1
|
Dinis-Ribeiro M, Libânio D, Uchima H, Spaander MCW, Bornschein J, Matysiak-Budnik T, Tziatzios G, Santos-Antunes J, Areia M, Chapelle N, Esposito G, Fernandez-Esparrach G, Kunovsky L, Garrido M, Tacheci I, Link A, Marcos P, Marcos-Pinto R, Moreira L, Pereira AC, Pimentel-Nunes P, Romanczyk M, Fontes F, Hassan C, Bisschops R, Feakins R, Schulz C, Triantafyllou K, Carneiro F, Kuipers EJ. Management of epithelial precancerous conditions and early neoplasia of the stomach (MAPS III): European Society of Gastrointestinal Endoscopy (ESGE), European Helicobacter and Microbiota Study Group (EHMSG) and European Society of Pathology (ESP) Guideline update 2025. Endoscopy 2025; 57:504-554. [PMID: 40112834 DOI: 10.1055/a-2529-5025] [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] [Indexed: 03/22/2025]
Abstract
At a population level, the European Society of Gastrointestinal Endoscopy (ESGE), the European Helicobacter and Microbiota Study Group (EHMSG), and the European Society of Pathology (ESP) suggest endoscopic screening for gastric cancer (and precancerous conditions) in high-risk regions (age-standardized rate [ASR] > 20 per 100 000 person-years) every 2 to 3 years or, if cost-effectiveness has been proven, in intermediate risk regions (ASR 10-20 per 100 000 person-years) every 5 years, but not in low-risk regions (ASR < 10).ESGE/EHMSG/ESP recommend that irrespective of country of origin, individual gastric risk assessment and stratification of precancerous conditions is recommended for first-time gastroscopy. ESGE/EHMSG/ESP suggest that gastric cancer screening or surveillance in asymptomatic individuals over 80 should be discontinued or not started, and that patients' comorbidities should be considered when treatment of superficial lesions is planned.ESGE/EHMSG/ESP recommend that a high quality endoscopy including the use of virtual chromoendoscopy (VCE), after proper training, is performed for screening, diagnosis, and staging of precancerous conditions (atrophy and intestinal metaplasia) and lesions (dysplasia or cancer), as well as after endoscopic therapy. VCE should be used to guide the sampling site for biopsies in the case of suspected neoplastic lesions as well as to guide biopsies for diagnosis and staging of gastric precancerous conditions, with random biopsies to be taken in the absence of endoscopically suspected changes. When there is a suspected early gastric neoplastic lesion, it should be properly described (location, size, Paris classification, vascular and mucosal pattern), photodocumented, and two targeted biopsies taken.ESGE/EHMSG/ESP do not recommend routine performance of endoscopic ultrasonography (EUS), computed tomography (CT), magnetic resonance imaging (MRI), or positron emission tomography (PET)-CT prior to endoscopic resection unless there are signs of deep submucosal invasion or if the lesion is not considered suitable for endoscopic resection.ESGE/EHMSG/ESP recommend endoscopic submucosal dissection (ESD) for differentiated gastric lesions clinically staged as dysplastic (low grade and high grade) or as intramucosal carcinoma (of any size if not ulcerated or ≤ 30 mm if ulcerated), with EMR being an alternative for Paris 0-IIa lesions of size ≤ 10 mm with low likelihood of malignancy.ESGE/EHMSG/ESP suggest that a decision about ESD can be considered for malignant lesions clinically staged as having minimal submucosal invasion if differentiated and ≤ 30 mm; or for malignant lesions clinically staged as intramucosal, undifferentiated and ≤ 20 mm; and in both cases with no ulcerative findings.ESGE/EHMSG/ESP recommends patient management based on the following histological risk after endoscopic resection: Curative/very low-risk resection (lymph node metastasis [LNM] risk < 0.5 %-1 %): en bloc R0 resection; dysplastic/pT1a, differentiated lesion, no lymphovascular invasion, independent of size if no ulceration and ≤ 30 mm if ulcerated. No further staging procedure or treatment is recommended.Curative/low-risk resection (LNM risk < 3 %): en bloc R0 resection; lesion with no lymphovascular invasion and: a) pT1b, invasion ≤ 500 µm, differentiated, size ≤ 30 mm; or b) pT1a, undifferentiated, size ≤ 20 mm and no ulceration. Staging should be completed, and further treatment is generally not necessary, but a multidisciplinary discussion is required. Local-risk resection (very low risk of LNM but increased risk of local persistence/recurrence): Piecemeal resection or tumor-positive horizontal margin of a lesion otherwise meeting curative/very low-risk criteria (or meeting low-risk criteria provided that there is no submucosal invasive tumor at the resection margin in the case of piecemeal resection or tumor-positive horizontal margin for pT1b lesions [invasion ≤ 500 µm; well-differentiated; size ≤ 30 mm, and VM0]). Endoscopic surveillance/re-treatment is recommended rather than other additional treatment. High-risk resection (noncurative): Any lesion with any of the following: (a) a positive vertical margin (if carcinoma) or lymphovascular invasion or deep submucosal invasion (> 500 µm from the muscularis mucosae); (b) poorly differentiated lesions if ulceration or size > 20 mm; (c) pT1b differentiated lesions with submucosal invasion ≤ 500 µm with size > 30 mm; or (d) intramucosal ulcerative lesion with size > 30 mm. Complete staging and strong consideration for additional treatments (surgery) in multidisciplinary discussion.ESGE/EHMSG/ESP suggest the use of validated endoscopic classifications of atrophy (e. g. Kimura-Takemoto) or intestinal metaplasia (e. g. endoscopic grading of gastric intestinal metaplasia [EGGIM]) to endoscopically stage precancerous conditions and stratify the risk for gastric cancer.ESGE/EHMSG/ESP recommend that biopsies should be taken from at least two topographic sites (2 biopsies from the antrum/incisura and 2 from the corpus, guided by VCE) in two separate, clearly labeled vials. Additional biopsy from the incisura is optional.ESGE/EHMSG/ESP recommend that patients with extensive endoscopic changes (Kimura C3 + or EGGIM 5 +) or advanced histological stages of atrophic gastritis (severe atrophic changes or intestinal metaplasia, or changes in both antrum and corpus, operative link on gastritis assessment/operative link on gastric intestinal metaplasia [OLGA/OLGIM] III/IV) should be followed up with high quality endoscopy every 3 years, irrespective of the individual's country of origin.ESGE/EHMSG/ESP recommend that no surveillance is proposed for patients with mild to moderate atrophy or intestinal metaplasia restricted to the antrum, in the absence of endoscopic signs of extensive lesions or other risk factors (family history, incomplete intestinal metaplasia, persistent H. pylori infection). This group constitutes most individuals found in clinical practice.ESGE/EHMSG/ESP recommend H. pylori eradication for patients with precancerous conditions and after endoscopic or surgical therapy.ESGE/EHMSG/ESP recommend that patients should be advised to stop smoking and low-dose daily aspirin use may be considered for the prevention of gastric cancer in selected individuals with high risk for cardiovascular events.
Collapse
Affiliation(s)
- Mário Dinis-Ribeiro
- Precancerous Lesions and Early Cancer Management Group, Research Center of IPO Porto (CI-IPOP)/CI-IPOP@RISE (Health Research Group), Portuguese Institute of Oncology of Porto (IPO Porto)/Porto Comprehensive Cancer Center (Porto.CCC), Porto, Portugal
- Gastroenterology Department, Portuguese Institute of Oncology of Porto, Porto, Portugal
| | - Diogo Libânio
- Precancerous Lesions and Early Cancer Management Group, Research Center of IPO Porto (CI-IPOP)/CI-IPOP@RISE (Health Research Group), Portuguese Institute of Oncology of Porto (IPO Porto)/Porto Comprehensive Cancer Center (Porto.CCC), Porto, Portugal
- Gastroenterology Department, Portuguese Institute of Oncology of Porto, Porto, Portugal
| | - Hugo Uchima
- Endoscopy Unit Gastroenterology Department Hospital Universitari Germans Trias i Pujol, Badalona, Spain
- Endoscopy Unit, Teknon Medical Center, Barcelona, Spain
| | - Manon C W Spaander
- Department of Gastroenterology and Hepatology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Jan Bornschein
- Medical Research Council Translational Immune Discovery Unit (MRC TIDU), Weatherall Institute of Molecular Medicine (WIMM), Radcliffe Department of Medicine, University of Oxford, Oxford, UK
- Translational Gastroenterology and Liver Unit, Nuffield Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford, UK
| | - Tamara Matysiak-Budnik
- Department of Hepato-Gastroenterology & Digestive Oncology, Institut des Maladies de l'Appareil Digestif, Centre Hospitalier Universitaire de Nantes Nantes, France
- INSERM, Center for Research in Transplantation and Translational Immunology, University of Nantes, Nantes, France
| | - Georgios Tziatzios
- Agia Olga General Hospital of Nea Ionia Konstantopouleio, Athens, Greece
| | - João Santos-Antunes
- Gastroenterology Department, Centro Hospitalar S. João, Porto, Portugal
- Faculty of Medicine, University of Porto, Portugal
- University of Porto, Institute of Molecular Pathology and Immunology of the University of Porto (IPATIMUP), Instituto de Investigação e Inovação na Saúde (I3S), Porto, Portugal
| | - Miguel Areia
- Gastroenterology Department, Portuguese Oncology Institute of Coimbra (IPO Coimbra), Coimbra, Portugal
- Precancerous Lesions and Early Cancer Management Group, Research Center of IPO Porto (CI-IPOP)/CI-IPOP@RISE (Health Research Group), RISE@CI-IPO, (Health Research Network), Portuguese Institute of Oncology of Porto (IPO Porto), Porto, Portugal
| | - Nicolas Chapelle
- Department of Hepato-Gastroenterology & Digestive Oncology, Institut des Maladies de l'Appareil Digestif, Centre Hospitalier Universitaire de Nantes Nantes, France
- INSERM, Center for Research in Transplantation and Translational Immunology, University of Nantes, Nantes, France
| | - Gianluca Esposito
- Department of Medical-Surgical Sciences and Translational Medicine, Sant'Andrea Hospital, Sapienza University of Rome, Italy
| | - Gloria Fernandez-Esparrach
- Gastroenterology Department, ICMDM, Hospital Clínic, Universitat de Barcelona, Barcelona, Spain
- Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, Barcelona, Spain
- Instituto de Investigaciones Biomédicas August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Spain
| | - Lumir Kunovsky
- 2nd Department of Internal Medicine - Gastroenterology and Geriatrics, University Hospital Olomouc, Faculty of Medicine and Dentistry, Palacky University Olomouc, Olomouc, Czech Republic
- Department of Surgery, University Hospital Brno, Faculty of Medicine, Masaryk University, Brno, Czech Republic
- Department of Gastroenterology and Digestive Endoscopy, Masaryk Memorial Cancer Institute, Brno, Czech Republic
| | - Mónica Garrido
- Gastroenterology Department, Portuguese Institute of Oncology of Porto, Porto, Portugal
| | - Ilja Tacheci
- Gastroenterology, Second Department of Internal Medicine, University Hospital Hradec Kralove, Faculty of Medicine in Hradec Kralove, Charles University of Prague, Czech Republic
| | | | - Pedro Marcos
- Department of Gastroenterology, Pêro da Covilhã Hospital, Covilhã, Portugal
- Department of Medical Sciences, Faculty of Health Sciences, University of Beira Interior, Covilhã, Portugal
| | - Ricardo Marcos-Pinto
- Precancerous Lesions and Early Cancer Management Group, Research Center of IPO Porto (CI-IPOP)/CI-IPOP@RISE (Health Research Group), RISE@CI-IPO, (Health Research Network), Portuguese Institute of Oncology of Porto (IPO Porto), Porto, Portugal
- Gastroenterology Department, Centro Hospitalar do Porto, Porto, Portugal
- Institute of Biomedical Sciences Abel Salazar, University of Porto, Porto, Portugal
| | - Leticia Moreira
- Gastroenterology Department, ICMDM, Hospital Clínic, Universitat de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Spain
| | - Ana Carina Pereira
- Precancerous Lesions and Early Cancer Management Group, Research Center of IPO Porto (CI-IPOP)/CI-IPOP@RISE (Health Research Group), Portuguese Institute of Oncology of Porto (IPO Porto)/Porto Comprehensive Cancer Center (Porto.CCC), Porto, Portugal
| | - Pedro Pimentel-Nunes
- Precancerous Lesions and Early Cancer Management Group, Research Center of IPO Porto (CI-IPOP)/CI-IPOP@RISE (Health Research Group), RISE@CI-IPO, (Health Research Network), Portuguese Institute of Oncology of Porto (IPO Porto), Porto, Portugal
- Department of Surgery and Physiology, Faculty of Medicine, University of Porto (FMUP), Portugal
- Gastroenterology and Clinical Research, Unilabs Portugal
| | - Marcin Romanczyk
- Department of Gastroenterology, Faculty of Medicine, Academy of Silesia, Katowice, Poland
- Endoterapia, H-T. Centrum Medyczne, Tychy, Poland
| | - Filipa Fontes
- Precancerous Lesions and Early Cancer Management Group, Research Center of IPO Porto (CI-IPOP)/CI-IPOP@RISE (Health Research Group), Portuguese Institute of Oncology of Porto (IPO Porto)/Porto Comprehensive Cancer Center (Porto.CCC), Porto, Portugal
- Public Health and Forensic Sciences, and Medical Education Department, Faculty of Medicine, University of Porto, Porto, Portugal
| | - Cesare Hassan
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy
- IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Raf Bisschops
- Department of Gastroenterology and Hepatology, UZ Leuven, Leuven, Belgium
- Department of Translational Research in Gastrointestinal Diseases (TARGID), KU Leuven, Leuven, Belgium
| | - Roger Feakins
- Department of Cellular Pathology, Royal Free London NHS Foundation Trust, London, United Kingdom
- University College London, London, United Kingdom
| | - Christian Schulz
- Department of Medicine II, University Hospital, LMU Munich, Germany
| | - Konstantinos Triantafyllou
- Hepatogastroenterology Unit, Second Department of Internal Medicine-Propaedeutic, Medical School, National and Kapodistrian University of Athens, Attikon University General Hospital, Athens, Greece
| | - Fatima Carneiro
- Institute of Molecular Pathology and Immunology at the University of Porto (IPATIMUP), Porto, Portugal
- Instituto de Investigação e Inovação em Saúde (i3S), University of Porto, Porto, Portugal
- Pathology Department, Centro Hospitalar de São João and Faculty of Medicine, Porto, Portugal
| | - Ernst J Kuipers
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore
| |
Collapse
|
2
|
Zheng J, Jiao Z, Yang X, Ruan Q, Huang Y, Jin C, Gui S, Xuan Z, Jia X. Network pharmacology-based exploration of the mechanism of Wenweishu granule in treating chronic atrophic gastritis with spleen-stomach cold deficiency syndrome. JOURNAL OF ETHNOPHARMACOLOGY 2025; 345:119591. [PMID: 40054637 DOI: 10.1016/j.jep.2025.119591] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Revised: 02/14/2025] [Accepted: 03/05/2025] [Indexed: 03/17/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Wenweishu (WWS) is a traditional Chinese medicine compound formulated for chronic atrophic gastritis (CAG) treatment by warming the stomach and alleviating pain. However, its pharmacological mechanisms remain underexplored. AIM OF THE STUDY This study investigated the therapeutic effects and potential mechanisms of WWS on CAG with spleen-stomach cold deficiency syndrome (SSCDS). METHODS To achieve this, an SSCDS-CAG rat model and a human gastric mucosal epithelial cells (GES-1) cell model were established using multi-factor modeling and N-Methyl-N'-nitro-N-nitrosoguanidine (MNNG) induction, respectively. WWS's effects on gastric injury were evaluated through pathology, inflammation, serum biomarkers, and apoptosis. Additionally, MNNG's effects on GES-1 cells were analyzed. Network pharmacology, involving protein-protein interaction networks, GO/KEGG enrichment, and molecular docking, was employed to predict WWS's potential targets and mechanisms in SSCDS-CAG. Mechanistic insights were further validated using immunohistochemistry, quantitative reverse transcription polymerase chain reaction, and western blotting. RESULTS In vivo results showed that WWS alleviated symptoms in SSCDS-CAG rats, lowering symptom scores and improving gastric histopathology. It modulated serum biomarkers and reduced inflammation and apoptosis in both in vivo and in vitro studies. Network pharmacology results revealed 263 overlapping targets between WWS and SSCDS-CAG, associated with apoptosis, inflammation, and the PI3K/AKT pathway. Molecular docking revealed strong binding affinity between the core target and active WWS components. In SSCDS-CAG rats and GES-1 cells, WWS inhibited PI3K/AKT phosphorylation, increased PTEN expression, and regulated Bcl-2, Bax, and cleaved caspase-3 levels. CONCLUSION WWS reduces inflammation and apoptosis in multi-factor CAG rats and MNNG-induced GES-1 cells by modulating the PTEN/PI3K/AKT signaling pathway and apoptosis-related proteins.
