics for pathologic diagnosis of gastric endoscopic submucosal dissection specimens. Gastric Cancer 2025:10.1007/s10120-025-01612-y. [PMID: 40232558 DOI: 10.1007/s10120-025-01612-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2025] [Accepted: 03/26/2025] [Indexed: 04/16/2025]
Abstract
BACKGROUND Accurate diagnosis of ESD specimens is crucial for managing early gastric cancer. Identifying tumor areas in serially sectioned ESD specimens requires experience and is time-consuming. This study aimed to develop and evaluate a deep learning model for diagnosing ESD specimens. METHODS Whole-slide images of 366 ESD specimens of adenocarcinoma were analyzed, with 2257 annotated regions of interest (tumor and muscularis mucosa) and 83,839 patch images. The development set was divided into training and internal validation sets. Tissue segmentation performance was evaluated using the internal validation set. A detection algorithm for tumor and submucosal invasion at the whole-slide image level was developed, and its performance was evaluated using a test set. RESULTS The model achieved Dice coefficients of 0.85 and 0.79 for segmentation of tumor and muscularis mucosa, respectively. In the test set, the diagnostic performance of tumor detection, measured by the AUROC, was 0.995, with a specificity of 1.000 and a sensitivity of 0.947. For detecting submucosal invasion, the model achieved an AUROC of 0.981, with a specificity of 0.956 and a sensitivity of 0.907. Pathologists' performance in diagnosing ESD specimens was evaluated with and without assistance from the deep learning model, and the model significantly reduced the mean diagnosis time (747 s without assistance vs. 478 s with assistance, P < 0.001). CONCLUSION The deep learning model demonstrated satisfactory performance in tissue segmentation and high accuracy in detecting tumors and submucosal invasion. This model can potentially serve as a screening tool in the histopathological diagnosis of ESD specimens.
Collapse
Affiliation(s)
- Soomin Ahn
- Department of Pathology and Translational Genomics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Yiyu Hong
- Department of R&D Center, Arontier Co., Ltd, Seoul, South Korea
| | - Sujin Park
- Department of Pathology and Translational Genomics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Yunjoo Cho
- Department of Pathology and Translational Genomics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Inwoo Hwang
- Department of Pathology and Translational Genomics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Ji Min Na
- Department of Pathology and Translational Genomics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Hyuk Lee
- Department of Internal Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Byung-Hoon Min
- Department of Internal Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Jun Haeng Lee
- Department of Internal Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Jae J Kim
- Department of Internal Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Kyoung-Mee Kim
- Department of Pathology and Translational Genomics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea.
| |
Collapse
|
2
|
Li R, Li J, Wang Y, Liu X, Xu W, Sun R, Xue B, Zhang X, Ai Y, Du Y, Jiang J. The artificial intelligence revolution in gastric cancer management: clinical applications. Cancer Cell Int 2025; 25:111. [PMID: 40119433 PMCID: PMC11929235 DOI: 10.1186/s12935-025-03756-4] [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: 08/27/2024] [Accepted: 03/18/2025] [Indexed: 03/24/2025] Open
Abstract
Nowadays, gastric cancer has become a significant issue in the global cancer burden, and its impact cannot be ignored. The rapid development of artificial intelligence technology is attempting to address this situation, aiming to change the clinical management landscape of gastric cancer fundamentally. In this transformative change, machine learning and deep learning, as two core technologies, play a pivotal role, bringing unprecedented innovations and breakthroughs in the diagnosis, treatment, and prognosis evaluation of gastric cancer. This article comprehensively reviews the latest research status and application of artificial intelligence algorithms in gastric cancer, covering multiple dimensions such as image recognition, pathological analysis, personalized treatment, and prognosis risk assessment. These applications not only significantly improve the sensitivity of gastric cancer risk monitoring, the accuracy of diagnosis, and the precision of survival prognosis but also provide robust data support and a scientific basis for clinical decision-making. The integration of artificial intelligence, from optimizing the diagnosis process and enhancing diagnostic efficiency to promoting the practice of precision medicine, demonstrates its promising prospects for reshaping the treatment model of gastric cancer. Although most of the current AI-based models have not been widely used in clinical practice, with the continuous deepening and expansion of precision medicine, we have reason to believe that a new era of AI-driven gastric cancer care is approaching.
Collapse
Affiliation(s)
- Runze Li
- Hebei University of Traditional Chinese Medicine, Hebei, 050011, China
| | - Jingfan Li
- Hebei University of Traditional Chinese Medicine, Hebei, 050011, China
| | - Yuman Wang
- Hebei University of Traditional Chinese Medicine, Hebei, 050011, China
| | - Xiaoyu Liu
- Hebei University of Traditional Chinese Medicine, Hebei, 050011, China
| | - Weichao Xu
- Hebei University of Traditional Chinese Medicine, Hebei, 050011, China
- Hebei Hospital of Traditional Chinese Medicine, Hebei, 050011, China
| | - Runxue Sun
- Hebei Hospital of Traditional Chinese Medicine, Hebei, 050011, China
| | - Binqing Xue
- Hebei University of Traditional Chinese Medicine, Hebei, 050011, China
| | - Xinqian Zhang
- Hebei University of Traditional Chinese Medicine, Hebei, 050011, China
| | - Yikun Ai
- North China University of Science and Technology, Tanshan 063000, China
| | - Yanru Du
- Hebei Hospital of Traditional Chinese Medicine, Hebei, 050011, China.
- Hebei Provincial Key Laboratory of Integrated Traditional and Western Medicine Research on Gastroenterology, Hebei, 050011, China.
- Hebei Key Laboratory of Turbidity and Toxicology, Hebei, 050011, China.
| | - Jianming Jiang
- Hebei University of Traditional Chinese Medicine, Hebei, 050011, China.
- Hebei Hospital of Traditional Chinese Medicine, Hebei, 050011, China.
| |
Collapse
|
3
|
Chen S, Ding P, Guo H, Meng L, Zhao Q, Li C. Applications of artificial intelligence in digital pathology for gastric cancer. Front Oncol 2024; 14:1437252. [PMID: 39529836 PMCID: PMC11551048 DOI: 10.3389/fonc.2024.1437252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Accepted: 10/07/2024] [Indexed: 11/16/2024] Open
Abstract
Gastric cancer is one of the most common cancers and is one of the leading causes of cancer-related deaths in worldwide. Early diagnosis and treatment are essential for a positive outcome. The integration of artificial intelligence in the pathology field is increasingly widespread, including histopathological images analysis. In recent years, the application of digital pathology technology emerged as a potential solution to enhance the understanding and management of gastric cancer. Through sophisticated image analysis algorithms, artificial intelligence technologies facilitate the accuracy and sensitivity of gastric cancer diagnosis and treatment and personalized therapeutic strategies. This review aims to evaluate the current landscape and future potential of artificial intelligence in transforming gastric cancer pathology, so as to provide ideas for future research.
Collapse
Affiliation(s)
- Sheng Chen
- School of Clinical Medicine, Hebei University, Affiliated Hospital of Hebei University, Baoding, China
| | - Ping’an Ding
- The Third Department of Surgery, the Fourth Hospital of Hebei Medical University, Shijiazhuang Hebei, China
- Hebei Key Laboratory of Precision Diagnosis and Comprehensive Treatment of Gastric Cancer, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
- Big Data Analysis and Mining Application for Precise Diagnosis and Treatment of Gastric Cancer Hebei Provincial Engineering Research Center, Shijiazhuang, Hebei, China
| | - Honghai Guo
- The Third Department of Surgery, the Fourth Hospital of Hebei Medical University, Shijiazhuang Hebei, China
- Hebei Key Laboratory of Precision Diagnosis and Comprehensive Treatment of Gastric Cancer, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
- Big Data Analysis and Mining Application for Precise Diagnosis and Treatment of Gastric Cancer Hebei Provincial Engineering Research Center, Shijiazhuang, Hebei, China
| | - Lingjiao Meng
- The Third Department of Surgery, the Fourth Hospital of Hebei Medical University, Shijiazhuang Hebei, China
- Hebei Key Laboratory of Precision Diagnosis and Comprehensive Treatment of Gastric Cancer, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
- Big Data Analysis and Mining Application for Precise Diagnosis and Treatment of Gastric Cancer Hebei Provincial Engineering Research Center, Shijiazhuang, Hebei, China
| | - Qun Zhao
- The Third Department of Surgery, the Fourth Hospital of Hebei Medical University, Shijiazhuang Hebei, China
- Hebei Key Laboratory of Precision Diagnosis and Comprehensive Treatment of Gastric Cancer, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
- Big Data Analysis and Mining Application for Precise Diagnosis and Treatment of Gastric Cancer Hebei Provincial Engineering Research Center, Shijiazhuang, Hebei, China
| | - Cong Li
- School of Clinical Medicine, Hebei University, Affiliated Hospital of Hebei University, Baoding, China
- Department of Hepatobiliary Surgery, Affiliated Hospital of Hebei University, Baoding, China
| |
Collapse
|
4
|
Shin JY, Son J, Kong ST, Park J, Park B, Park KH, Jung KH, Park SJ. Clinical Utility of Deep Learning Assistance for Detecting Various Abnormal Findings in Color Retinal Fundus Images: A Reader Study. Transl Vis Sci Technol 2024; 13:34. [PMID: 39441571 PMCID: PMC11512572 DOI: 10.1167/tvst.13.10.34] [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: 07/27/2023] [Accepted: 02/28/2024] [Indexed: 10/25/2024] Open
Abstract
Purpose To evaluate the clinical usefulness of a deep learning-based detection device for multiple abnormal findings on retinal fundus photographs for readers with varying expertise. Methods Fourteen ophthalmologists (six residents, eight specialists) assessed 399 fundus images with respect to 12 major ophthalmologic findings, with or without the assistance of a deep learning algorithm, in two separate reading sessions. Sensitivity, specificity, and reading time per image were compared. Results With algorithmic assistance, readers significantly improved in sensitivity for all 12 findings (P < 0.05) but tended to be less specific (P < 0.05) for hemorrhage, drusen, membrane, and vascular abnormality, more profoundly so in residents. Sensitivity without algorithmic assistance was significantly lower in residents (23.1%∼75.8%) compared to specialists (55.1%∼97.1%) in nine findings, but it improved to similar levels with algorithmic assistance (67.8%∼99.4% in residents, 83.2%∼99.5% in specialists) with only hemorrhage remaining statistically significantly lower. Variances in sensitivity were significantly reduced for all findings. Reading time per image decreased in images with fewer than three findings per image, more profoundly in residents. When simulated based on images acquired from a health screening center, average reading time was estimated to be reduced by 25.9% (from 16.4 seconds to 12.1 seconds per image) for residents, and by 2.0% (from 9.6 seconds to 9.4 seconds) for specialists. Conclusions Deep learning-based computer-assisted detection devices increase sensitivity, reduce inter-reader variance in sensitivity, and reduce reading time in less complicated images. Translational Relevance This study evaluated the influence that algorithmic assistance in detecting abnormal findings on retinal fundus photographs has on clinicians, possibly predicting its influence on clinical application.
Collapse
Affiliation(s)
- Joo Young Shin
- Department of Ophthalmology, Seoul Metropolitan Government Seoul National University Boramae Medical Centre, Seoul, Republic of Korea
| | | | | | | | | | - Kyu Hyung Park
- Department of Ophthalmology, Seoul National University College of Medicine, Seoul National University Bundang Hospital, Seongnam, Republic of Korea
| | - Kyu-Hwan Jung
- VUNO Inc., Seoul, Republic of Korea
- Department of Medical Device Research and Management, Samsung Advanced Institute for Health Sciences and Technology, Sungkyunkwan University, Seoul, Republic of Korea
| | - Sang Jun Park
- Department of Ophthalmology, Seoul National University College of Medicine, Seoul National University Bundang Hospital, Seongnam, Republic of Korea
| |
Collapse
|
5
|
Yilmaz F, Brickman A, Najdawi F, Yakirevich E, Egger R, Resnick MB. Advancing Artificial Intelligence Integration Into the Pathology Workflow: Exploring Opportunities in Gastrointestinal Tract Biopsies. J Transl Med 2024; 104:102043. [PMID: 38431118 DOI: 10.1016/j.labinv.2024.102043] [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: 10/30/2023] [Revised: 02/14/2024] [Accepted: 02/26/2024] [Indexed: 03/05/2024] Open
Abstract
This review aims to present a comprehensive overview of the current landscape of artificial intelligence (AI) applications in the analysis of tubular gastrointestinal biopsies. These publications cover a spectrum of conditions, ranging from inflammatory ailments to malignancies. Moving beyond the conventional diagnosis based on hematoxylin and eosin-stained whole-slide images, the review explores additional implications of AI, including its involvement in interpreting immunohistochemical results, molecular subtyping, and the identification of cellular spatial biomarkers. Furthermore, the review examines how AI can contribute to enhancing the quality and control of diagnostic processes, introducing new workflow options, and addressing the limitations and caveats associated with current AI platforms in this context.
Collapse
Affiliation(s)
- Fazilet Yilmaz
- The Warren Alpert Medical School of Brown University, Rhode Island Hospital, Providence, Rhode Island
| | - Arlen Brickman
- The Warren Alpert Medical School of Brown University, Rhode Island Hospital, Providence, Rhode Island
| | - Fedaa Najdawi
- The Warren Alpert Medical School of Brown University, Rhode Island Hospital, Providence, Rhode Island
| | - Evgeny Yakirevich
- The Warren Alpert Medical School of Brown University, Rhode Island Hospital, Providence, Rhode Island
| | | | - Murray B Resnick
- The Warren Alpert Medical School of Brown University, Rhode Island Hospital, Providence, Rhode Island.
| |
Collapse
|
6
|
Fang S, Liu Z, Qiu Q, Tang Z, Yang Y, Kuang Z, Du X, Xiao S, Liu Y, Luo Y, Gu L, Tian L, Liang X, Fan G, Zhang Y, Zhang P, Zhou W, Liu X, Tian J, Wei W. Diagnosing and grading gastric atrophy and intestinal metaplasia using semi-supervised deep learning on pathological images: development and validation study. Gastric Cancer 2024; 27:343-354. [PMID: 38095766 PMCID: PMC10896941 DOI: 10.1007/s10120-023-01451-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Accepted: 11/09/2023] [Indexed: 02/28/2024]
Abstract
OBJECTIVE Patients with gastric atrophy and intestinal metaplasia (IM) were at risk for gastric cancer, necessitating an accurate risk assessment. We aimed to establish and validate a diagnostic approach for gastric biopsy specimens using deep learning and OLGA/OLGIM for individual gastric cancer risk classification. METHODS In this study, we prospectively enrolled 545 patients suspected of atrophic gastritis during endoscopy from 13 tertiary hospitals between December 22, 2017, to September 25, 2020, with a total of 2725 whole-slide images (WSIs). Patients were randomly divided into a training set (n = 349), an internal validation set (n = 87), and an external validation set (n = 109). Sixty patients from the external validation set were randomly selected and divided into two groups for an observer study, one with the assistance of algorithm results and the other without. We proposed a semi-supervised deep learning algorithm to diagnose and grade IM and atrophy, and we compared it with the assessments of 10 pathologists. The model's performance was evaluated based on the area under the curve (AUC), sensitivity, specificity, and weighted kappa value. RESULTS The algorithm, named GasMIL, was established and demonstrated encouraging performance in diagnosing IM (AUC 0.884, 95% CI 0.862-0.902) and atrophy (AUC 0.877, 95% CI 0.855-0.897) in the external test set. In the observer study, GasMIL achieved an 80% sensitivity, 85% specificity, a weighted kappa value of 0.61, and an AUC of 0.953, surpassing the performance of all ten pathologists in diagnosing atrophy. Among the 10 pathologists, GasMIL's AUC ranked second in OLGA (0.729, 95% CI 0.625-0.833) and fifth in OLGIM (0.792, 95% CI 0.688-0.896). With the assistance of GasMIL, pathologists demonstrated improved AUC (p = 0.013), sensitivity (p = 0.014), and weighted kappa (p = 0.016) in diagnosing IM, and improved specificity (p = 0.007) in diagnosing atrophy compared to pathologists working alone. CONCLUSION GasMIL shows the best overall performance in diagnosing IM and atrophy when compared to pathologists, significantly enhancing their diagnostic capabilities.