Collapse
Affiliation(s)
- Jia Zheng
- College of Pharmacy, Anhui University of Chinese Medicine, Hefei, 230012, China; Anhui Province Key Laboratory of Bioactive Natural Products, Hefei, 230012, China
| | - Zhiyong Jiao
- College of Pharmacy, Anhui University of Chinese Medicine, Hefei, 230012, China; Anhui Province Key Laboratory of Bioactive Natural Products, Hefei, 230012, China
| | - Xinyu Yang
- College of Pharmacy, Anhui University of Chinese Medicine, Hefei, 230012, China; Anhui Province Key Laboratory of Bioactive Natural Products, Hefei, 230012, China
| | - Qing Ruan
- College of Pharmacy, Anhui University of Chinese Medicine, Hefei, 230012, China; Anhui Province Key Laboratory of Bioactive Natural Products, Hefei, 230012, China
| | - Yuzhe Huang
- College of Pharmacy, Anhui University of Chinese Medicine, Hefei, 230012, China; Institute of Pharmaceutics, Anhui Academy of Chinese Medicine, Hefei, Anhui, 230012, China
| | - Cheng Jin
- College of Pharmacy, Anhui University of Chinese Medicine, Hefei, 230012, China; Institute of Pharmaceutics, Anhui Academy of Chinese Medicine, Hefei, Anhui, 230012, China
| | - Shuangying Gui
- College of Pharmacy, Anhui University of Chinese Medicine, Hefei, 230012, China; Institute of Pharmaceutics, Anhui Academy of Chinese Medicine, Hefei, Anhui, 230012, China
| | - Zihua Xuan
- College of Pharmacy, Anhui University of Chinese Medicine, Hefei, 230012, China; Anhui Province Key Laboratory of Bioactive Natural Products, Hefei, 230012, China
| | - Xiaoyi Jia
- College of Pharmacy, Anhui University of Chinese Medicine, Hefei, 230012, China; Anhui Province Key Laboratory of Bioactive Natural Products, Hefei, 230012, China.
| |
Collapse
|
3
|
Tahmasebzadeh A, Sadeghi M, Naseripour M, Mirshahi R, Ghaderi R. Artificial intelligence and different image modalities in uveal melanoma diagnosis and prognosis: A narrative review. Photodiagnosis Photodyn Ther 2025; 52:104528. [PMID: 39986588 DOI: 10.1016/j.pdpdt.2025.104528] [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: 01/18/2025] [Revised: 02/06/2025] [Accepted: 02/19/2025] [Indexed: 02/24/2025]
Abstract
BACKGROUND The most widespread primary intraocular tumor in adults is called uveal melanoma (UM), if detected early enough, it can be curable. Various methods are available to treat UM, but the most commonly used and effective approach is plaque radiotherapy using Iodine-125 and Ruthenium-106. METHOD The authors performed searches to distinguish relevant studies from 2017 to 2024 by three databases (PubMed, Scopus, and Google Scholar). RESULTS Imaging technologies such as ultrasound (US), fundus photography (FP), optical coherent tomography (OCT), fluorescein angiography (FA), and magnetic resonance images (MRI) play a vital role in the diagnosis and prognosis of UM. The present review assessed the power of different image modalities when integrated with artificial intelligence (AI) to diagnose and prognosis of patients affected by UM. CONCLUSION Finally, after reviewing the studies conducted, it was concluded that AI is a developing tool in image analysis and enhances workflows in diagnosis from data and image processing to clinical decisions, improving tailored treatment scenarios, response prediction, and prognostication.
Collapse
Affiliation(s)
- Atefeh Tahmasebzadeh
- Medical Physics Department, School of Medicine, Iran University of Medical Sciences, Tehran, , Iran
| | - Mahdi Sadeghi
- Medical Physics Department, School of Medicine, Iran University of Medical Sciences, Tehran, , Iran.
| | - Masood Naseripour
- Eye Research Center, The Five Senses Institute, Moheb Kowsar Hospital, Iran University of Medical Sciences, Tehran, Iran; Finetech in Medicine Research Center, Iran University of Medical Sciences, Tehran, Iran.
| | - Reza Mirshahi
- Eye Research Center, The Five Senses Institute, Moheb Kowsar Hospital, Iran University of Medical Sciences, Tehran, Iran
| | - Reza Ghaderi
- Department of Electrical Engineering, Shahid Beheshti University, Tehran, Iran
| |
Collapse
|
4
|
Wan N, Chan C, Tan JL, Chinnaratha MA, Singh R. Endoscopists' knowledge, perceptions, and attitudes toward the use of artificial intelligence in endoscopy: A systematic review. Gastrointest Endosc 2025:S0016-5107(25)00148-8. [PMID: 40081742 DOI: 10.1016/j.gie.2025.03.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Revised: 02/20/2025] [Accepted: 03/03/2025] [Indexed: 03/16/2025]
Abstract
BACKGROUND AND AIMS Artificial intelligence (AI) is rapidly evolving in the field of GI endoscopy. This systematic review aims to summarize the current perspectives of endoscopists on AI in endoscopy and to identify its challenges. METHODS Electronic databases were searched to identify studies conducted on endoscopists' opinions on the use of AI in endoscopy. A qualitative synthesis of included studies was conducted by categorizing into 5 domains: knowledge, perception, and attitude toward AI; impacts of AI on endoscopic performance; impact of AI on endoscopists; impact of AI on patients; and barriers toward the implementation of AI. RESULTS Ten studies were included, comprising 1587 endoscopists across Europe (32.6%), North America (42.8%), and Asia (24.6%). For domain 1, most endoscopists (69%-100%) had a basic knowledge of AI and most (79.5%-87.5%) expressed interest and optimism. For domain 2, most endoscopists (62.5%-97%) believed AI would positively impact endoscopic performance and quality. Domain 3 saw mixed perceptions, with 6.2% to 62.8% of endoscopists suggesting AI would lead to operator dependence, 21% to 81.3% believing AI would prolong procedural time, and most (71%-100%) disagreeing that AI would replace them. For domain 4, most endoscopists (81.3%) believed AI would improve patient care. For domain 5, most (75.2%-91%) identified costs as barriers to AI implementation. Opinions on ethics and regulation varied (12.5%-100% and 35%-88%, respectively), with most advocating for clear guidelines and regulations. CONCLUSIONS There is a positive attitude toward the use of AI in endoscopy. Concerns regarding the impact on clinical practice, costs, and medicolegal considerations remain. Establishing robust regulatory frameworks is crucial to the integration of AI into endoscopy.
Collapse
Affiliation(s)
- Nicholas Wan
- Department of Gastroenterology, Lyell McEwin Hospital, Adelaide, Australia; Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, Australia
| | - Celine Chan
- Department of Gastroenterology, Lyell McEwin Hospital, Adelaide, Australia
| | - Jin Lin Tan
- Department of Gastroenterology, Lyell McEwin Hospital, Adelaide, Australia; Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, Australia
| | - Mohamed Asif Chinnaratha
- Department of Gastroenterology, Lyell McEwin Hospital, Adelaide, Australia; Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, Australia
| | - Rajvinder Singh
- Department of Gastroenterology, Lyell McEwin Hospital, Adelaide, Australia; Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, Australia.
| |
Collapse
|
5
|
Ebigbo A, Messmann H, Lee SH. Artificial Intelligence Applications in Image-Based Diagnosis of Early Esophageal and Gastric Neoplasms. Gastroenterology 2025:S0016-5085(25)00471-8. [PMID: 40043857 DOI: 10.1053/j.gastro.2025.01.253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Revised: 01/14/2025] [Accepted: 01/22/2025] [Indexed: 04/03/2025]
Abstract
Artificial intelligence (AI) holds the potential to transform the management of upper gastrointestinal (GI) conditions, such as Barrett's esophagus, esophageal squamous cell cancer, and early gastric cancer. Advancements in deep learning and convolutional neural networks offer improved diagnostic accuracy and reduced diagnostic variability across different clinical settings, particularly where human error or fatigue may impair diagnostic precision. Deep learning models have shown the potential to improve early cancer detection and lesion characterization, predict invasion depth, and delineate lesion margins with remarkable accuracy, all contributing to effective treatment planning. Several challenges, however, limit the broad application of AI in GI endoscopy, particularly in the upper GI tract. Subtle lesion morphology and restricted diversity in training datasets, which are often sourced from specialized centers, may constrain the generalizability of AI models in various clinical settings. Furthermore, the "black box" nature of some AI systems can impede explainability and clinician trust. To address these issues, efforts are underway to incorporate multimodal data, such as combining endoscopic and histopathologic imaging, to bolster model robustness and transparency. In the future, AI promises substantial advancements in automated real-time endoscopic guidance, personalized risk assessment, and optimized biopsy decision making. As it evolves, it would substantially impact not only early diagnosis and prognosis, but also the cost-effectiveness of managing upper GI diseases, ultimately leading to improved patient outcomes and more efficient health care delivery.