Collapse
Affiliation(s)
- Shuangshuang Fang
- Beijing Key Laboratory of Functional Gastrointestinal Disorders Diagnosis and Treatment of Traditional Chinese Medicine; Department of Gastroenterology, Wangjing Hospital, China Academy of Chinese Medical Sciences, No. 6, Zhonghuan South Road, Wangjing, Beijing, 100102, China
| | - Zhenyu Liu
- CAS Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing, 100190, China
- School of Artificial Intelligence, University of Chinese Academy of Science, Beijing, 100190, China
| | - Qi Qiu
- CAS Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing, 100190, China
- School of Artificial Intelligence, University of Chinese Academy of Science, Beijing, 100190, China
| | - Zhenchao Tang
- Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, School of Engineering Medicine, Beihang University, Beijing, 100191, China
| | - Yang Yang
- Beijing Key Laboratory of Functional Gastrointestinal Disorders Diagnosis and Treatment of Traditional Chinese Medicine; Department of Gastroenterology, Wangjing Hospital, China Academy of Chinese Medical Sciences, No. 6, Zhonghuan South Road, Wangjing, Beijing, 100102, China
| | - Zhongsheng Kuang
- Department of Pathology, The First Affiliated Hospital of Guangdong University of Traditional Chinese Medicine, Guangzhou, 510405, China
| | - Xiaohua Du
- Department of Pathology, Guangdong Provincial Hospital of Traditional Chinese Medicine, Guangzhou, 510120, China
| | - Shanshan Xiao
- Department of Pathology, The First Affiliated Hospital of Guangdong University of Traditional Chinese Medicine, Guangzhou, 510405, China
| | - Yanyan Liu
- Department of Pathology, The First Affiliated Hospital of Guangdong University of Traditional Chinese Medicine, Guangzhou, 510405, China
| | - Yuanbin Luo
- Department of Pathology, Gansu Provincial Hospital of Traditional Chinese Medicine, Lanzhou, 730050, China
| | - Liping Gu
- Department of Pathology, Gansu Provincial Hospital of Traditional Chinese Medicine, Lanzhou, 730050, China
| | - Li Tian
- Department of Pathology, Gansu Provincial Hospital of Traditional Chinese Medicine, Lanzhou, 730050, China
| | - Xiaoxia Liang
- Department of Pathology, Shanxi Provincial Hospital of Traditional Chinese Medicine, Taiyuan, 030012, China
| | - Guiling Fan
- Department of Pathology, Shanxi Provincial Hospital of Traditional Chinese Medicine, Taiyuan, 030012, China
| | - Yu Zhang
- Department of Pathology, Shanxi Provincial Hospital of Traditional Chinese Medicine, Taiyuan, 030012, China
| | - Ping Zhang
- Department of Pathology, Wangjing Hospital, China Academy of Chinese Medical Sciences, Beijing, 100102, China
| | - Weixun Zhou
- Department of Pathology, Peking Union Medical College Hospital, Beijing, 100730, China
| | - Xiuli Liu
- Department of Pathology and Immunology, Washington University, St. Louis, MO, 98195, USA
| | - Jie Tian
- CAS Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing, 100190, China
- School of Artificial Intelligence, University of Chinese Academy of Science, Beijing, 100190, China
- Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, School of Engineering Medicine, Beihang University, Beijing, 100191, China
- Engineering Research Center of Molecular and Neuro Imaging of Ministry of Education, School of Life Science and Technology, Xidian University, Xi'an, 710126, Shaanxi, China
| | - Wei Wei
- Beijing Key Laboratory of Functional Gastrointestinal Disorders Diagnosis and Treatment of Traditional Chinese Medicine; Department of Gastroenterology, Wangjing Hospital, China Academy of Chinese Medical Sciences, No. 6, Zhonghuan South Road, Wangjing, Beijing, 100102, China.
| |
Collapse
|
7
|
Yousif M, Pantanowitz L. Artificial Intelligence-Enabled Gastric Cancer Interpretations: Are We There yet? Surg Pathol Clin 2023; 16:673-686. [PMID: 37863559 DOI: 10.1016/j.path.2023.05.005] [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] [Indexed: 10/22/2023]
Abstract
The integration of digital pathology and artificial intelligence (AI) is revolutionizing pathology by providing pathologists with new tools to improve workflow, enhance diagnostic accuracy, and undertake novel discovery. The capability of AI to recognize patterns and features in digital images beyond human perception is particularly valuable, thereby providing additional information for prognostic and predictive purposes. AI-based tools diagnose gastric carcinoma in digital images, detect gastric carcinoma metastases in lymph nodes, automate Ki-67 scoring in gastric neuroendocrine tumors, and quantify tumor-infiltrating lymphocytes. This article provides an overview of all of these applications of AI pertaining to gastric cancer.
Collapse
Affiliation(s)
- Mustafa Yousif
- Department of Pathology, University of Michigan, NCRC Building 35, 2800 Plymouth Road, Ann Arbor, MI 48109, USA.
| | - Liron Pantanowitz
- Department of Pathology, UPMC Shadyside Hospital, 5150 Centre Avenue Cancer Pavilion, POB2, Suite 3B, Room 347, Pittsburgh, PA 15232, USA
| |
Collapse
|
8
|
Oh Y, Bae GE, Kim KH, Yeo MK, Ye JC. Multi-Scale Hybrid Vision Transformer for Learning Gastric Histology: AI-Based Decision Support System for Gastric Cancer Treatment. IEEE J Biomed Health Inform 2023; 27:4143-4153. [PMID: 37192031 DOI: 10.1109/jbhi.2023.3276778] [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: 05/18/2023]
Abstract
Gastric endoscopic screening is an effective way to decide appropriate gastric cancer treatment at an early stage, reducing gastric cancer-associated mortality rate. Although artificial intelligence has brought a great promise to assist pathologist to screen digitalized endoscopic biopsies, existing artificial intelligence systems are limited to be utilized in planning gastric cancer treatment. We propose a practical artificial intelligence-based decision support system that enables five subclassifications of gastric cancer pathology, which can be directly matched to general gastric cancer treatment guidance. The proposed framework is designed to efficiently differentiate multi-classes of gastric cancer through multiscale self-attention mechanism using 2-stage hybrid vision transformer networks, by mimicking the way how human pathologists understand histology. The proposed system demonstrates its reliable diagnostic performance by achieving class-average sensitivity of above 0.85 for multicentric cohort tests. Moreover, the proposed system demonstrates its great generalization capability on gastrointestinal track organ cancer by achieving the best class-average sensitivity among contemporary networks. Furthermore, in the observational study, artificial intelligence-assisted pathologists show significantly improved diagnostic sensitivity within saved screening time compared to human pathologists. Our results demonstrate that the proposed artificial intelligence system has a great potential for providing presumptive pathologic opinion and supporting decision of appropriate gastric cancer treatment in practical clinical settings.
Collapse
|
9
|
Meng Z, Wang G, Su F, Liu Y, Wang Y, Yang J, Luo J, Cao F, Zhen P, Huang B, Yin Y, Zhao Z, Guo L. A Deep Learning-Based System Trained for Gastrointestinal Stromal Tumor Screening Can Identify Multiple Types of Soft Tissue Tumors. THE AMERICAN JOURNAL OF PATHOLOGY 2023; 193:899-912. [PMID: 37068638 DOI: 10.1016/j.ajpath.2023.03.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 03/26/2023] [Accepted: 03/28/2023] [Indexed: 04/19/2023]
Abstract
The accuracy and timeliness of the pathologic diagnosis of soft tissue tumors (STTs) critically affect treatment decision and patient prognosis. Thus, it is crucial to make a preliminary judgement on whether the tumor is benign or malignant with hematoxylin and eosin-stained images. A deep learning-based system, Soft Tissue Tumor Box (STT-BOX), is presented herein, with only hematoxylin and eosin images for malignant STT identification from benign STTs with histopathologic similarity. STT-BOX assumed gastrointestinal stromal tumor as a baseline for malignant STT evaluation, and distinguished gastrointestinal stromal tumor from leiomyoma and schwannoma with 100% area under the curve in patients from three hospitals, which achieved higher accuracy than the interpretation of experienced pathologists. Particularly, this system performed well on six common types of malignant STTs from The Cancer Genome Atlas data set, accurately highlighting the malignant mass lesion. STT-BOX was able to distinguish ovarian malignant sex-cord stromal tumors without any fine-tuning. This study included mesenchymal tumors that originated from the digestive system, bone and soft tissues, and reproductive system, where the high accuracy of migration verification may reveal the morphologic similarity of the nine types of malignant tumors. Further evaluation in a pan-STT setting would be potential and prospective, obviating the overuse of immunohistochemistry and molecular tests, and providing a practical basis for clinical treatment selection in a timely manner.
Collapse
Affiliation(s)
- Zhu Meng
- Beijing University of Posts and Telecommunications and Department of Pathology, Peking University Third Hospital, Beijing Key Laboratory of Tumor Systems Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Guangxi Wang
- Beijing University of Posts and Telecommunications and Department of Pathology, Peking University Third Hospital, Beijing Key Laboratory of Tumor Systems Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Fei Su
- Beijing University of Posts and Telecommunications and Department of Pathology, Peking University Third Hospital, Beijing Key Laboratory of Tumor Systems Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China; Beijing Key Laboratory of Network System and Network Culture, Beijing, China
| | - Yan Liu
- Beijing University of Posts and Telecommunications and Department of Pathology, Peking University Third Hospital, Beijing Key Laboratory of Tumor Systems Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Yuxiang Wang
- Beijing University of Posts and Telecommunications and Department of Pathology, Peking University Third Hospital, Beijing Key Laboratory of Tumor Systems Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Jing Yang
- Beijing University of Posts and Telecommunications and Department of Pathology, Peking University Third Hospital, Beijing Key Laboratory of Tumor Systems Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Jianyuan Luo
- Department of Medical Genetics, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Fang Cao
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Pathology, Peking University Cancer Hospital and Institute, Beijing, China
| | - Panpan Zhen
- Department of Pathology, Beijing Luhe Hospital, Capital Medical University, Beijing, China
| | - Binhua Huang
- Department of Pathology, Dongguan Houjie Hospital, Dongguan, China
| | - Yuxin Yin
- Beijing University of Posts and Telecommunications and Department of Pathology, Peking University Third Hospital, Beijing Key Laboratory of Tumor Systems Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Zhicheng Zhao
- Beijing University of Posts and Telecommunications and Department of Pathology, Peking University Third Hospital, Beijing Key Laboratory of Tumor Systems Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China; Beijing Key Laboratory of Network System and Network Culture, Beijing, China.
| | - Limei Guo
- Beijing University of Posts and Telecommunications and Department of Pathology, Peking University Third Hospital, Beijing Key Laboratory of Tumor Systems Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China.
| |
Collapse
|
10
|
Choi S, Kim S. Artificial Intelligence in the Pathology of Gastric Cancer. J Gastric Cancer 2023; 23:410-427. [PMID: 37553129 PMCID: PMC10412971 DOI: 10.5230/jgc.2023.23.e25] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Revised: 07/09/2023] [Accepted: 07/14/2023] [Indexed: 08/10/2023] Open
Abstract
Recent advances in artificial intelligence (AI) have provided novel tools for rapid and precise pathologic diagnosis. The introduction of digital pathology has enabled the acquisition of scanned slide images that are essential for the application of AI. The application of AI for improved pathologic diagnosis includes the error-free detection of potentially negligible lesions, such as a minute focus of metastatic tumor cells in lymph nodes, the accurate diagnosis of potentially controversial histologic findings, such as very well-differentiated carcinomas mimicking normal epithelial tissues, and the pathological subtyping of the cancers. Additionally, the utilization of AI algorithms enables the precise decision of the score of immunohistochemical markers for targeted therapies, such as human epidermal growth factor receptor 2 and programmed death-ligand 1. Studies have revealed that AI assistance can reduce the discordance of interpretation between pathologists and more accurately predict clinical outcomes. Several approaches have been employed to develop novel biomarkers from histologic images using AI. Moreover, AI-assisted analysis of the cancer microenvironment showed that the distribution of tumor-infiltrating lymphocytes was related to the response to the immune checkpoint inhibitor therapy, emphasizing its value as a biomarker. As numerous studies have demonstrated the significance of AI-assisted interpretation and biomarker development, the AI-based approach will advance diagnostic pathology.
Collapse
Affiliation(s)
- Sangjoon Choi
- Department of Pathology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Seokhwi Kim
- Department of Pathology, Ajou University School of Medicine, Suwon, Korea
- Department of Biomedical Sciences, Ajou University Graduate School of Medicine, Suwon, Korea.
| |
Collapse
|
11
|
Kim DH, Sun S, Cho SI, Kong HJ, Lee JW, Lee JH, Suh DH. Automated Facial Acne Lesion Detecting and Counting Algorithm for Acne Severity Evaluation and Its Utility in Assisting Dermatologists. Am J Clin Dermatol 2023:10.1007/s40257-023-00777-5. [PMID: 37160644 DOI: 10.1007/s40257-023-00777-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/27/2023] [Indexed: 05/11/2023]
Abstract
BACKGROUND Although lesion counting is an evaluation method that effectively analyzes facial acne severity, its usage is limited because of difficult implementation. OBJECTIVES We aimed to develop and validate an automated algorithm that detects and counts acne lesions by type, and to evaluate its clinical applicability as an assistance tool through a reader test. METHODS A total of 20,699 lesions (closed and open comedones, papules, nodules/cysts, and pustules) were manually labeled on 1213 facial images of 398 facial acne photography sets (frontal and both lateral views) acquired from 258 patients and used for training and validating algorithms based on a convolutional neural network for classifying five classes of acne lesions or for binary classification into noninflammatory and inflammatory lesions. RESULTS In the validation dataset, the highest mean average precision was 28.48 for the binary classification algorithm. Pearson's correlation of lesion counts between algorithm and ground-truth was 0.72 (noninflammatory) and 0.90 (inflammatory), respectively. In the reader test, eight readers (100.0%) detected and counted lesions more accurately using the algorithm compared with the reader-alone evaluation. CONCLUSIONS Overall, our algorithm demonstrated clinically applicable performance in detecting and counting facial acne lesions by type and its utility as an assistance tool for evaluating acne severity.
Collapse
Affiliation(s)
- Dong Hyo Kim
- Department of Dermatology, Seoul National University College of Medicine, Seoul, South Korea
- Acne, Rosacea, Seborrheic Dermatitis and Hidradenitis Suppurativa Research Laboratory, Seoul National University Hospital, Seoul, South Korea
| | - Sukkyu Sun
- Biomedical Research Institute, Seoul National University Hospital, Seoul, South Korea
| | - Soo Ick Cho
- Department of Dermatology, Seoul National University College of Medicine, Seoul, South Korea
| | - Hyoun-Joong Kong
- Department of Transdisciplinary Medicine, Seoul National University Hospital, Seoul, South Korea
- Department of Medicine, Seoul National University College of Medicine, Seoul, South Korea
| | - Ji Won Lee
- Department of Dermatology, Seoul National University College of Medicine, Seoul, South Korea
| | - Jun Hyo Lee
- Department of Dermatology, Seoul National University College of Medicine, Seoul, South Korea
- Acne, Rosacea, Seborrheic Dermatitis and Hidradenitis Suppurativa Research Laboratory, Seoul National University Hospital, Seoul, South Korea
| | - Dae Hun Suh
- Department of Dermatology, Seoul National University College of Medicine, Seoul, South Korea.