Collapse
Affiliation(s)
- Alanna Ebigbo
- Department of Gastroenterology, University Hospital Augsburg, Augsburg, Germany.
| | - Helmut Messmann
- Department of Gastroenterology, University Hospital Augsburg, Augsburg, Germany.
| | - Sung Hak Lee
- Department of Hospital Pathology, College of Medicine, The Catholic University of Korea, Seoul, South Korea; Seoul St. Mary's Hospital, Seoul, South Korea.
| |
Collapse
|
6
|
Kafetzis I, Sodmann P, Herghelegiu B, Brand M, Zoller WG, Seyfried F, Fuchs K, Meining A, Hann A. Prospective Evaluation of Real-Time Artificial Intelligence for the Hill Classification of the Gastroesophageal Junction. United European Gastroenterol J 2025; 13:240-246. [PMID: 39668544 PMCID: PMC11975621 DOI: 10.1002/ueg2.12721] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Revised: 09/30/2024] [Accepted: 10/12/2024] [Indexed: 12/14/2024] Open
Abstract
BACKGROUND Assessment of the gastroesophageal junction (GEJ) is an integral part of gastroscopy; however, the absence of standardized reporting hinders consistency of examination documentation. The Hill classification offers a standardized approach for evaluating the GEJ. This study aims to compare the accuracy of an artificial intelligence (AI) system with that of physicians in classifying the GEJ according to Hill in a prospective, blinded, superiority trial. METHODS Consecutive patients scheduled for gastroscopy with an intact GEJ were recruited during clinical routine from October 2023 to December 2023. Nine physicians (six experienced, three inexperienced) assessed the Hill grade, and the AI system operated in the background in real-time. The gold standard was determined by a majority vote of independent assessments by three expert endoscopists who did not participate in the study. The primary outcome was accuracy. Secondary outcomes were per-Hill grade analysis and result comparison for experienced and inexperienced endoscopists separately. RESULTS In 131 analysed examinations the AI's accuracy of 84.7% (95% CI: 78.6-90.8) was significantly higher than 62.5% (95% CI: 54.2-71) of physicians (p < 0.01). The AI outperformed physicians in all but one cases in the per-Hill-class analysis. AI was significantly more accurate than inexperienced physicians (85% vs. 56%, p < 0.01) and in trend better than experienced physicians (84% vs. 69.6%, p = 0.07). CONCLUSIONS AI was significantly more accurate than examiners in assessing the Hill classification. This superior model performance can prove beneficial for endoscopists, especially those with limited experience. TRIAL REGISTRATION ClinicalTrials.gov identifier: NCT06040723.
Collapse
Affiliation(s)
- Ioannis Kafetzis
- Interventional and Experimental Endoscopy (InExEn)Department of Internal Medicine 2University Hospital WürzburgWürzburgGermany
| | - Philipp Sodmann
- Interventional and Experimental Endoscopy (InExEn)Department of Internal Medicine 2University Hospital WürzburgWürzburgGermany
| | - Bianca‐Elena Herghelegiu
- Interventional and Experimental Endoscopy (InExEn)Department of Internal Medicine 2University Hospital WürzburgWürzburgGermany
| | - Markus Brand
- Interventional and Experimental Endoscopy (InExEn)Department of Internal Medicine 2University Hospital WürzburgWürzburgGermany
| | - Wolfram G. Zoller
- Department of Internal Medicine and GastroenterologyKatharinenhospitalStuttgartGermany
| | - Florian Seyfried
- Department of General, Visceral, Transplantation, Vascular, and Pediatric SurgeryCenter of Operative Medicine (ZOM)University Hospital WürzburgWürzburgGermany
| | - Karl‐Hermann Fuchs
- Interventional and Experimental Endoscopy (InExEn)Department of Internal Medicine 2University Hospital WürzburgWürzburgGermany
| | - Alexander Meining
- Interventional and Experimental Endoscopy (InExEn)Department of Internal Medicine 2University Hospital WürzburgWürzburgGermany
| | - Alexander Hann
- Interventional and Experimental Endoscopy (InExEn)Department of Internal Medicine 2University Hospital WürzburgWürzburgGermany
| |
Collapse
|
7
|
Tkach S, Kharchenko N, Dorofieiev A, Hurkalo Y, Ryzhiy L. Current concepts of chronic gastritis and gastropathies: new international consensus RE.GA.IN. Gastroenterology 2024; 58:286-294. [DOI: 10.22141/2308-2097.58.4.2024.641] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
Abstract
Recent decades have been marked by several important milestones in the field of gastritis. New epidemiological reports were received, especially from countries with a high incidence of gastric cancer, and H.pylori infection was recognized as the most important cause of gastritis, due to which it was quickly included in the primary infectious oncogenic agents. Clinical strategies for the detection and eradication of H.pylori infection were developed, the previously unsuspected clinical and pathological significance of gastric microbiota was established, and extraordinary progress was made in the technology of esophagogastroduodenoscopy. Histological determination of gastritis in terms of stage (that is, risk stratification of stomach cancer) was implemented into clinical practice. In addition to the Sydney and Houston gastritis classifications, the six editions of the Maastricht Consensus (1996–2022) and the Kyoto Global Consensus deserve special attention, thanks to which broad agreements and significant scientific achievements became possible in the most controversial issues of the entire spectrum of gastritis, with a special emphasis on H.pylori gastritis, which accounts for more than 90 % of all forms of gastritis worldwide. The Real-world Gastritis Initiative (RE.GA.IN) marks the next step in the ongoing search to achieve a better understanding of various gastric conditions. The RE.GA.IN consensus was concluded in Venice in November 2022 after 8 months of intensive global scientific reasonings. RE.GA.IN mission itself has been focused on critically reviewing, updating, sharing and building consensus on the current scientific knowledge of inflammatory gastric lesions. It includes eight sections on clinicopathological topics, each consisting of preamble, which presents brief statements and corresponding explanatory texts. For each statement, the level of evidence (assessed according to a predefined four-level scale) and the strength of recommendations on the GRADE system are reported. The following topics were considered in the consensus: 1) definition and classification issues of gastritis; 2) spectrum of H.pylori gastritis; 3) key diagnosis of H.pylori gastritis; 4) H.pylori gastritis: clinical results; 5) autoimmune gastritis; 6) gastritis of low prevalence; 7) gastritis and gastric microbiota; 8) epidemiology of gastritis and associated precancerous and neoplastic lesions. Thus, the most controversial aspects of gastritis were addressed, a comprehensive and diverse body of knowledge was brought together to use patient-oriented evidence to assist physicians in their real-world clinical practice.
Collapse
|
8
|
Bai Z, Yin Y, Xu W, Cheng G, Qi X. Predictive model of in-hospital mortality in liver cirrhosis patients with hyponatremia: an artificial neural network approach. Sci Rep 2024; 14:28719. [PMID: 39567595 PMCID: PMC11579295 DOI: 10.1038/s41598-024-73256-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Accepted: 09/16/2024] [Indexed: 11/22/2024] Open
Abstract
Hyponatremia can worsen the outcomes of patients with liver cirrhosis. However, it remains unclear about how to predict the risk of death in cirrhotic patients with hyponatremia. Patients with liver cirrhosis and hyponatremia were screened. Eligible patients were randomly divided into the training (n = 472) and validation (n = 471) cohorts. In the training cohort, the independent predictors for in-hospital death were identified by logistic regression analyses. Odds ratios (ORs) were calculated. An artificial neural network (ANN) model was established in the training cohort. Areas under curve (AUCs) of ANN model, Child-Pugh, model for end-stage liver disease (MELD), and MELD-Na scores were calculated by receiver operating characteristic curve analyses. In multivariate logistic regression analyses, ascites (OR = 2.705, P = 0.042), total bilirubin (OR = 1.004, P = 0.003), serum creatinine (OR = 1.004, P = 0.017), and international normalized ratio (OR = 1.457, P = 0.005) were independently associated with in-hospital death. Based on the four variables, an ANN model was established. Its AUC was 0.865 and 0.810 in the training and validation cohorts, respectively, which was significantly larger than those of Child-Pugh (AUC = 0.757), MELD (AUC = 0.765), and MELD-Na (AUC = 0.769) scores. An ANN model has been developed and validated for the prediction of in-hospital death in patients with liver cirrhosis and hyponatremia.
Collapse
Affiliation(s)
- Zhaohui Bai
- Liver Cirrhosis Study Group, Department of Gastroenterology, General Hospital of Northern Theater Command (Teaching Hospital of Shenyang Pharmaceutical University), Shenyang, China
- Department of Pharmacy Administration and Clinical Pharmacy, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Yuhang Yin
- Liver Cirrhosis Study Group, Department of Gastroenterology, General Hospital of Northern Theater Command (Teaching Hospital of Shenyang Pharmaceutical University), Shenyang, China
- Postgraduate College, China Medical University, Shenyang, China
| | - Wentao Xu
- Liver Cirrhosis Study Group, Department of Gastroenterology, General Hospital of Northern Theater Command (Teaching Hospital of Shenyang Pharmaceutical University), Shenyang, China
| | - Gang Cheng
- NMPA Key Laboratory for Research and Evaluation of Drug Regulatory Technology, Shenyang Pharmaceutical University, Shenyang, China.
| | - Xingshun Qi
- Liver Cirrhosis Study Group, Department of Gastroenterology, General Hospital of Northern Theater Command (Teaching Hospital of Shenyang Pharmaceutical University), Shenyang, China.
- NMPA Key Laboratory for Research and Evaluation of Drug Regulatory Technology, Shenyang Pharmaceutical University, Shenyang, China.
- Department of Gastroenterology, General Hospital of Northern Theater Command Shenyang (Teaching Hospital of Shenyang Pharmaceutical University), Shenyang, China.
| |
Collapse
|
9
|
Ma XZ, Zhou N, Luo X, Guo SQ, Mai P. Update understanding on diagnosis and histopathological examination of atrophic gastritis: A review. World J Gastrointest Oncol 2024; 16:4080-4091. [DOI: 10.4251/wjgo.v16.i10.4080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 08/14/2024] [Accepted: 08/21/2024] [Indexed: 09/26/2024] Open
Abstract
Chronic atrophic gastritis (CAG) is a complex syndrome in which long-term chronic inflammatory stimulation causes gland atrophy in the gastric mucosa, reducing the stomach's ability to secrete gastric juice and pepsin, and interfering with its normal physiological function. Multiple pathogenic factors contribute to CAG incidence, the most common being Helicobacter pylori infection and the immune reactions resulting from gastric autoimmunity. Furthermore, CAG has a broad spectrum of clinical manifestations, including gastroenterology and extra-intestinal symptoms and signs, such as hematology, neurology, and oncology. Therefore, the initial CAG evaluation should involve the examination of clinical and serological indicators, as well as diagnosis confirmation via gastroscopy and histopathology if necessary. Depending on the severity and scope of atrophy affecting the gastric mucosa, a histologic staging system (Operative Link for Gastritis Assessment or Operative Link on Gastritis intestinal metaplasia) could also be employed. Moreover, chronic gastritis has a higher risk of progressing to gastric cancer (GC). In this regard, early diagnosis, treatment, and regular testing could reduce the risk of GC in CAG patients. However, the optimal interval for endoscopic monitoring in CAG patients remains uncertain, and it should ideally be tailored based on individual risk evaluations and shared decision-making processes. Although there have been many reports on CAG, the precise etiology and histopathological features of the disease, as well as the diagnosis of CAG patients, are yet to be fully elucidated. Consequently, this review offers a detailed account of CAG, including its key clinical aspects, aiming to enhance the overall understanding of the disease.
Collapse
Affiliation(s)
- Xiu-Zhen Ma
- The First School of Clinical Medicine, Lanzhou University, Lanzhou 730000, Gansu Province, China
- Department of Gastroenterology, Gansu Provincial People's Hospital, Lanzhou 730000, Gansu Province, China
| | - Ni Zhou
- Department of Gastroenterology, Xi'an International Medical Center, Xi’an 710000, Shaanxi Province, China
| | - Xiu Luo
- The First School of Clinical Medicine, Lanzhou University, Lanzhou 730000, Gansu Province, China
| | - Si-Qi Guo
- Department of Gastroenterology, Gansu Provincial People's Hospital, Lanzhou 730000, Gansu Province, China
- The First School of Clinical Medicine, Gansu University of Chinese Medicine, Lanzhou 730000, Gansu Province, China
| | - Ping Mai
- The First School of Clinical Medicine, Lanzhou University, Lanzhou 730000, Gansu Province, China
- Department of Gastroenterology, Gansu Provincial People's Hospital, Lanzhou 730000, Gansu Province, China
| |
Collapse
|
10
|
Huang M, Luo S, Yang J, Xiong H, Lu X, Ma X, Zeng J, Efferth T. Optimized therapeutic potential of Sijunzi-similar formulae for chronic atrophic gastritis via Bayesian network meta-analysis. EXCLI JOURNAL 2024; 23:1185-1207. [PMID: 39421026 PMCID: PMC11484511 DOI: 10.17179/excli2024-7618] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Accepted: 08/29/2024] [Indexed: 10/19/2024]
Abstract
Chronic atrophic gastritis (CAG) is considered as a significant risk factor for triggering gastric cancer incidence, if not effectively treated. Sijunzi decoction (SD) is a well-known classic formula for treating gastric disorders, and Sijunzi-similar formulae (SF) derived from SD have also been highly regarded by Chinese clinical practitioners for their effectiveness in treating chronic atrophic gastritis. Currently, there is a lack of meta-analysis for these formulae, leaving unclear which exhibits optimal efficacy. Therefore, we employed Bayesian network meta-analysis (BNMA) to evaluate the efficacy and safety of SF as an intervention for CAG and to establish a scientific foundation for the clinical utilization of SF. The result of meta-analysis demonstrated that the combination of SF and basic therapy outperformed basic therapy alone in terms of clinical efficacy rate, eradication rate of H. pylori, and incidence of adverse events. As indicated by the SUCRA value, Chaishao Liujunzi decoction (CLD) demonstrated superior efficacy in enhancing clinical effectiveness and ameliorating H. pylori infection, and it also showed remarkable effectiveness in minimizing the occurrence of adverse events. Comprehensive analysis of therapeutic efficacy suggests that CLD is most likely the optimal choice among these six formulations, holding potential value for optimizing clinical treatment strategies. See also the graphical abstract(Fig. 1).
Collapse
Affiliation(s)
- Meilan Huang
- Department of Gastroenterology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610072, China
- School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu 610075, China
| | - Shiman Luo
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Jiayue Yang
- Department of Gastroenterology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610072, China
| | - Huiling Xiong
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Xiaohua Lu
- Department of Pharmaceutical Biology, Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University, 55128 Mainz, Germany
| | - Xiao Ma
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
- Department of Pharmaceutical Biology, Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University, 55128 Mainz, Germany
| | - Jinhao Zeng
- Department of Gastroenterology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610072, China
- School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu 610075, China
- TCM Regulating Metabolic Disease Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610072, China
| | - Thomas Efferth
- Department of Pharmaceutical Biology, Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University, 55128 Mainz, Germany
| |
Collapse
|
11
|
Li Z, Zheng X, Mu Y, Zhang M, Liu G. The intelligent gastrointestinal metaplasia assessment based on deformable transformer with token merging. Biomed Signal Process Control 2024; 95:106454. [DOI: 10.1016/j.bspc.2024.106454] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
12
|
Turtoi DC, Brata VD, Incze V, Ismaiel A, Dumitrascu DI, Militaru V, Munteanu MA, Botan A, Toc DA, Duse TA, Popa SL. Artificial Intelligence for the Automatic Diagnosis of Gastritis: A Systematic Review. J Clin Med 2024; 13:4818. [PMID: 39200959 PMCID: PMC11355427 DOI: 10.3390/jcm13164818] [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: 07/23/2024] [Revised: 08/11/2024] [Accepted: 08/13/2024] [Indexed: 09/02/2024] Open
Abstract
Background and Objective: Gastritis represents one of the most prevalent gastrointestinal diseases and has a multifactorial etiology, many forms of manifestation, and various symptoms. Diagnosis of gastritis is made based on clinical, endoscopic, and histological criteria, and although it is a thorough process, many cases are misdiagnosed or overlooked. This systematic review aims to provide an extensive overview of current artificial intelligence (AI) applications in gastritis diagnosis and evaluate the precision of these systems. This evaluation could highlight the role of AI as a helpful and useful tool in facilitating timely and accurate diagnoses, which in turn could improve patient outcomes. Methods: We have conducted an extensive and comprehensive literature search of PubMed, Scopus, and Web of Science, including studies published until July 2024. Results: Despite variations in study design, participant numbers and characteristics, and outcome measures, our observations suggest that implementing an AI automatic diagnostic tool into clinical practice is currently feasible, with the current systems achieving high levels of accuracy, sensitivity, and specificity. Our findings indicate that AI outperformed human experts in most studies, with multiple studies exhibiting an accuracy of over 90% for AI compared to human experts. These results highlight the significant potential of AI to enhance diagnostic accuracy and efficiency in gastroenterology. Conclusions: AI-based technologies can now automatically diagnose using images provided by gastroscopy, digital pathology, and radiology imaging. Deep learning models exhibited high levels of accuracy, sensitivity, and specificity while assessing the diagnosis, staging, and risk of neoplasia for different types of gastritis, results that are superior to those of human experts in most studies.
Collapse
Affiliation(s)
- Daria Claudia Turtoi
- Faculty of Medicine, “Iuliu Hatieganu” University of Medicine and Pharmacy, 400000 Cluj-Napoca, Romania; (D.C.T.); (V.I.); (A.B.); (T.A.D.)
| | - Vlad Dumitru Brata
- Faculty of Medicine, “Iuliu Hatieganu” University of Medicine and Pharmacy, 400000 Cluj-Napoca, Romania; (D.C.T.); (V.I.); (A.B.); (T.A.D.)
| | - Victor Incze
- Faculty of Medicine, “Iuliu Hatieganu” University of Medicine and Pharmacy, 400000 Cluj-Napoca, Romania; (D.C.T.); (V.I.); (A.B.); (T.A.D.)
| | - Abdulrahman Ismaiel
- 2nd Medical Department, “Iuliu Hatieganu” University of Medicine and Pharmacy, 400000 Cluj-Napoca, Romania; (A.I.); (S.L.P.)
| | - Dinu Iuliu Dumitrascu
- Department of Anatomy, “Iuliu Hatieganu” University of Medicine and Pharmacy, 400000 Cluj-Napoca, Romania;
| | - Valentin Militaru
- Department of Internal Medicine, Clinical Municipal Hospital, 400139 Cluj-Napoca, Romania;
| | - Mihai Alexandru Munteanu
- Department of Medical Disciplines, Faculty of Medicine and Pharmacy, University of Oradea, 410087 Oradea, Romania;
| | - Alexandru Botan
- Faculty of Medicine, “Iuliu Hatieganu” University of Medicine and Pharmacy, 400000 Cluj-Napoca, Romania; (D.C.T.); (V.I.); (A.B.); (T.A.D.)
| | - Dan Alexandru Toc
- Department of Microbiology, “Iuliu Hatieganu” University of Medicine and Pharmacy, 400000 Cluj-Napoca, Romania;
| | - Traian Adrian Duse
- Faculty of Medicine, “Iuliu Hatieganu” University of Medicine and Pharmacy, 400000 Cluj-Napoca, Romania; (D.C.T.); (V.I.); (A.B.); (T.A.D.)
| | - Stefan Lucian Popa
- 2nd Medical Department, “Iuliu Hatieganu” University of Medicine and Pharmacy, 400000 Cluj-Napoca, Romania; (A.I.); (S.L.P.)
| |
Collapse
|
13
|
Bou Jaoude J, Al Bacha R, Abboud B. Will artificial intelligence reach any limit in gastroenterology? Artif Intell Gastroenterol 2024; 5:91336. [DOI: 10.35712/aig.v5.i2.91336] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Revised: 04/25/2024] [Accepted: 06/07/2024] [Indexed: 08/08/2024] Open
Abstract
Endoscopy is the cornerstone in the management of digestive diseases. Over the last few decades, technology has played an important role in the development of this field, helping endoscopists in better detecting and characterizing luminal lesions. However, despite ongoing advancements in endoscopic technology, the incidence of missed pre-neoplastic and neoplastic lesions remains high due to the operator-dependent nature of endoscopy and the challenging learning curve associated with new technologies. Artificial intelligence (AI), an operator-independent field, could be an invaluable solution. AI can serve as a “second observer”, enhancing the performance of endoscopists in detecting and characterizing luminal lesions. By utilizing deep learning (DL), an innovation within machine learning, AI automatically extracts input features from targeted endoscopic images. DL encompasses both computer-aided detection and computer-aided diagnosis, assisting endoscopists in reducing missed detection rates and predicting the histology of luminal digestive lesions. AI applications in clinical gastrointestinal diseases are continuously expanding and evolving the entire digestive tract. In all published studies, real-time AI assists endoscopists in improving the performance of non-expert gastroenterologists, bringing it to a level comparable to that of experts. The development of DL may be affected by selection biases. Studies have utilized different AI-assisted models, which are heterogeneous. In the future, algorithms need validation through large, randomized trials. Theoretically, AI has no limit to assist endoscopists in increasing the accuracy and the quality of endoscopic exams. However, practically, we still have a long way to go before standardizing our AI models to be accepted and applied by all gastroenterologists.
Collapse
Affiliation(s)
- Joseph Bou Jaoude
- Department of Gastroenterology, Levant Hospital, Beirut 166830, Lebanon
| | - Rose Al Bacha
- Department of Gastroenterology, Levant Hospital, Beirut 166830, Lebanon
| | - Bassam Abboud
- Department of General Surgery, Geitaoui Hospital, Faculty of Medicine, Lebanese University, Lebanon, Beirut 166830, Lebanon
| |
Collapse
|
14
|
Tao X, Zhu Y, Dong Z, Huang L, Shang R, Du H, Wang J, Zeng X, Wang W, Wang J, Li Y, Deng Y, Wu L, Yu H. An artificial intelligence system for chronic atrophic gastritis diagnosis and risk stratification under white light endoscopy. Dig Liver Dis 2024; 56:1319-1326. [PMID: 38246825 DOI: 10.1016/j.dld.2024.01.177] [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: 07/12/2023] [Revised: 11/06/2023] [Accepted: 01/05/2024] [Indexed: 01/23/2024]
Abstract
BACKGROUND AND AIMS The diagnosis and stratification of gastric atrophy (GA) predict patients' gastric cancer progression risk and determine endoscopy surveillance interval. We aimed to construct an artificial intelligence (AI) system for GA endoscopic identification and risk stratification based on the Kimura-Takemoto classification. METHODS We constructed the system using two trained models and verified its performance. First, we retrospectively collected 869 images and 119 videos to compare its performance with that of endoscopists in identifying GA. Then, we included original image cases of 102 patients to validate the system for stratifying GA and comparing it with endoscopists with different experiences. RESULTS The sensitivity of model 1 was higher than that of endoscopists (92.72% vs. 76.85 %) at image level and also higher than that of experts (94.87% vs. 85.90 %) at video level. The system outperformed experts in stratifying GA (overall accuracy: 81.37 %, 73.04 %, p = 0.045). The accuracy of this system in classifying non-GA, mild GA, moderate GA, and severe GA was 80.00 %, 77.42 %, 83.33 %, and 85.71 %, comparable to that of experts and better than that of seniors and novices. CONCLUSIONS We established an expert-level system for GA endoscopic identification and risk stratification. It has great potential for endoscopic assessment and surveillance determinations.
Collapse
Affiliation(s)
- Xiao Tao
- Renmin Hospital of Wuhan University, Wuhan, PR China; Key Laboratory of Hubei Province for Digestive System Disease, Renmin Hospital of Wuhan University, Wuhan, PR China; Hubei Provincial Clinical Research Center for Digestive Disease Minimally Invasive Incision, Renmin Hospital of Wuhan University, Wuhan, PR China
| | - Yijie Zhu
- Renmin Hospital of Wuhan University, Wuhan, PR China; Key Laboratory of Hubei Province for Digestive System Disease, Renmin Hospital of Wuhan University, Wuhan, PR China; Hubei Provincial Clinical Research Center for Digestive Disease Minimally Invasive Incision, Renmin Hospital of Wuhan University, Wuhan, PR China; Department of Gastroenterology, Yunnan Digestive Endoscopy Clinical Medical Center, The First People's Hospital of Yunnan Province, Kunming, 650032, PR China
| | - Zehua Dong
- Renmin Hospital of Wuhan University, Wuhan, PR China; Key Laboratory of Hubei Province for Digestive System Disease, Renmin Hospital of Wuhan University, Wuhan, PR China; Hubei Provincial Clinical Research Center for Digestive Disease Minimally Invasive Incision, Renmin Hospital of Wuhan University, Wuhan, PR China
| | - Li Huang
- Renmin Hospital of Wuhan University, Wuhan, PR China; Key Laboratory of Hubei Province for Digestive System Disease, Renmin Hospital of Wuhan University, Wuhan, PR China; Hubei Provincial Clinical Research Center for Digestive Disease Minimally Invasive Incision, Renmin Hospital of Wuhan University, Wuhan, PR China
| | - Renduo Shang
- Renmin Hospital of Wuhan University, Wuhan, PR China; Key Laboratory of Hubei Province for Digestive System Disease, Renmin Hospital of Wuhan University, Wuhan, PR China; Hubei Provincial Clinical Research Center for Digestive Disease Minimally Invasive Incision, Renmin Hospital of Wuhan University, Wuhan, PR China
| | - Hongliu Du
- Renmin Hospital of Wuhan University, Wuhan, PR China; Key Laboratory of Hubei Province for Digestive System Disease, Renmin Hospital of Wuhan University, Wuhan, PR China; Hubei Provincial Clinical Research Center for Digestive Disease Minimally Invasive Incision, Renmin Hospital of Wuhan University, Wuhan, PR China
| | - Junxiao Wang
- Renmin Hospital of Wuhan University, Wuhan, PR China; Key Laboratory of Hubei Province for Digestive System Disease, Renmin Hospital of Wuhan University, Wuhan, PR China; Hubei Provincial Clinical Research Center for Digestive Disease Minimally Invasive Incision, Renmin Hospital of Wuhan University, Wuhan, PR China
| | - Xiaoquan Zeng
- Renmin Hospital of Wuhan University, Wuhan, PR China; Key Laboratory of Hubei Province for Digestive System Disease, Renmin Hospital of Wuhan University, Wuhan, PR China; Hubei Provincial Clinical Research Center for Digestive Disease Minimally Invasive Incision, Renmin Hospital of Wuhan University, Wuhan, PR China
| | - Wen Wang
- Renmin Hospital of Wuhan University, Wuhan, PR China; Key Laboratory of Hubei Province for Digestive System Disease, Renmin Hospital of Wuhan University, Wuhan, PR China; Hubei Provincial Clinical Research Center for Digestive Disease Minimally Invasive Incision, Renmin Hospital of Wuhan University, Wuhan, PR China
| | - Jiamin Wang
- Renmin Hospital of Wuhan University, Wuhan, PR China; Key Laboratory of Hubei Province for Digestive System Disease, Renmin Hospital of Wuhan University, Wuhan, PR China; Hubei Provincial Clinical Research Center for Digestive Disease Minimally Invasive Incision, Renmin Hospital of Wuhan University, Wuhan, PR China
| | - Yanxia Li
- Renmin Hospital of Wuhan University, Wuhan, PR China; Key Laboratory of Hubei Province for Digestive System Disease, Renmin Hospital of Wuhan University, Wuhan, PR China; Hubei Provincial Clinical Research Center for Digestive Disease Minimally Invasive Incision, Renmin Hospital of Wuhan University, Wuhan, PR China
| | - Yunchao Deng
- Renmin Hospital of Wuhan University, Wuhan, PR China; Key Laboratory of Hubei Province for Digestive System Disease, Renmin Hospital of Wuhan University, Wuhan, PR China; Hubei Provincial Clinical Research Center for Digestive Disease Minimally Invasive Incision, Renmin Hospital of Wuhan University, Wuhan, PR China
| | - Lianlian Wu
- Renmin Hospital of Wuhan University, Wuhan, PR China; Key Laboratory of Hubei Province for Digestive System Disease, Renmin Hospital of Wuhan University, Wuhan, PR China; Hubei Provincial Clinical Research Center for Digestive Disease Minimally Invasive Incision, Renmin Hospital of Wuhan University, Wuhan, PR China.