- Acne, Rosacea, Seborrheic Dermatitis and Hidradenitis Suppurativa Research Laboratory, Seoul National University Hospital, Seoul, South Korea.
| |
Collapse
|
12
|
Using deep learning to predict survival outcome in non-surgical cervical cancer patients based on pathological images. J Cancer Res Clin Oncol 2023:10.1007/s00432-022-04446-8. [PMID: 36653539 PMCID: PMC10356676 DOI: 10.1007/s00432-022-04446-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Accepted: 10/19/2022] [Indexed: 01/20/2023]
Abstract
PURPOSE We analyzed clinical features and the representative HE-stained pathologic images to predict 5-year overall survival via the deep-learning approach in cervical cancer patients in order to assist oncologists in designing the optimal treatment strategies. METHODS The research retrospectively collected 238 non-surgical cervical cancer patients treated with radiochemotherapy from 2014 to 2017. These patients were randomly divided into the training set (n = 165) and test set (n = 73). Then, we extract deep features after segmenting the HE-stained image into patches of size 224 × 224. A Lasso-Cox model was constructed with clinical data to predict 5-year OS. C-index evaluated this model performance with 95% CI, calibration curve, and ROC. RESULTS Based on multivariate analysis, 2 of 11 clinical characteristics (C-index 0.68) and 2 of 2048 pathomic features (C-index 0.74) and clinical-pathomic model (C-index 0.83) of nomograms predict 5-year survival in the training set, respectively. In test set, compared with the pathomic and clinical characteristics used alone, the clinical-pathomic model had an AUC of 0.750 (95% CI 0.540-0.959), the clinical predictor model had an AUC of 0.729 (95% CI 0.551-0.909), and the pathomic model AUC was 0.703 (95% CI 0.487-0.919). Based on appropriate nomogram scores, we divided patients into high-risk and low-risk groups, and Kaplan-Meier survival probability curves for both groups showed statistical differences. CONCLUSION We built a clinical-pathomic model to predict 5-year OS in non-surgical cervical cancer patients, which may be a promising method to improve the precision of personalized therapy.
Collapse
|
13
|
Ko YS, Choi YM, Kim M, Park Y, Ashraf M, Quiñones Robles WR, Kim MJ, Jang J, Yun S, Hwang Y, Jang H, Yi MY. Improving quality control in the routine practice for histopathological interpretation of gastrointestinal endoscopic biopsies using artificial intelligence. PLoS One 2022; 17:e0278542. [PMID: 36520777 PMCID: PMC9754254 DOI: 10.1371/journal.pone.0278542] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Accepted: 11/18/2022] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Colorectal and gastric cancer are major causes of cancer-related deaths. In Korea, gastrointestinal (GI) endoscopic biopsy specimens account for a high percentage of histopathologic examinations. Lack of a sufficient pathologist workforce can cause an increase in human errors, threatening patient safety. Therefore, we developed a digital pathology total solution combining artificial intelligence (AI) classifier models and pathology laboratory information system for GI endoscopic biopsy specimens to establish a post-analytic daily fast quality control (QC) system, which was applied in clinical practice for a 3-month trial run by four pathologists. METHODS AND FINDINGS Our whole slide image (WSI) classification framework comprised patch-generator, patch-level classifier, and WSI-level classifier. The classifiers were both based on DenseNet (Dense Convolutional Network). In laboratory tests, the WSI classifier achieved accuracy rates of 95.8% and 96.0% in classifying histopathological WSIs of colorectal and gastric endoscopic biopsy specimens, respectively, into three classes (Negative for dysplasia, Dysplasia, and Malignant). Classification by pathologic diagnosis and AI prediction were compared and daily reviews were conducted, focusing on discordant cases for early detection of potential human errors by the pathologists, allowing immediate correction, before the pathology report error is conveyed to the patients. During the 3-month AI-assisted daily QC trial run period, approximately 7-10 times the number of slides compared to that in the conventional monthly QC (33 months) were reviewed by pathologists; nearly 100% of GI endoscopy biopsy slides were double-checked by the AI models. Further, approximately 17-30 times the number of potential human errors were detected within an average of 1.2 days. CONCLUSIONS The AI-assisted daily QC system that we developed and established demonstrated notable improvements in QC, in quantitative, qualitative, and time utility aspects. Ultimately, we developed an independent AI-assisted post-analytic daily fast QC system that was clinically applicable and influential, which could enhance patient safety.
Collapse
Affiliation(s)
- Young Sin Ko
- Pathology Center, Seegene Medical Foundation, Seoul, Republic of Korea
| | - Yoo Mi Choi
- Pathology Center, Seegene Medical Foundation, Seoul, Republic of Korea
| | - Mujin Kim
- Graduate School of Data Science, Department of Industrial & Systems Engineering, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea
| | - Youngjin Park
- Graduate School of Data Science, Department of Industrial & Systems Engineering, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea
| | - Murtaza Ashraf
- Graduate School of Data Science, Department of Industrial & Systems Engineering, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea
| | - Willmer Rafell Quiñones Robles
- Graduate School of Data Science, Department of Industrial & Systems Engineering, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea
| | - Min-Ju Kim
- Department of Pathology, Incheon Sejong Hospital, Incheon, Republic of Korea
| | - Jiwook Jang
- AI Research Team, Digital Innovation Sector, Seegene Medical Foundation, Seoul, Republic of Korea
| | - Seokju Yun
- AI Research Team, Digital Innovation Sector, Seegene Medical Foundation, Seoul, Republic of Korea
| | - Yuri Hwang
- AI Research Team, Digital Innovation Sector, Seegene Medical Foundation, Seoul, Republic of Korea
| | - Hani Jang
- AI Research Team, Digital Innovation Sector, Seegene Medical Foundation, Seoul, Republic of Korea
| | - Mun Yong Yi
- Graduate School of Data Science, Department of Industrial & Systems Engineering, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea
| |
Collapse
|
14
|
Zhang X, Ba W, Zhao X, Wang C, Li Q, Zhang Y, Lu S, Wang L, Wang S, Song Z, Shen D. Clinical-grade endometrial cancer detection system via whole-slide images using deep learning. Front Oncol 2022; 12:1040238. [PMID: 36408137 PMCID: PMC9668742 DOI: 10.3389/fonc.2022.1040238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Accepted: 10/19/2022] [Indexed: 11/05/2022] Open
Abstract
The accurate pathological diagnosis of endometrial cancer (EC) improves the curative effect and reduces the mortality rate. Deep learning has demonstrated expert-level performance in pathological diagnosis of a variety of organ systems using whole-slide images (WSIs). It is urgent to build the deep learning system for endometrial cancer detection using WSIs. The deep learning model was trained and validated using a dataset of 601 WSIs from PUPH. The model performance was tested on three independent datasets containing a total of 1,190 WSIs. For the retrospective test, we evaluated the model performance on 581 WSIs from PUPH. In the prospective study, 317 consecutive WSIs from PUPH were collected from April 2022 to May 2022. To further evaluate the generalizability of the model, 292 WSIs were gathered from PLAHG as part of the external test set. The predictions were thoroughly analyzed by expert pathologists. The model achieved an area under the receiver operating characteristic curve (AUC), sensitivity, and specificity of 0.928, 0.924, and 0.801, respectively, on 1,190 WSIs in classifying EC and non-EC. On the retrospective dataset from PUPH/PLAGH, the model achieved an AUC, sensitivity, and specificity of 0.948/0.971, 0.928/0.947, and 0.80/0.938, respectively. On the prospective dataset, the AUC, sensitivity, and specificity were, in order, 0.933, 0.934, and 0.837. Falsely predicted results were analyzed to further improve the pathologists’ confidence in the model. The deep learning model achieved a high degree of accuracy in identifying EC using WSIs. By pre-screening the suspicious EC regions, it would serve as an assisted diagnostic tool to improve working efficiency for pathologists.