| | - Honggang Yu
- Renmin Hospital of Wuhan University, Wuhan, PR China; Key Laboratory of Hubei Province for Digestive System Disease, Renmin Hospital of Wuhan University, Wuhan, PR China; Hubei Provincial Clinical Research Center for Digestive Disease Minimally Invasive Incision, Renmin Hospital of Wuhan University, Wuhan, PR China.
| |
Collapse
|
15
|
Jia J, Zhao H, Li F, Zheng Q, Wang G, Li D, Liu Y. Research on drug treatment and the novel signaling pathway of chronic atrophic gastritis. Biomed Pharmacother 2024; 176:116912. [PMID: 38850667 DOI: 10.1016/j.biopha.2024.116912] [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: 04/16/2024] [Revised: 06/04/2024] [Accepted: 06/06/2024] [Indexed: 06/10/2024] Open
Abstract
BACKGROUND Chronic atrophic gastritis (CAG) is a global digestive system disease and one of the important causes of gastric cancer. The incidence of CAG has been increasing yearly worldwide. PURPOSE This article reviews the latest research on the common causes and future therapeutic targets of CAG as well as the pharmacological effects of corresponding clinical drugs. We provide a detailed theoretical basis for further research on possible methods for the treatment of CAG and reversal of the CAG process. RESULTS CAG often develops from chronic gastritis, and its main pathological manifestation is atrophy of the gastric mucosa, which can develop into gastric cancer. The drug treatment of CAG can be divided into agents that regulate gastric acid secretion, eradicate Helicobacter. pylori (H. pylori), protect gastric mucous membrane, or inhibit inflammatory factors according to their mechanism of action. Although there are limited specific drugs for the treatment of CAG, progress is being made in defining the pathogenesis and therapeutic targets of the disease. Growing evidence shows that NF-κB, PI3K/AKT, Wnt/ β-catenin, MAPK, Toll-like receptors (TLRs), Hedgehog, and VEGF signaling pathways play an important role in the development of CAG.
Collapse
Affiliation(s)
- Jinhao Jia
- Featured Laboratory for Biosynthesis and Target Discovery of Active Components of Traditional Chinese Medicine, School of Traditional Chinese Medicine & Binzhou Hospital of Traditional Chinese Medicine, Binzhou Medical University, Yantai, Shandong 264003, PR China
| | - Huijie Zhao
- Featured Laboratory for Biosynthesis and Target Discovery of Active Components of Traditional Chinese Medicine, School of Traditional Chinese Medicine & Binzhou Hospital of Traditional Chinese Medicine, Binzhou Medical University, Yantai, Shandong 264003, PR China
| | - Fangfei Li
- Shum Yiu Foon Shum Bik Chuen Memorial Centre for Cancer and Inflammation Research, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, Special Administrative Region of China
| | - Qiusheng Zheng
- Featured Laboratory for Biosynthesis and Target Discovery of Active Components of Traditional Chinese Medicine, School of Traditional Chinese Medicine & Binzhou Hospital of Traditional Chinese Medicine, Binzhou Medical University, Yantai, Shandong 264003, PR China; Key Laboratory of Xinjiang Phytomedicine Resource and Utilization, Ministry of Education, School of Pharmacy, Shihezi University, Shihezi, Xinjiang 832003, PR China
| | - Guoli Wang
- Featured Laboratory for Biosynthesis and Target Discovery of Active Components of Traditional Chinese Medicine, School of Traditional Chinese Medicine & Binzhou Hospital of Traditional Chinese Medicine, Binzhou Medical University, Yantai, Shandong 264003, PR China
| | - Defang Li
- Featured Laboratory for Biosynthesis and Target Discovery of Active Components of Traditional Chinese Medicine, School of Traditional Chinese Medicine & Binzhou Hospital of Traditional Chinese Medicine, Binzhou Medical University, Yantai, Shandong 264003, PR China; Key Laboratory of Xinjiang Phytomedicine Resource and Utilization, Ministry of Education, School of Pharmacy, Shihezi University, Shihezi, Xinjiang 832003, PR China.
| | - Ying Liu
- Featured Laboratory for Biosynthesis and Target Discovery of Active Components of Traditional Chinese Medicine, School of Traditional Chinese Medicine & Binzhou Hospital of Traditional Chinese Medicine, Binzhou Medical University, Yantai, Shandong 264003, PR China.
| |
Collapse
|
16
|
Matsubayashi CO, Cheng S, Hulchafo I, Zhang Y, Tada T, Buxbaum JL, Ochiai K. Artificial intelligence for gastric cancer in endoscopy: From diagnostic reasoning to market. Dig Liver Dis 2024; 56:1156-1163. [PMID: 38763796 DOI: 10.1016/j.dld.2024.04.019] [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: 10/19/2023] [Revised: 04/15/2024] [Accepted: 04/16/2024] [Indexed: 05/21/2024]
Abstract
Recognition of gastric conditions during endoscopy exams, including gastric cancer, usually requires specialized training and a long learning curve. Besides that, the interobserver variability is frequently high due to the different morphological characteristics of the lesions and grades of mucosal inflammation. In this sense, artificial intelligence tools based on deep learning models have been developed to support physicians to detect, classify, and predict gastric lesions more efficiently. Even though a growing number of studies exists in the literature, there are multiple challenges to bring a model to practice in this field, such as the need for more robust validation studies and regulatory hurdles. Therefore, the aim of this review is to provide a comprehensive assessment of the current use of artificial intelligence applied to endoscopic imaging to evaluate gastric precancerous and cancerous lesions and the barriers to widespread implementation of this technology in clinical routine.
Collapse
Affiliation(s)
- Carolina Ogawa Matsubayashi
- Endoscopy Unit, Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, University of São Paulo, São Paulo, Brasil; AI Medical Service Inc., Tokyo, Japan.
| | - Shuyan Cheng
- Department of Population Health Science, Weill Cornell Medical College, New York, NY 10065, USA
| | - Ismael Hulchafo
- Columbia University School of Nursing, New York, NY 10032, USA
| | - Yifan Zhang
- Department of Population Health Science, Weill Cornell Medical College, New York, NY 10065, USA
| | - Tomohiro Tada
- AI Medical Service Inc., Tokyo, Japan; Department of Surgical Oncology, Faculty of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - James L Buxbaum
- Division of Gastrointestinal and Liver Diseases, Keck School of Medicine of the University of Southern California, Los Angeles, California, USA
| | - Kentaro Ochiai
- Department of Surgical Oncology, Faculty of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo 113-0033, Japan; Department of Colon and Rectal Surgery, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| |
Collapse
|
17
|
Bangolo A, Wadhwani N, Nagesh VK, Dey S, Tran HHV, Aguilar IK, Auda A, Sidiqui A, Menon A, Daoud D, Liu J, Pulipaka SP, George B, Furman F, Khan N, Plumptre A, Sekhon I, Lo A, Weissman S. Impact of artificial intelligence in the management of esophageal, gastric and colorectal malignancies. Artif Intell Gastrointest Endosc 2024; 5:90704. [DOI: 10.37126/aige.v5.i2.90704] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 01/28/2024] [Accepted: 03/04/2024] [Indexed: 05/11/2024] Open
Abstract
The incidence of gastrointestinal malignancies has increased over the past decade at an alarming rate. Colorectal and gastric cancers are the third and fifth most commonly diagnosed cancers worldwide but are cited as the second and third leading causes of mortality. Early institution of appropriate therapy from timely diagnosis can optimize patient outcomes. Artificial intelligence (AI)-assisted diagnostic, prognostic, and therapeutic tools can assist in expeditious diagnosis, treatment planning/response prediction, and post-surgical prognostication. AI can intercept neoplastic lesions in their primordial stages, accurately flag suspicious and/or inconspicuous lesions with greater accuracy on radiologic, histopathological, and/or endoscopic analyses, and eliminate over-dependence on clinicians. AI-based models have shown to be on par, and sometimes even outperformed experienced gastroenterologists and radiologists. Convolutional neural networks (state-of-the-art deep learning models) are powerful computational models, invaluable to the field of precision oncology. These models not only reliably classify images, but also accurately predict response to chemotherapy, tumor recurrence, metastasis, and survival rates post-treatment. In this systematic review, we analyze the available evidence about the diagnostic, prognostic, and therapeutic utility of artificial intelligence in gastrointestinal oncology.
Collapse
Affiliation(s)
- Ayrton Bangolo
- Department of Internal Medicine, Palisades Medical Center, North Bergen, NJ 07047, United States
| | - Nikita Wadhwani
- Department of Internal Medicine, Palisades Medical Center, North Bergen, NJ 07047, United States
| | - Vignesh K Nagesh
- Department of Internal Medicine, Palisades Medical Center, North Bergen, NJ 07047, United States
| | - Shraboni Dey
- Department of Internal Medicine, Palisades Medical Center, North Bergen, NJ 07047, United States
| | - Hadrian Hoang-Vu Tran
- Department of Internal Medicine, Palisades Medical Center, North Bergen, NJ 07047, United States
| | - Izage Kianifar Aguilar
- Department of Internal Medicine, Palisades Medical Center, North Bergen, NJ 07047, United States
| | - Auda Auda
- Department of Medicine, Palisades Medical Center, North Bergen, NJ 07047, United States
| | - Aman Sidiqui
- Department of Internal Medicine, Palisades Medical Center, North Bergen, NJ 07047, United States
| | - Aiswarya Menon
- Department of Internal Medicine, Palisades Medical Center, North Bergen, NJ 07047, United States
| | - Deborah Daoud
- Department of Medicine, Palisades Medical Center, North Bergen, NJ 07047, United States
| | - James Liu
- Department of Internal Medicine, Palisades Medical Center, North Bergen, NJ 07047, United States
| | - Sai Priyanka Pulipaka
- Department of Medicine, Palisades Medical Center, North Bergen, NJ 07047, United States
| | - Blessy George
- Department of Internal Medicine, Palisades Medical Center, North Bergen, NJ 07047, United States
| | - Flor Furman
- Department of Internal Medicine, Palisades Medical Center, North Bergen, NJ 07047, United States
| | - Nareeman Khan
- Department of Internal Medicine, Palisades Medical Center, North Bergen, NJ 07047, United States
| | - Adewale Plumptre
- Department of Internal Medicine, Palisades Medical Center, North Bergen, NJ 07047, United States
| | - Imranjot Sekhon
- Department of Internal Medicine, Palisades Medical Center, North Bergen, NJ 07047, United States
| | - Abraham Lo
- Department of Medicine, Palisades Medical Center, North Bergen, NJ 07047, United States
| | - Simcha Weissman
- Department of Internal Medicine, Palisades Medical Center, North Bergen, NJ 07047, United States
| |
Collapse
|
18
|
Deng B, Zheng X, Chen X, Zhang M. A Swin transformer encoder-based StyleGAN for unbalanced endoscopic image enhancement. Comput Biol Med 2024; 175:108472. [PMID: 38663349 DOI: 10.1016/j.compbiomed.2024.108472] [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: 10/23/2023] [Revised: 02/22/2024] [Accepted: 04/10/2024] [Indexed: 05/15/2024]
Abstract
With the rapid development of artificial intelligence, automated endoscopy-assisted diagnostic systems have become an effective tool for reducing the diagnostic costs and shortening the treatment cycle of patients. Typically, the performance of these systems depends on deep learning models which are pre-trained with large-scale labeled data, for example, early gastric cancer based on endoscopic images. However, the expensive annotation and the subjectivity of the annotators lead to an insufficient and class-imbalanced endoscopic image dataset, and these datasets are detrimental to the training of deep learning models. Therefore, we proposed a Swin Transformer encoder-based StyleGAN (STE-StyleGAN) for unbalanced endoscopic image enhancement, which is composed of an adversarial learning encoder and generator. Firstly, a pre-trained Swin Transformer is introduced into the encoder to extract multi-scale features layer by layer from endoscopic images. The features are subsequently fed into a mapping block for aggregation and recombination. Secondly, a self-attention mechanism is applied to the generator, which adds detailed information of the image layer by layer through recoded features, enabling the generator to autonomously learn the coupling between different image regions. Finally, we conducted extensive experiments on a private intestinal metaplasia grading dataset from a Grade-A tertiary hospital. The experimental results show that the images generated by STE-StyleGAN are closer to the initial image distribution, achieving a Fréchet Inception Distance (FID) value of 100.4. Then, these generated images are used to enhance the initial dataset to improve the robustness of the classification model, and achieved a top accuracy of 86 %.
Collapse
Affiliation(s)
- Bo Deng
- School of Information Science and Engineering, Shandong Normal University, Jinan, 250352, China
| | - Xiangwei Zheng
- School of Information Science and Engineering, Shandong Normal University, Jinan, 250352, China; Shandong Provincial Key Laboratory for Distributed Computer Software Novel Technology, Jinan, 250352, China; State Key Laboratory of High-end Server & Storage Technology, Jinan 250101, China.
| | - Xuanchi Chen
- School of Information Science and Engineering, Shandong Normal University, Jinan, 250352, China
| | - Mingzhe Zhang
- School of Information Science and Engineering, Shandong Normal University, Jinan, 250352, China
| |
Collapse
|
19
|
Du K, Dong L, Zhang K, Guan M, Chen C, Xie L, Kong W, Li H, Zhang R, Zhou W, Wu H, Dong H, Wei W. Deep learning system for screening AIDS-related cytomegalovirus retinitis with ultra-wide-field fundus images. Heliyon 2024; 10:e30881. [PMID: 38803983 PMCID: PMC11128864 DOI: 10.1016/j.heliyon.2024.e30881] [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: 12/03/2023] [Revised: 05/03/2024] [Accepted: 05/07/2024] [Indexed: 05/29/2024] Open
Abstract
Background Ophthalmological screening for cytomegalovirus retinitis (CMVR) for HIV/AIDS patients is important to prevent lifelong blindness. Previous studies have shown good properties of automated CMVR screening using digital fundus images. However, the application of a deep learning (DL) system to CMVR with ultra-wide-field (UWF) fundus images has not been studied, and the feasibility and efficiency of this method are uncertain. Methods In this study, we developed, internally validated, externally validated, and prospectively validated a DL system to detect AIDS-related from UWF fundus images from different clinical datasets. We independently used the InceptionResnetV2 network to develop and internally validate a DL system for identifying active CMVR, inactive CMVR, and non-CMVR in 6960 UWF fundus images from 862 AIDS patients and validated the system in a prospective and an external validation data set using the area under the curve (AUC), accuracy, sensitivity, and specificity. A heat map identified the most important area (lesions) used by the DL system for differentiating CMVR. Results The DL system showed AUCs of 0.945 (95 % confidence interval [CI]: 0.929, 0.962), 0.964 (95 % CI: 0.870, 0.999) and 0.968 (95 % CI: 0.860, 1.000) for detecting active CMVR from non-CMVR and 0.923 (95 % CI: 0.908, 0.938), 0.902 (0.857, 0.948) and 0.884 (0.851, 0.917) for detecting active CMVR from non-CMVR in the internal cross-validation, external validation, and prospective validation, respectively. Deep learning performed promisingly in screening CMVR. It also showed the ability to differentiate active CMVR from non-CMVR and inactive CMVR as well as to identify active CMVR and inactive CMVR from non-CMVR (all AUCs in the three independent data sets >0.900). The heat maps successfully highlighted lesion locations. Conclusions Our UWF fundus image-based DL system showed reliable performance for screening AIDS-related CMVR showing its potential for screening CMVR in HIV/AIDS patients, especially in the absence of ophthalmic resources.
Collapse
Affiliation(s)
- Kuifang Du
- Department of Ophthalmology, Beijing Youan Hospital, Capital Medical University, Beijing, China
| | - Li Dong
- Beijing Tongren Eye Centre, Beijing Key Laboratory of Intraocular Tumour Diagnosis and Treatment, Beijing Ophthalmology & Visual Sciences Key Lab, Medical Artificial Intelligence Research and Verification Key Laboratory of the Ministry of Industry and Information Technology, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Kai Zhang
- Chongqing Chang'an Industrial Group Co. Ltd, Chongqing, China
| | - Meilin Guan
- Department of Ophthalmology, Beijing Youan Hospital, Capital Medical University, Beijing, China
| | - Chao Chen
- Department of Ophthalmology, Beijing Youan Hospital, Capital Medical University, Beijing, China
| | - Lianyong Xie
- Department of Ophthalmology, Beijing Youan Hospital, Capital Medical University, Beijing, China
| | - Wenjun Kong
- Department of Ophthalmology, Beijing Youan Hospital, Capital Medical University, Beijing, China
| | - Heyan Li
- Beijing Tongren Eye Centre, Beijing Key Laboratory of Intraocular Tumour Diagnosis and Treatment, Beijing Ophthalmology & Visual Sciences Key Lab, Medical Artificial Intelligence Research and Verification Key Laboratory of the Ministry of Industry and Information Technology, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Ruiheng Zhang
- Beijing Tongren Eye Centre, Beijing Key Laboratory of Intraocular Tumour Diagnosis and Treatment, Beijing Ophthalmology & Visual Sciences Key Lab, Medical Artificial Intelligence Research and Verification Key Laboratory of the Ministry of Industry and Information Technology, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Wenda Zhou
- Beijing Tongren Eye Centre, Beijing Key Laboratory of Intraocular Tumour Diagnosis and Treatment, Beijing Ophthalmology & Visual Sciences Key Lab, Medical Artificial Intelligence Research and Verification Key Laboratory of the Ministry of Industry and Information Technology, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Haotian Wu
- Beijing Tongren Eye Centre, Beijing Key Laboratory of Intraocular Tumour Diagnosis and Treatment, Beijing Ophthalmology & Visual Sciences Key Lab, Medical Artificial Intelligence Research and Verification Key Laboratory of the Ministry of Industry and Information Technology, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Hongwei Dong
- Department of Ophthalmology, Beijing Youan Hospital, Capital Medical University, Beijing, China
| | - Wenbin Wei
- Beijing Tongren Eye Centre, Beijing Key Laboratory of Intraocular Tumour Diagnosis and Treatment, Beijing Ophthalmology & Visual Sciences Key Lab, Medical Artificial Intelligence Research and Verification Key Laboratory of the Ministry of Industry and Information Technology, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
20
|
Bai CY, Tian W, Zhang Q. Clinical study on microscopic syndrome differentiation and traditional Chinese medicine treatment for liver stomach disharmony in chronic gastritis. World J Gastrointest Surg 2024; 16:1377-1384. [PMID: 38817300 PMCID: PMC11135293 DOI: 10.4240/wjgs.v16.i5.1377] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Revised: 04/05/2024] [Accepted: 04/11/2024] [Indexed: 05/23/2024] Open
Abstract
BACKGROUND Chronic gastritis (CG) is a common gastrointestinal disorder characterized by inflammation of the stomach lining. Liver-stomach disharmony (LSD) syndrome is believed to contribute to CG symptoms. AIM To evaluate the efficacy and safety of microcosmic syndrome differentiation and Chinese herbal medicine (CHM) treatment in patients with CG and LSD syndrome. METHODS Sixty-four patients with CG and LSD syndrome were randomly divided into two groups: The treatment group received CHM based on microcosmic syndrome differentiation and the control group received conventional Western medicine. The treatment course lasted 12 wk. The primary outcome was improvement in dyspeptic symptoms, measured using the Nepean Dyspepsia Index. The secondary outcomes included the improvement rate of endoscopic findings, histopathological findings, and microcosmic syndrome scores and the incidence of adverse events. RESULTS After 12 wk of treatment, the treatment group showed significantly greater improvement in dyspeptic symptoms than the control group (93.75% vs 65.63%, P < 0.01). The treatment group also showed a significantly higher improvement rate in endoscopic findings than the control group (81.25% vs 53.13%, P < 0.05). The improvement rates of histopathological findings and microcosmic syndrome scores were not significantly different between the two groups (P > 0.05). No serious adverse events were observed in either group. CONCLUSION Microcosmic syndrome differentiation and CHM treatment can effectively improve dyspeptic symptoms and endoscopic findings in patients with CG and LSD syndrome and have a good safety profile. Further studies with larger sample sizes and longer follow-up periods are required to confirm the long-term efficacy and mechanism of action of this treatment.
Collapse
Affiliation(s)
- Chun-Yan Bai
- Department of Rehabilitation Medicine, Beijing Aerospace General Hospital, Beijing 100076, China
| | - Wei Tian
- Department of Rehabilitation Medicine, People’s Hospital of Hengshui, Hengshui 053000, Hebei Province, China
| | - Qian Zhang
- Department of Internal Medicine, Hebei Academy of Chinese Medicine Sciences, Shijiazhuang 050000, Hebei Province, China
| |
Collapse
|
21
|
Gong EJ, Bang CS, Lee JJ. Computer-aided diagnosis in real-time endoscopy for all stages of gastric carcinogenesis: Development and validation study. United European Gastroenterol J 2024; 12:487-495. [PMID: 38400815 PMCID: PMC11091781 DOI: 10.1002/ueg2.12551] [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: 08/28/2023] [Accepted: 01/14/2024] [Indexed: 02/26/2024] Open
Abstract
OBJECTIVE Using endoscopic images, we have previously developed computer-aided diagnosis models to predict the histopathology of gastric neoplasms. However, no model that categorizes every stage of gastric carcinogenesis has been published. In this study, a deep-learning-based diagnosis model was developed and validated to automatically classify all stages of gastric carcinogenesis, including atrophy and intestinal metaplasia, in endoscopy images. DESIGN A total of 18,701 endoscopic images were collected retrospectively and randomly divided into train, validation, and internal-test datasets in an 8:1:1 ratio. The primary outcome was lesion-classification accuracy in six categories: normal/atrophy/intestinal metaplasia/dysplasia/early /advanced gastric cancer. External-validation of performance in the established model used 1427 novel images from other institutions that were not used in training, validation, or internal-tests. RESULTS The internal-test lesion-classification accuracy was 91.2% (95% confidence interval: 89.9%-92.5%). For performance validation, the established model achieved an accuracy of 82.3% (80.3%-84.3%). The external-test per-class receiver operating characteristic in the diagnosis of atrophy and intestinal metaplasia was 93.4 ± 0% and 91.3 ± 0%, respectively. CONCLUSIONS The established model demonstrated high performance in the diagnosis of preneoplastic lesions (atrophy and intestinal metaplasia) as well as gastric neoplasms.
Collapse
Affiliation(s)
- Eun Jeong Gong
- Department of Internal MedicineHallym University College of MedicineChuncheonKorea
- Institute for Liver and Digestive DiseasesHallym UniversityChuncheonKorea
- Institute of New Frontier ResearchHallym University College of MedicineChuncheonKorea
| | - Chang Seok Bang
- Department of Internal MedicineHallym University College of MedicineChuncheonKorea
- Institute for Liver and Digestive DiseasesHallym UniversityChuncheonKorea
- Institute of New Frontier ResearchHallym University College of MedicineChuncheonKorea
- Division of Big Data and Artificial IntelligenceChuncheon Sacred Heart HospitalChuncheonKorea
| | - Jae Jun Lee
- Institute of New Frontier ResearchHallym University College of MedicineChuncheonKorea
- Division of Big Data and Artificial IntelligenceChuncheon Sacred Heart HospitalChuncheonKorea
- Department of Anesthesiology and Pain MedicineHallym University College of MedicineChuncheonKorea
| |
Collapse
|
22
|
Zhao SQ, Liu WT. Progress in artificial intelligence assisted digestive endoscopy diagnosis of digestive system diseases. WORLD CHINESE JOURNAL OF DIGESTOLOGY 2024; 32:171-181. [DOI: 10.11569/wcjd.v32.i3.171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/29/2024]
|
23
|
Rugge M, Genta RM, Malfertheiner P, Dinis-Ribeiro M, El-Serag H, Graham DY, Kuipers EJ, Leung WK, Park JY, Rokkas T, Schulz C, El-Omar EM. RE.GA.IN.: the Real-world Gastritis Initiative-updating the updates. Gut 2024; 73:407-441. [PMID: 38383142 DOI: 10.1136/gutjnl-2023-331164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Accepted: 12/18/2023] [Indexed: 02/23/2024]
Abstract
At the end of the last century, a far-sighted 'working party' held in Sydney, Australia addressed the clinicopathological issues related to gastric inflammatory diseases. A few years later, an international conference held in Houston, Texas, USA critically updated the seminal Sydney classification. In line with these initiatives, Kyoto Global Consensus Report, flanked by the Maastricht-Florence conferences, added new clinical evidence to the gastritis clinicopathological puzzle.The most relevant topics related to the gastric inflammatory diseases have been addressed by the Real-world Gastritis Initiative (RE.GA.IN.), from disease definitions to the clinical diagnosis and prognosis. This paper reports the conclusions of the RE.GA.IN. consensus process, which culminated in Venice in November 2022 after more than 8 months of intense global scientific deliberations. A forum of gastritis scholars from five continents participated in the multidisciplinary RE.GA.IN. consensus. After lively debates on the most controversial aspects of the gastritis spectrum, the RE.GA.IN. Faculty amalgamated complementary knowledge to distil patient-centred, evidence-based statements to assist health professionals in their real-world clinical practice. The sections of this report focus on: the epidemiology of gastritis; Helicobacter pylori as dominant aetiology of environmental gastritis and as the most important determinant of the gastric oncogenetic field; the evolving knowledge on gastric autoimmunity; the clinicopathological relevance of gastric microbiota; the new diagnostic horizons of endoscopy; and the clinical priority of histologically reporting gastritis in terms of staging. The ultimate goal of RE.GA.IN. was and remains the promotion of further improvement in the clinical management of patients with gastritis.