Collapse
Affiliation(s)
- Xiaobo Zhang
- Department of Pathology, Peking University People’s Hospital, Beijing, China
| | - Wei Ba
- Department of Pathology, Chinese PLA General Hospital, Beijing, China
| | - Xiaoya Zhao
- Department of Pathology, Peking University People’s Hospital, Beijing, China
| | - Chen Wang
- Department of Pathology, Peking University People’s Hospital, Beijing, China
| | - Qiting Li
- R&D Department, China Academy of Launch Vehicle Technology, Beijing, China
| | - Yinli Zhang
- Department of Pathology, Peking University People’s Hospital, Beijing, China
| | - Shanshan Lu
- Department of Pathology, Peking University People’s Hospital, Beijing, China
| | - Lang Wang
- Thorough Lab, Thorough Future, Beijing, China
| | - Shuhao Wang
- Thorough Lab, Thorough Future, Beijing, China
- *Correspondence: Danhua Shen, ; Zhigang Song, ; Shuhao Wang,
| | - Zhigang Song
- Department of Pathology, Chinese PLA General Hospital, Beijing, China
- *Correspondence: Danhua Shen, ; Zhigang Song, ; Shuhao Wang,
| | - Danhua Shen
- Department of Pathology, Peking University People’s Hospital, Beijing, China
- *Correspondence: Danhua Shen, ; Zhigang Song, ; Shuhao Wang,
| |
Collapse
|
15
|
Qiao Y, Zhao L, Luo C, Luo Y, Wu Y, Li S, Bu D, Zhao Y. Multi-modality artificial intelligence in digital pathology. Brief Bioinform 2022; 23:6702380. [PMID: 36124675 PMCID: PMC9677480 DOI: 10.1093/bib/bbac367] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Revised: 07/27/2022] [Accepted: 08/05/2022] [Indexed: 12/14/2022] Open
Abstract
In common medical procedures, the time-consuming and expensive nature of obtaining test results plagues doctors and patients. Digital pathology research allows using computational technologies to manage data, presenting an opportunity to improve the efficiency of diagnosis and treatment. Artificial intelligence (AI) has a great advantage in the data analytics phase. Extensive research has shown that AI algorithms can produce more up-to-date and standardized conclusions for whole slide images. In conjunction with the development of high-throughput sequencing technologies, algorithms can integrate and analyze data from multiple modalities to explore the correspondence between morphological features and gene expression. This review investigates using the most popular image data, hematoxylin-eosin stained tissue slide images, to find a strategic solution for the imbalance of healthcare resources. The article focuses on the role that the development of deep learning technology has in assisting doctors' work and discusses the opportunities and challenges of AI.
Collapse
Affiliation(s)
- Yixuan Qiao
- Research Center for Ubiquitous Computing Systems, Institute of Computing Technology, Chinese Academy of Sciences, Beijing 100190, China,University of Chinese Academy of Sciences, Beijing 100049, China
| | - Lianhe Zhao
- Corresponding authors: Yi Zhao, Research Center for Ubiquitous Computing Systems, Institute of Computing Technology, Chinese Academy of Sciences, Beijing 100190, China; University of Chinese Academy of Sciences; Shandong First Medical University & Shandong Academy of Medical Sciences. Tel.: +86 10 6260 0822; Fax: +86 10 6260 1356; E-mail: ; Lianhe Zhao, Research Center for Ubiquitous Computing Systems, Institute of Computing Technology, Chinese Academy of Sciences, Beijing 100190, China; University of Chinese Academy of Sciences. Tel.: +86 18513983324; E-mail:
| | - Chunlong Luo
- Research Center for Ubiquitous Computing Systems, Institute of Computing Technology, Chinese Academy of Sciences, Beijing 100190, China,University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yufan Luo
- Research Center for Ubiquitous Computing Systems, Institute of Computing Technology, Chinese Academy of Sciences, Beijing 100190, China,University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yang Wu
- Research Center for Ubiquitous Computing Systems, Institute of Computing Technology, Chinese Academy of Sciences, Beijing 100190, China
| | - Shengtong Li
- Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Dechao Bu
- Research Center for Ubiquitous Computing Systems, Institute of Computing Technology, Chinese Academy of Sciences, Beijing 100190, China
| | - Yi Zhao
- Corresponding authors: Yi Zhao, Research Center for Ubiquitous Computing Systems, Institute of Computing Technology, Chinese Academy of Sciences, Beijing 100190, China; University of Chinese Academy of Sciences; Shandong First Medical University & Shandong Academy of Medical Sciences. Tel.: +86 10 6260 0822; Fax: +86 10 6260 1356; E-mail: ; Lianhe Zhao, Research Center for Ubiquitous Computing Systems, Institute of Computing Technology, Chinese Academy of Sciences, Beijing 100190, China; University of Chinese Academy of Sciences. Tel.: +86 18513983324; E-mail:
| |
Collapse
|
16
|
Park J, Chung YR, Nose A. Comparative analysis of high- and low-level deep learning approaches in microsatellite instability prediction. Sci Rep 2022; 12:12218. [PMID: 35851285 PMCID: PMC9293930 DOI: 10.1038/s41598-022-16283-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Accepted: 07/07/2022] [Indexed: 11/09/2022] Open
Abstract
Deep learning-based approaches in histopathology can be largely divided into two categories: a high-level approach using an end-to-end model and a low-level approach using feature extractors. Although the advantages and disadvantages of both approaches are empirically well known, there exists no scientific basis for choosing a specific approach in research, and direct comparative analysis of the two approaches has rarely been performed. Using the Cancer Genomic Atlas (TCGA)-based dataset, we compared these two different approaches in microsatellite instability (MSI) prediction and analyzed morphological image features associated with MSI. Our high-level approach was based solely on EfficientNet, while our low-level approach relied on LightGBM and multiple deep learning models trained on publicly available multiclass tissue, nuclei, and gland datasets. We compared their performance and important image features. Our high-level approach showed superior performance compared to our low-level approach. In both approaches, debris, lymphocytes, and necrotic cells were revealed as important features of MSI, which is consistent with clinical knowledge. Then, during qualitative analysis, we discovered the weaknesses of our low-level approach and demonstrated that its performance can be improved by using different image features in a complementary way. We performed our study using open-access data, and we believe this study can serve as a useful basis for discovering imaging biomarkers for clinical application.
Collapse
Affiliation(s)
- Jeonghyuk Park
- Department of Physics, Graduate School of Science, The University of Tokyo, Tokyo, Japan.
| | - Yul Ri Chung
- Pathology Center, Seegene Medical Foundation, Seoul, Korea
| | - Akinao Nose
- Department of Physics, Graduate School of Science, The University of Tokyo, Tokyo, Japan.,Department of Complexity Science and Engineering, Graduate School of Frontier Sciences, The University of Tokyo, Chiba, Japan
| |
Collapse
|
17
|
Shi Z, Zhu C, Zhang Y, Wang Y, Hou W, Li X, Lu J, Guo X, Xu F, Jiang X, Wang Y, Liu J, Jin M. Deep learning for automatic diagnosis of gastric dysplasia using whole-slide histopathology images in endoscopic specimens. Gastric Cancer 2022; 25:751-760. [PMID: 35394573 DOI: 10.1007/s10120-022-01294-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/26/2021] [Accepted: 03/07/2022] [Indexed: 02/07/2023]
Abstract
BACKGROUND Distinguishing gastric epithelial regeneration change from dysplasia and histopathological diagnosis of dysplasia is subject to interobserver disagreement in endoscopic specimens. In this study, we developed a method to distinguish gastric epithelial regeneration change from dysplasia and further subclassify dysplasia. Meanwhile, optimized the cross-hospital diagnosis using domain adaption (DA). METHODS 897 whole slide images (WSIs) of endoscopic specimens from two hospitals were divided into training, internal validation, and external validation cohorts. We developed a deep learning (DL) with DA (DLDA) model to classify gastric dysplasia and epithelial regeneration change into three categories: negative for dysplasia (NFD), low-grade dysplasia (LGD), and high-grade dysplasia (HGD)/intramucosal invasion neoplasia (IMN). The diagnosis based on the DLDA model was compared to 12 pathologists using 100 gastric biopsy cases. RESULTS In the internal validation cohort, the diagnostic performance measured by the macro-averaged area under the receiver operating characteristic curve (AUC) was 0.97. In the independent external validation cohort, our DLDA models increased macro-averaged AUC from 0.67 to 0.82. In terms of the NFD and HGD cases, our model's diagnostic sensitivity, specificity, positive predictive value (PPV), and negative predictive value (NPV) were significantly higher than junior and senior pathologists. Our model's diagnostic sensitivity, NPV, was higher than specialist pathologists. CONCLUSIONS We demonstrated that our DLDA model could distinguish gastric epithelial regeneration change from dysplasia and further subclassify dysplasia in endoscopic specimens. Meanwhile, achieved significant improvement of diagnosis cross-hospital.