Collapse
Affiliation(s)
- Massimo Rugge
- Department of Medicine-DIMED, University of Padova, Padua, Italy
- Azienda Zero, Veneto Tumour Registry, Padua, Italy
| | - Robert M Genta
- Gastrointestinal Pathology, Inform Diagnostics Research Institute, Dallas, Texas, USA
- Pathology, Baylor College of Medicine, Houston, Texas, USA
| | - Peter Malfertheiner
- Medizinische Klinik und Poliklinik II, Ludwig Maximilian Universität Klinikum München, Munich, Germany
- Klinik für Gastroenterologie, Hepatologie und Infektiologie, Otto-von-Guericke Universität Magdeburg, Magdeburg, Germany
| | - Mario Dinis-Ribeiro
- Porto Comprehensive Cancer Center & RISE@CI-IPO, University of Porto, Porto, Portugal
- Gastroenterology Department, Portuguese Institute of Oncology of Porto, Porto, Portugal
| | - Hashem El-Serag
- Gastroenterology and Hepatology, Baylor College of Medicine, Houston, Texas, USA
- Houston VA Health Services Research & Development Center of Excellence, Michael E DeBakey Veterans Affairs Medical Center, Houston, Texas, USA
| | - David Y Graham
- Department of Medicine, Michael E DeBakey Veterans Affairs Medical Center, Houston, Texas, USA
| | - Ernst J Kuipers
- Erasmus University Medical Center, Rotterdam, The Netherlands
| | | | - Jin Young Park
- International Agency for Research on Cancer, Lyon, France
| | - Theodore Rokkas
- Gastroenterology, Henry Dunant Hospital Center, Athens, Greece
| | | | - Emad M El-Omar
- Microbiome Research Centre, University of New South Wales, Sydney, New South Wales, Australia
| |
Collapse
|
24
|
Li X, Wu Q, Wang M, Wu K. Uncertainty-aware network for fine-grained and imbalanced reflux esophagitis grading. Comput Biol Med 2024; 168:107751. [PMID: 38016373 DOI: 10.1016/j.compbiomed.2023.107751] [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: 07/15/2023] [Revised: 10/22/2023] [Accepted: 11/20/2023] [Indexed: 11/30/2023]
Abstract
Computer-aided diagnosis (CAD) assists endoscopists in analyzing endoscopic images, reducing misdiagnosis rates and enabling timely treatment. A few studies have focused on CAD for gastroesophageal reflux disease, but CAD studies on reflux esophagitis (RE) are still inadequate. This paper presents a CAD study on RE using a dataset collected from hospital, comprising over 3000 images. We propose an uncertainty-aware network with handcrafted features, utilizing representation and classifier decoupling with metric learning to address class imbalance and achieve fine-grained RE classification. To enhance interpretability, the network estimates uncertainty through test time augmentation. The experimental results demonstrate that the proposed network surpasses previous methods, achieving an accuracy of 90.2% and an F1 score of 90.1%.
Collapse
Affiliation(s)
- Xingcun Li
- School of Management, Huazhong University of Science and Technology, Wuhan, 430074, China.
| | - Qinghua Wu
- School of Management, Huazhong University of Science and Technology, Wuhan, 430074, China.
| | - Mi Wang
- Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| | - Kun Wu
- Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| |
Collapse
|
25
|
Zhao Q, Jia Q, Chi T. U-Net deep learning model for endoscopic diagnosis of chronic atrophic gastritis and operative link for gastritis assessment staging: a prospective nested case-control study. Therap Adv Gastroenterol 2023; 16:17562848231208669. [PMID: 37928896 PMCID: PMC10624012 DOI: 10.1177/17562848231208669] [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: 01/15/2023] [Accepted: 10/02/2023] [Indexed: 11/07/2023] Open
Abstract
Background The operative link for the gastritis assessment (OLGA) system can objectively reflect the stratification of gastric cancer risk in patients with chronic atrophic gastritis (CAG). Objectives We developed a real-time video monitoring model for the endoscopic diagnosis of CAG and OLGA staging based on U-Net deep learning (DL). To further validate and improve its performance, we designed a study to evaluate the diagnostic evaluation indices. Design A prospective nested case-control study. Methods Our cohort consisted of 1306 patients from 31 July 2021 to 31 January 2022. According to the pathological results, patients in the cohort were divided into the CAG group and the chronic non-atrophic gastritis group to evaluate the diagnostic evaluation indices. Each atrophy lesion was automatically labeled and the atrophy severity was assessed by the model. Propensity score matching was used to minimize selection bias. Results The diagnostic evaluation indices and the consistency between OLGA staging and pathological diagnosis of the model were superior to those of endoscopists [sensitivity (89.31% versus 67.56%), specificity (90.46% versus 70.23%), positive predictive value (90.35% versus 69.41%), negative predictive value (89.43% versus 68.40%), accuracy rate (89.89% versus 68.89%), Youden index (79.77% versus 37.79%), odd product (79.23 versus 4.91), positive likelihood ratio (9.36 versus 2.27), negative likelihood ratio (0.12 versus 0.46)], areas under the curves (AUC) (95% CI) (0.919 (0.893-0.945) versus 0.749 (0.707-0.792), p < 0.001) and kappa (0.816 versus 0.291)]. Conclusion Our study demonstrated that the DL model can assist endoscopists in real-time diagnosis of CAG during gastroscopy and synchronous identification of high-risk OLGA stage (OLGA stages III and IV) patients. Trial registration ChiCTR2100044458.
Collapse
Affiliation(s)
- Quchuan Zhao
- Department of Gastroenterology, Xuanwu Hospital of Capital Medical University, Beijing, China
| | - Qing Jia
- Department of Anesthesiology, Guang’anmen Hospital China Academy of Chinese Medical Sciences, 5 North Court Street, Beijing 100053, China
| | - Tianyu Chi
- Department of Gastroenterology, Xuanwu Hospital of Capital Medical University, 45 Chang-Chun Street, Beijing 100053, China
| |
Collapse
|
26
|
Gong EJ, Bang CS, Lee JJ, Jeong HM, Baik GH, Jeong JH, Dick S, Lee GH. Clinical Decision Support System for All Stages of Gastric Carcinogenesis in Real-Time Endoscopy: Model Establishment and Validation Study. J Med Internet Res 2023; 25:e50448. [PMID: 37902818 PMCID: PMC10644184 DOI: 10.2196/50448] [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: 06/30/2023] [Revised: 07/27/2023] [Accepted: 10/12/2023] [Indexed: 10/31/2023] Open
Abstract
BACKGROUND Our research group previously established a deep-learning-based clinical decision support system (CDSS) for real-time endoscopy-based detection and classification of gastric neoplasms. However, preneoplastic conditions, such as atrophy and intestinal metaplasia (IM) were not taken into account, and there is no established model that classifies all stages of gastric carcinogenesis. OBJECTIVE This study aims to build and validate a CDSS for real-time endoscopy for all stages of gastric carcinogenesis, including atrophy and IM. METHODS A total of 11,868 endoscopic images were used for training and internal testing. The primary outcomes were lesion classification accuracy (6 classes: advanced gastric cancer, early gastric cancer, dysplasia, atrophy, IM, and normal) and atrophy and IM lesion segmentation rates for the segmentation model. The following tests were carried out to validate the performance of lesion classification accuracy: (1) external testing using 1282 images from another institution and (2) evaluation of the classification accuracy of atrophy and IM in real-world procedures in a prospective manner. To estimate the clinical utility, 2 experienced endoscopists were invited to perform a blind test with the same data set. A CDSS was constructed by combining the established 6-class lesion classification model and the preneoplastic lesion segmentation model with the previously established lesion detection model. RESULTS The overall lesion classification accuracy (95% CI) was 90.3% (89%-91.6%) in the internal test. For the performance validation, the CDSS achieved 85.3% (83.4%-97.2%) overall accuracy. The per-class external test accuracies for atrophy and IM were 95.3% (92.6%-98%) and 89.3% (85.4%-93.2%), respectively. CDSS-assisted endoscopy showed an accuracy of 92.1% (88.8%-95.4%) for atrophy and 95.5% (92%-99%) for IM in the real-world application of 522 consecutive screening endoscopies. There was no significant difference in the overall accuracy between the invited endoscopists and established CDSS in the prospective real-clinic evaluation (P=.23). The CDSS demonstrated a segmentation rate of 93.4% (95% CI 92.4%-94.4%) for atrophy or IM lesion segmentation in the internal testing. CONCLUSIONS The CDSS achieved high performance in terms of computer-aided diagnosis of all stages of gastric carcinogenesis and demonstrated real-world application potential.
Collapse
Affiliation(s)
- Eun Jeong Gong
- Department of Internal Medicine, Hallym University College of Medicine, Chuncheon, Republic of Korea
- Institute for Liver and Digestive Diseases, Hallym University, Chuncheon, Republic of Korea
- Institute of New Frontier Research, Hallym University College of Medicine, Chuncheon, Republic of Korea
| | - Chang Seok Bang
- Department of Internal Medicine, Hallym University College of Medicine, Chuncheon, Republic of Korea
- Institute for Liver and Digestive Diseases, Hallym University, Chuncheon, Republic of Korea
- Institute of New Frontier Research, Hallym University College of Medicine, Chuncheon, Republic of Korea
| | - Jae Jun Lee
- Institute of New Frontier Research, Hallym University College of Medicine, Chuncheon, Republic of Korea
- Department of Anesthesiology, Hallym University College of Medicine, Chuncheon, Republic of Korea
| | - Hae Min Jeong
- Department of Internal Medicine, Hallym University College of Medicine, Chuncheon, Republic of Korea
| | - Gwang Ho Baik
- Department of Internal Medicine, Hallym University College of Medicine, Chuncheon, Republic of Korea
- Institute for Liver and Digestive Diseases, Hallym University, Chuncheon, Republic of Korea
| | | | | | | |
Collapse
|
27
|
Wang L, Yang Y, Yang A, Li T. Lightweight deep learning model incorporating an attention mechanism and feature fusion for automatic classification of gastric lesions in gastroscopic images. BIOMEDICAL OPTICS EXPRESS 2023; 14:4677-4695. [PMID: 37791283 PMCID: PMC10545198 DOI: 10.1364/boe.487456] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 06/11/2023] [Accepted: 06/29/2023] [Indexed: 10/05/2023]
Abstract
Accurate diagnosis of various lesions in the formation stage of gastric cancer is an important problem for doctors. Automatic diagnosis tools based on deep learning can help doctors improve the accuracy of gastric lesion diagnosis. Most of the existing deep learning-based methods have been used to detect a limited number of lesions in the formation stage of gastric cancer, and the classification accuracy needs to be improved. To this end, this study proposed an attention mechanism feature fusion deep learning model with only 14 million (M) parameters. Based on that model, the automatic classification of a wide range of lesions covering the stage of gastric cancer formation was investigated, including non-neoplasm(including gastritis and intestinal metaplasia), low-grade intraepithelial neoplasia, and early gastric cancer (including high-grade intraepithelial neoplasia and early gastric cancer). 4455 magnification endoscopy with narrow-band imaging(ME-NBI) images from 1188 patients were collected to train and test the proposed method. The results of the test dataset showed that compared with the advanced gastric lesions classification method with the best performance (overall accuracy = 94.3%, parameters = 23.9 M), the proposed method achieved both higher overall accuracy and a relatively lightweight model (overall accuracy =95.6%, parameter = 14 M). The accuracy, sensitivity, and specificity of low-grade intraepithelial neoplasia were 94.5%, 93.0%, and 96.5%, respectively, achieving state-of-the-art classification performance. In conclusion, our method has demonstrated its potential in diagnosing various lesions at the stage of gastric cancer formation.
Collapse
Affiliation(s)
- Lingxiao Wang
- Institute of Biomedical Engineering, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300192, China
| | - Yingyun Yang
- Department of Gastroenterology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, China
| | - Aiming Yang
- Department of Gastroenterology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, China
| | - Ting Li
- Institute of Biomedical Engineering, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300192, China
| |
Collapse
|
28
|
Martins ML, Pedroso M, Libanio D, Dinis-Ribeiro M, Coimbra M, Renna F. Diagnostic Performance of Deep Learning Models for Gastric Intestinal Metaplasia Detection in Narrow-band Images. ANNUAL INTERNATIONAL CONFERENCE OF THE IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. ANNUAL INTERNATIONAL CONFERENCE 2023; 2023:1-4. [PMID: 38083501 DOI: 10.1109/embc40787.2023.10340055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2023]
Abstract
Gastric Intestinal Metaplasia (GIM) is one of the precancerous conditions in the gastric carcinogenesis cascade and its optical diagnosis during endoscopic screening is challenging even for seasoned endoscopists. Several solutions leveraging pre-trained deep neural networks (DNNs) have been recently proposed in order to assist human diagnosis. In this paper, we present a comparative study of these architectures in a new dataset containing GIM and non-GIM Narrow-band imaging still frames. We find that the surveyed DNNs perform remarkably well on average, but still measure sizeable inter-fold variability during cross-validation. An additional ad-hoc analysis suggests that these baseline architectures may not perform equally well at all scales when diagnosing GIM.Clinical relevance- Enhanching a clinician's ability to detect and localize intestinal metaplasia can be a crucial tool for gastric cancer management policies.
Collapse
|
29
|
Shi Y, Wei N, Wang K, Tao T, Yu F, Lv B. Diagnostic value of artificial intelligence-assisted endoscopy for chronic atrophic gastritis: a systematic review and meta-analysis. Front Med (Lausanne) 2023; 10:1134980. [PMID: 37200961 PMCID: PMC10185804 DOI: 10.3389/fmed.2023.1134980] [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: 12/31/2022] [Accepted: 04/10/2023] [Indexed: 05/20/2023] Open
Abstract
Background and aims The diagnosis of chronic atrophic gastritis (CAG) under normal white-light endoscopy depends on the endoscopist's experience and is not ideal. Artificial intelligence (AI) is increasingly used to diagnose diseases with good results. This review aimed to evaluate the accuracy of AI-assisted diagnosis of CAG through a meta-analysis. Methods We conducted a comprehensive literature search of four databases: PubMed, Embase, Web of Science, and the Cochrane Library. Studies published by November 21, 2022, on AI diagnosis CAG with endoscopic images or videos were included. We assessed the diagnostic performance of AI using meta-analysis, explored the sources of heterogeneity through subgroup analysis and meta-regression, and compared the accuracy of AI and endoscopists in diagnosing CAG. Results Eight studies that included a total of 25,216 patients of interest, 84,678 image training set images, and 10,937 test set images/videos were included. The results of the meta-analysis showed that the sensitivity of AI in identifying CAG was 94% (95% confidence interval [CI]: 0.88-0.97, I2 = 96.2%), the specificity was 96% (95% CI: 0.88-0.98, I2 = 98.04%), and the area under the summary receiver operating characteristic curve was 0.98 (95% CI: 0.96-0.99). The accuracy of AI in diagnosing CAG was significantly higher than that of endoscopists. Conclusions AI-assisted diagnosis of CAG in endoscopy has high accuracy and clinical diagnostic value. Systematic review registration http://www.crd.york.ac.uk/PROSPERO/, identifier: CRD42023391853.