Collapse
Affiliation(s)
- Zhongyue Shi
- Department of Pathology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, China
| | - Chuang Zhu
- School of Artificial Intelligence, Beijing University of Posts and Telecommunications, Beijing, China
| | - Yu Zhang
- School of Artificial Intelligence, Beijing University of Posts and Telecommunications, Beijing, China
| | - Yakun Wang
- Department of Pathology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, China
| | - Weihua Hou
- Department of Pathology, PLA Joint Logistics Support Force 989 Hospital (Formerly, the 152 Central Hospital), Henan, China
| | - Xue Li
- Department of Pathology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, China
| | - Jun Lu
- Department of Pathology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, China
| | - Xinmeng Guo
- Department of Pathology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, China
| | - Feng Xu
- Department of Breast Surgery, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Xingran Jiang
- Department of Pathology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, China
| | - Ying Wang
- Department of Pathology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, China
| | - Jun Liu
- School of Artificial Intelligence, Beijing University of Posts and Telecommunications, Beijing, China
| | - Mulan Jin
- Department of Pathology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, China.
| |
Collapse
|
18
|
Choi S, Cho SI, Ma M, Park S, Pereira S, Aum BJ, Shin S, Paeng K, Yoo D, Jung W, Ock CY, Lee SH, Choi YL, Chung JH, Mok TS, Kim H, Kim S. Artificial intelligence–powered programmed death ligand 1 analyser reduces interobserver variation in tumour proportion score for non–small cell lung cancer with better prediction of immunotherapy response. Eur J Cancer 2022; 170:17-26. [DOI: 10.1016/j.ejca.2022.04.011] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 03/10/2022] [Accepted: 04/04/2022] [Indexed: 12/23/2022]
|
19
|
Zhang P, She Y, Gao J, Feng Z, Tan Q, Min X, Xu S. Development of a Deep Learning System to Detect Esophageal Cancer by Barium Esophagram. Front Oncol 2022; 12:766243. [PMID: 35800062 PMCID: PMC9253273 DOI: 10.3389/fonc.2022.766243] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 05/23/2022] [Indexed: 12/24/2022] Open
Abstract
Background Implementation of deep learning systems (DLSs) for analysis of barium esophagram, a cost-effective diagnostic test for esophageal cancer detection, is expected to reduce the burden to radiologists while ensuring the accuracy of diagnosis. Objective To develop an automated DLS to detect esophageal cancer on barium esophagram. Methods This was a retrospective study using deep learning for esophageal cancer detection. A two-stage DLS (including a Selection network and a Classification network) was developed. Five datasets based on barium esophagram were used for stepwise training, validation, and testing of the DLS. Datasets 1 and 2 were used to respectively train and test the Selection network, while Datasets 3, 4, and 5 were respectively used to train, validate, and test the Classification network. Finally, a positioning box with a probability value was outputted by the DLS. A region of interest delineated by experienced radiologists was selected as the ground truth to evaluate the detection and classification efficiency of the DLS. Standard machine learning metrics (accuracy, recall, precision, sensitivity, and specificity) were calculated. A comparison with the conventional visual inspection approach was also conducted. Results The accuracy, sensitivity, and specificity of our DLS in detecting esophageal cancer were 90.3%, 92.5%, and 88.7%, respectively. With the aid of DLS, the radiologists’ interpretation time was significantly shortened (Reader1, 45.7 s vs. 72.2 s without DLS aid; Reader2, 54.1 s vs. 108.7 s without DLS aid). Respective diagnostic efficiencies for Reader1 with and without DLS aid were 96.8% vs. 89.3% for accuracy, 97.5% vs. 87.5% for sensitivity, 96.2% vs. 90.6% for specificity, and 0.969 vs. 0.890 for AUC. Respective diagnostic efficiencies for Reader2 with and without DLS aid were 95.7% vs. 88.2% for accuracy, 92.5% vs. 77.5% for sensitivity, 98.1% vs. 96.2% for specificity, and 0.953 vs. 0.869 for AUC. Of note, the positioning boxes outputted by the DLS almost overlapped with those manually labeled by the radiologists on Dataset 5. Conclusions The proposed two-stage DLS for detecting esophageal cancer on barium esophagram could effectively shorten the interpretation time with an excellent diagnostic performance. It may well assist radiologists in clinical practice to reduce their burden.
Collapse
Affiliation(s)
- Peipei Zhang
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yifei She
- College of Computer Science, South-Central University for Nationalities, Wuhan, China
| | - Junfeng Gao
- College of Biomedical Engineering, South-Central of University for Nationalities, Wuhan, China
| | - Zhaoyan Feng
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qinghai Tan
- Department of Gastroenterology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiangde Min
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- *Correspondence: Shengzhou Xu, ; Xiangde Min,
| | - Shengzhou Xu
- College of Computer Science, South-Central University for Nationalities, Wuhan, China
- *Correspondence: Shengzhou Xu, ; Xiangde Min,
| |
Collapse
|
20
|
Cho BJ, Kim JW, Park J, Kwon GY, Hong M, Jang SH, Bang H, Kim G, Park ST. Automated Diagnosis of Cervical Intraepithelial Neoplasia in Histology Images via Deep Learning. Diagnostics (Basel) 2022; 12:diagnostics12020548. [PMID: 35204638 PMCID: PMC8871214 DOI: 10.3390/diagnostics12020548] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 02/05/2022] [Accepted: 02/17/2022] [Indexed: 02/04/2023] Open
Abstract
Artificial intelligence has enabled the automated diagnosis of several cancer types. We aimed to develop and validate deep learning models that automatically classify cervical intraepithelial neoplasia (CIN) based on histological images. Microscopic images of CIN3, CIN2, CIN1, and non-neoplasm were obtained. The performances of two pre-trained convolutional neural network (CNN) models adopting DenseNet-161 and EfficientNet-B7 architectures were evaluated and compared with those of pathologists. The dataset comprised 1106 images from 588 patients; images of 10% of patients were included in the test dataset. The mean accuracies for the four-class classification were 88.5% (95% confidence interval [CI], 86.3–90.6%) by DenseNet-161 and 89.5% (95% CI, 83.3–95.7%) by EfficientNet-B7, which were similar to human performance (93.2% and 89.7%). The mean per-class area under the receiver operating characteristic curve values by EfficientNet-B7 were 0.996, 0.990, 0.971, and 0.956 in the non-neoplasm, CIN3, CIN1, and CIN2 groups, respectively. The class activation map detected the diagnostic area for CIN lesions. In the three-class classification of CIN2 and CIN3 as one group, the mean accuracies of DenseNet-161 and EfficientNet-B7 increased to 91.4% (95% CI, 88.8–94.0%), and 92.6% (95% CI, 90.4–94.9%), respectively. CNN-based deep learning is a promising tool for diagnosing CIN lesions on digital histological images.
Collapse
Affiliation(s)
- Bum-Joo Cho
- Medical Artificial Intelligence Center, Hallym University Medical Center, Anyang 14068, Korea;
- Department of Ophthalmology, Hallym University Sacred Heart Hospital, Hallym University College of Medicine, Anyang 14068, Korea
- Correspondence: (B.-J.C.); (J.-W.K.)
| | - Jeong-Won Kim
- Department of Pathology, Kangnam Sacred Heart Hospital, Hallym University College of Medicine, Seoul 07441, Korea;
- Correspondence: (B.-J.C.); (J.-W.K.)
| | - Jungkap Park
- Medical Artificial Intelligence Center, Hallym University Medical Center, Anyang 14068, Korea;
| | | | - Mineui Hong
- Department of Pathology, Chung-Ang University Hospital, Chung-Ang University College of Medicine, Seoul 06973, Korea;
| | - Si-Hyong Jang
- Department of Pathology, Soonchunhyang University Cheonan Hospital, Soonchunhyang University College of Medicine, Cheonan 31151, Korea;
| | - Heejin Bang
- Department of Pathology, Konkuk University Medical Center, Konkuk University School of Medicine, Seoul 05030, Korea;
| | - Gilhyang Kim
- Department of Pathology, Kangnam Sacred Heart Hospital, Hallym University College of Medicine, Seoul 07441, Korea;
| | - Sung-Taek Park
- Department of Obstetrics and Gynecology, Kangnam Sacred Heart Hospital, Hallym University College of Medicine, Seoul 07441, Korea;
| |
Collapse
|
21
|
Ayyaz MS, Lali MIU, Hussain M, Rauf HT, Alouffi B, Alyami H, Wasti S. Hybrid Deep Learning Model for Endoscopic Lesion Detection and Classification Using Endoscopy Videos. Diagnostics (Basel) 2021; 12:diagnostics12010043. [PMID: 35054210 PMCID: PMC8775223 DOI: 10.3390/diagnostics12010043] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2021] [Revised: 12/22/2021] [Accepted: 12/23/2021] [Indexed: 02/06/2023] Open
Abstract
In medical imaging, the detection and classification of stomach diseases are challenging due to the resemblance of different symptoms, image contrast, and complex background. Computer-aided diagnosis (CAD) plays a vital role in the medical imaging field, allowing accurate results to be obtained in minimal time. This article proposes a new hybrid method to detect and classify stomach diseases using endoscopy videos. The proposed methodology comprises seven significant steps: data acquisition, preprocessing of data, transfer learning of deep models, feature extraction, feature selection, hybridization, and classification. We selected two different CNN models (VGG19 and Alexnet) to extract features. We applied transfer learning techniques before using them as feature extractors. We used a genetic algorithm (GA) in feature selection, due to its adaptive nature. We fused selected features of both models using a serial-based approach. Finally, the best features were provided to multiple machine learning classifiers for detection and classification. The proposed approach was evaluated on a personally collected dataset of five classes, including gastritis, ulcer, esophagitis, bleeding, and healthy. We observed that the proposed technique performed superbly on Cubic SVM with 99.8% accuracy. For the authenticity of the proposed technique, we considered these statistical measures: classification accuracy, recall, precision, False Negative Rate (FNR), Area Under the Curve (AUC), and time. In addition, we provided a fair state-of-the-art comparison of our proposed technique with existing techniques that proves its worthiness.
Collapse
Affiliation(s)
- M Shahbaz Ayyaz
- Department of Computer Science, University of Gujrat, Gujrat 50700, Pakistan; (M.S.A.); (M.H.)
| | - Muhammad Ikram Ullah Lali
- Department of Information Sciences, University of Education Lahore, Lahore 41000, Pakistan; (M.I.U.L.); (S.W.)
| | - Mubbashar Hussain
- Department of Computer Science, University of Gujrat, Gujrat 50700, Pakistan; (M.S.A.); (M.H.)
| | - Hafiz Tayyab Rauf
- Centre for Smart Systems, AI and Cybersecurity, Staffordshire University, Stoke-on-Trent ST4 2DE, UK
- Correspondence:
| | - Bader Alouffi
- Department of Computer Science, College of Computers and Information Technology, Taif University, P. O. Box 11099, Taif 21944, Saudi Arabia; (B.A.); (H.A.)
| | - Hashem Alyami
- Department of Computer Science, College of Computers and Information Technology, Taif University, P. O. Box 11099, Taif 21944, Saudi Arabia; (B.A.); (H.A.)
| | - Shahbaz Wasti
- Department of Information Sciences, University of Education Lahore, Lahore 41000, Pakistan; (M.I.U.L.); (S.W.)
| |
Collapse
|
22
|
Zhuang H, Zhang J, Liao F. A systematic review on application of deep learning in digestive system image processing. THE VISUAL COMPUTER 2021; 39:2207-2222. [PMID: 34744231 PMCID: PMC8557108 DOI: 10.1007/s00371-021-02322-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 09/30/2021] [Indexed: 05/07/2023]
Abstract
With the advent of the big data era, the application of artificial intelligence represented by deep learning in medicine has become a hot topic. In gastroenterology, deep learning has accomplished remarkable accomplishments in endoscopy, imageology, and pathology. Artificial intelligence has been applied to benign gastrointestinal tract lesions, early cancer, tumors, inflammatory bowel diseases, livers, pancreas, and other diseases. Computer-aided diagnosis significantly improve diagnostic accuracy and reduce physicians' workload and provide a shred of evidence for clinical diagnosis and treatment. In the near future, artificial intelligence will have high application value in the field of medicine. This paper mainly summarizes the latest research on artificial intelligence in diagnosing and treating digestive system diseases and discussing artificial intelligence's future in digestive system diseases. We sincerely hope that our work can become a stepping stone for gastroenterologists and computer experts in artificial intelligence research and facilitate the application and development of computer-aided image processing technology in gastroenterology.
Collapse
Affiliation(s)
- Huangming Zhuang
- Gastroenterology Department, Renmin Hospital of Wuhan University, Wuhan, 430060 Hubei China
| | - Jixiang Zhang
- Gastroenterology Department, Renmin Hospital of Wuhan University, Wuhan, 430060 Hubei China
| | - Fei Liao
- Gastroenterology Department, Renmin Hospital of Wuhan University, Wuhan, 430060 Hubei China
| |
Collapse
|
23
|
Klein S, Duda DG. Machine Learning for Future Subtyping of the Tumor Microenvironment of Gastro-Esophageal Adenocarcinomas. Cancers (Basel) 2021; 13:4919. [PMID: 34638408 PMCID: PMC8507866 DOI: 10.3390/cancers13194919] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2021] [Revised: 09/27/2021] [Accepted: 09/28/2021] [Indexed: 12/11/2022] Open
Abstract
Tumor progression involves an intricate interplay between malignant cells and their surrounding tumor microenvironment (TME) at specific sites. The TME is dynamic and is composed of stromal, parenchymal, and immune cells, which mediate cancer progression and therapy resistance. Evidence from preclinical and clinical studies revealed that TME targeting and reprogramming can be a promising approach to achieve anti-tumor effects in several cancers, including in GEA. Thus, it is of great interest to use modern technology to understand the relevant components of programming the TME. Here, we discuss the approach of machine learning, which recently gained increasing interest recently because of its ability to measure tumor parameters at the cellular level, reveal global features of relevance, and generate prognostic models. In this review, we discuss the relevant stromal composition of the TME in GEAs and discuss how they could be integrated. We also review the current progress in the application of machine learning in different medical disciplines that are relevant for the management and study of GEA.
Collapse
Affiliation(s)
- Sebastian Klein
- Gerhard-Domagk-Institute for Pathology, University Hospital Münster, 48149 Münster, Germany
- Institute for Pathology, Faculty of Medicine, University Hospital Cologne, University of Cologne, 50931 Cologne, Germany
| | - Dan G. Duda
- Edwin L. Steele Laboratories for Tumor Biology, Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02478, USA
| |
Collapse
|
24
|
Park J, Chung YR, Kong ST, Kim YW, Park H, Kim K, Kim DI, Jung KH. Aggregation of cohorts for histopathological diagnosis with deep morphological analysis. Sci Rep 2021; 11:2876. [PMID: 33536550 PMCID: PMC7858624 DOI: 10.1038/s41598-021-82642-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Accepted: 01/21/2021] [Indexed: 12/13/2022] Open
Abstract
There have been substantial efforts in using deep learning (DL) to diagnose cancer from digital images of pathology slides. Existing algorithms typically operate by training deep neural networks either specialized in specific cohorts or an aggregate of all cohorts when there are only a few images available for the target cohort. A trade-off between decreasing the number of models and their cancer detection performance was evident in our experiments with The Cancer Genomic Atlas dataset, with the former approach achieving higher performance at the cost of having to acquire large datasets from the cohort of interest. Constructing annotated datasets for individual cohorts is extremely time-consuming, with the acquisition cost of such datasets growing linearly with the number of cohorts. Another issue associated with developing cohort-specific models is the difficulty of maintenance: all cohort-specific models may need to be adjusted when a new DL algorithm is to be used, where training even a single model may require a non-negligible amount of computation, or when more data is added to some cohorts. In resolving the sub-optimal behavior of a universal cancer detection model trained on an aggregate of cohorts, we investigated how cohorts can be grouped to augment a dataset without increasing the number of models linearly with the number of cohorts. This study introduces several metrics which measure the morphological similarities between cohort pairs and demonstrates how the metrics can be used to control the trade-off between performance and the number of models.
Collapse
Affiliation(s)
| | - Yul Ri Chung
- Pathology Center, Seegene Medical Foundation, Seoul, Korea
| | | | | | | | | | - Dong-Il Kim
- Department of Pathology, Green Cross Laboratories, Yong-in, Gyeonggi, Korea
| | | |
Collapse
|