Collapse
Affiliation(s)
- Yanting Shi
- Department of Gastroenterology, Zibo Central Hospital, Zibo, Shandong, China
| | - Ning Wei
- Department of Gastroenterology, Zibo Central Hospital, Zibo, Shandong, China
| | - Kunhong Wang
- Department of Gastroenterology, Zibo Central Hospital, Zibo, Shandong, China
| | - Tao Tao
- Department of Gastroenterology, Zibo Central Hospital, Zibo, Shandong, China
| | - Feng Yu
- Department of Gastroenterology, Zibo Central Hospital, Zibo, Shandong, China
- Feng Yu
| | - Bing Lv
- School of Computer Science and Technology, Shandong University of Technology, Zibo, Shandong, China
- *Correspondence: Bing Lv
| |
Collapse
|
30
|
Ali S. Where do we stand in AI for endoscopic image analysis? Deciphering gaps and future directions. NPJ Digit Med 2022; 5:184. [PMID: 36539473 PMCID: PMC9767933 DOI: 10.1038/s41746-022-00733-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Accepted: 11/29/2022] [Indexed: 12/24/2022] Open
Abstract
Recent developments in deep learning have enabled data-driven algorithms that can reach human-level performance and beyond. The development and deployment of medical image analysis methods have several challenges, including data heterogeneity due to population diversity and different device manufacturers. In addition, more input from experts is required for a reliable method development process. While the exponential growth in clinical imaging data has enabled deep learning to flourish, data heterogeneity, multi-modality, and rare or inconspicuous disease cases still need to be explored. Endoscopy being highly operator-dependent with grim clinical outcomes in some disease cases, reliable and accurate automated system guidance can improve patient care. Most designed methods must be more generalisable to the unseen target data, patient population variability, and variable disease appearances. The paper reviews recent works on endoscopic image analysis with artificial intelligence (AI) and emphasises the current unmatched needs in this field. Finally, it outlines the future directions for clinically relevant complex AI solutions to improve patient outcomes.
Collapse
Affiliation(s)
- Sharib Ali
- School of Computing, University of Leeds, LS2 9JT, Leeds, UK.
| |
Collapse
|
31
|
Ochiai K, Ozawa T, Shibata J, Ishihara S, Tada T. Current Status of Artificial Intelligence-Based Computer-Assisted Diagnosis Systems for Gastric Cancer in Endoscopy. Diagnostics (Basel) 2022; 12:diagnostics12123153. [PMID: 36553160 PMCID: PMC9777622 DOI: 10.3390/diagnostics12123153] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 12/07/2022] [Accepted: 12/10/2022] [Indexed: 12/15/2022] Open
Abstract
Artificial intelligence (AI) is gradually being utilized in various fields as its performance has been improving with the development of deep learning methods, availability of big data, and the progression of computer processing units. In the field of medicine, AI is mainly implemented in image recognition, such as in radiographic and pathologic diagnoses. In the realm of gastrointestinal endoscopy, although AI-based computer-assisted detection/diagnosis (CAD) systems have been applied in some areas, such as colorectal polyp detection and diagnosis, so far, their implementation in real-world clinical settings is limited. The accurate detection or diagnosis of gastric cancer (GC) is one of the challenges in which performance varies greatly depending on the endoscopist's skill. The diagnosis of early GC is especially challenging, partly because early GC mimics atrophic gastritis in the background mucosa. Therefore, several CAD systems for GC are being actively developed. The development of a CAD system for GC is considered challenging because it requires a large number of GC images. In particular, early stage GC images are rarely available, partly because it is difficult to diagnose gastric cancer during the early stages. Additionally, the training image data should be of a sufficiently high quality to conduct proper CAD training. Recently, several AI systems for GC that exhibit a robust performance, owing to being trained on a large number of high-quality images, have been reported. This review outlines the current status and prospects of AI use in esophagogastroduodenoscopy (EGDS), focusing on the diagnosis of GC.
Collapse
Affiliation(s)
- Kentaro Ochiai
- Department of Surgical Oncology, Faculty of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Tsuyoshi Ozawa
- Department of Surgical Oncology, Faculty of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo 113-0033, Japan
- Tomohiro Tada the Institute of Gastroenterology and Proctology, Musashi-Urawa, Saitama 336-0021, Japan
- AI Medical Service Inc. Toshima-ku, Tokyo 104-0061, Japan
| | - Junichi Shibata
- Tomohiro Tada the Institute of Gastroenterology and Proctology, Musashi-Urawa, Saitama 336-0021, Japan
- AI Medical Service Inc. Toshima-ku, Tokyo 104-0061, Japan
| | - Soichiro Ishihara
- Department of Surgical Oncology, Faculty of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Tomohiro Tada
- Department of Surgical Oncology, Faculty of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo 113-0033, Japan
- Tomohiro Tada the Institute of Gastroenterology and Proctology, Musashi-Urawa, Saitama 336-0021, Japan
- AI Medical Service Inc. Toshima-ku, Tokyo 104-0061, Japan
| |
Collapse
|
32
|
Luo Q, Yang H, Hu B. Application of artificial intelligence in the endoscopic diagnosis of early gastric cancer, atrophic gastritis, and Helicobacter pylori infection. J Dig Dis 2022; 23:666-674. [PMID: 36661411 DOI: 10.1111/1751-2980.13154] [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: 08/12/2022] [Revised: 01/04/2023] [Accepted: 01/17/2023] [Indexed: 01/21/2023]
Abstract
Gastric cancer (GC) is one of the most serious health problems worldwide. Chronic atrophic gastritis (CAG) is most commonly caused by Helicobacter pylori (H. pylori) infection. Currently, endoscopic detection of early gastric cancer (EGC) and CAG remains challenging for endoscopists, and the diagnostic accuracy of H. pylori infection by endoscopy is approximately 70%. Artificial intelligence (AI) can assist endoscopic diagnosis including detection, prediction of depth of invasion, boundary delineation, and anatomical location of EGC, and has achievable diagnostic ability even comparable to experienced endoscopists. In this review we summarized various AI-assisted systems in the diagnosis of EGC, CAG, and H. pylori infection.
Collapse
Affiliation(s)
- Qi Luo
- Department of Gastroenterology, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Hang Yang
- Department of Gastroenterology, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Bing Hu
- Department of Gastroenterology, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| |
Collapse
|
33
|
Chong Y, Xie N, Liu X, Zhang M, Huang F, Fang J, Wang F, Pan S, Nie H, Zhao Q. A deep learning network based on multi-scale and attention for the diagnosis of chronic atrophic gastritis. ZEITSCHRIFT FUR GASTROENTEROLOGIE 2022; 60:1770-1778. [PMID: 35697062 DOI: 10.1055/a-1828-1441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
BACKGROUND AND STUDY AIM Chronic atrophic gastritis plays an important role in the process of gastric cancer. Deep learning is gradually introduced in the medical field, and how to better apply a convolutional neural network (CNN) to the diagnosis of chronic atrophic gastritis remains a research hotspot. This study was designed to improve the performance of CNN on diagnosing chronic atrophic gastritis by constructing and evaluating a network structure based on the characteristics of gastroscopic images. METHODS Three endoscopists reviewed the endoscopic images of the gastric antrum from the Gastroscopy Image Database of Zhongnan Hospital and labelled available images according to pathological results. Two novel modules proposed recently were introduced to construct the Multi-scale with Attention net (MWA-net) considering the characters of similar medical images. After training the network using images of training sets, the diagnostic ability of the MWA-net was evaluated by comparing it with those of other deep learning models and endoscopists with varying degrees of expertise. RESULTS As a result, 5,159 images of the gastric antrum from 2,240 patients were used to train and test the MWA-net. Compared with the direct application of famous networks, the MWA-net achieved the best performance (accuracy, 92.13%) with an increase of 1.80% compared to that of ResNet. The suspicious lesions indicated by the network are consistent with the conclusion of experts. The sensitivity and specificity of the convolutional network for gastric atrophy diagnosis are 90.19% and 94.51%, respectively, which are higher than those of experts. CONCLUSIONS Highly similar images of chronic atrophic gastritis can be identified by the proposed MWA-net, which has a better performance than other well-known networks. This work can further reduce the workload of gastroscopists, simplify the diagnostic process and provide medical assistance to more residents.
Collapse
Affiliation(s)
| | | | - Xin Liu
- Wuhan University, Wuhan City, China
| | - Meng Zhang
- Wuhan University Zhongnan Hospital, Wuhan, China
| | | | - Jun Fang
- Wuhan University Zhongnan Hospital, Wuhan, China
| | - Fan Wang
- Wuhan University Zhongnan Hospital, Wuhan, China
| | | | - Haihang Nie
- Wuhan University Zhongnan Hospital, Wuhan, China
| | - Qiu Zhao
- Wuhan University, Wuhan City, China
| |
Collapse
|
34
|
Dilaghi E, Lahner E, Annibale B, Esposito G. Systematic review and meta-analysis: Artificial intelligence for the diagnosis of gastric precancerous lesions and Helicobacter pylori infection. Dig Liver Dis 2022; 54:1630-1638. [PMID: 35382973 DOI: 10.1016/j.dld.2022.03.007] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Revised: 03/12/2022] [Accepted: 03/16/2022] [Indexed: 02/08/2023]
Abstract
BACKGROUND The endoscopic diagnosis of Helicobacter-pylori(H.pylori) infection and gastric precancerous lesions(GPL), namely atrophic-gastritis and intestinal-metaplasia, still remains challenging. Artificial intelligence(AI) may represent a powerful resource for the endoscopic recognition of these conditions. AIMS To explore the diagnostic performance(DP) of AI in the diagnosis of GPL and H.pylori infection. METHODS A systematic-review was performed by two independent authors up to September 2021. Inclusion criteria were studies focusing on the DP of AI-system in the diagnosis of GPL and H.pylori infection. The pooled accuracy of studies included was reported. RESULTS Overall, 128 studies were found (PubMed-Embase-Cochrane Library) and four and nine studies were finally included regarding GPL and H.pylori infection, respectively. The pooled-accuracy(random effects model) was 90.3%(95%CI 84.3-94.9) and 79.6%(95%CI 66.7-90.0) with a significant heterogeneity[I2=90.4%(95%CI 78.5-95.7);I2=97.9%(97.2-98.6)] for GPL and H.pylori infection, respectively. The Begg's-test showed a significant publication-bias(p = 0.0371) only among studies regarding H.pylori infection. The pooled-accuracy(random-effects-model) was similar considering only studies using CNN-model for the diagnosis of H.pylori infection: 74.1%[(95%CI 51.6-91.3);I2=98.9%(95%CI 98.5-99.3)], Begg's-test(p = 0.1416) did not show publication-bias. CONCLUSION AI-system seems to be a good resource for an easier diagnosis of GPL and H.pylori infection, showing a pooled-diagnostic-accuracy of 90% and 80%, respectively. However, considering the high heterogeneity, these promising data need an external validation by randomized control trials and prospective real-time studies.
Collapse
Affiliation(s)
- E Dilaghi
- Department of Medical-Surgical Sciences and Translational Medicine, Sant'Andrea Hospital, Sapienza University of Rome, Via di Grottarossa, Roma 1035 - 00189, Italy
| | - E Lahner
- Department of Medical-Surgical Sciences and Translational Medicine, Sant'Andrea Hospital, Sapienza University of Rome, Via di Grottarossa, Roma 1035 - 00189, Italy
| | - B Annibale
- Department of Medical-Surgical Sciences and Translational Medicine, Sant'Andrea Hospital, Sapienza University of Rome, Via di Grottarossa, Roma 1035 - 00189, Italy
| | - G Esposito
- Department of Medical-Surgical Sciences and Translational Medicine, Sant'Andrea Hospital, Sapienza University of Rome, Via di Grottarossa, Roma 1035 - 00189, Italy.
| |
Collapse
|
35
|
Cao R, Tang L, Fang M, Zhong L, Wang S, Gong L, Li J, Dong D, Tian J. Artificial intelligence in gastric cancer: applications and challenges. Gastroenterol Rep (Oxf) 2022; 10:goac064. [PMID: 36457374 PMCID: PMC9707405 DOI: 10.1093/gastro/goac064] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Revised: 09/27/2022] [Accepted: 10/18/2022] [Indexed: 08/10/2023] Open
Abstract
Gastric cancer (GC) is one of the most common malignant tumors with high mortality. Accurate diagnosis and treatment decisions for GC rely heavily on human experts' careful judgments on medical images. However, the improvement of the accuracy is hindered by imaging conditions, limited experience, objective criteria, and inter-observer discrepancies. Recently, the developments of machine learning, especially deep-learning algorithms, have been facilitating computers to extract more information from data automatically. Researchers are exploring the far-reaching applications of artificial intelligence (AI) in various clinical practices, including GC. Herein, we aim to provide a broad framework to summarize current research on AI in GC. In the screening of GC, AI can identify precancerous diseases and assist in early cancer detection with endoscopic examination and pathological confirmation. In the diagnosis of GC, AI can support tumor-node-metastasis (TNM) staging and subtype classification. For treatment decisions, AI can help with surgical margin determination and prognosis prediction. Meanwhile, current approaches are challenged by data scarcity and poor interpretability. To tackle these problems, more regulated data, unified processing procedures, and advanced algorithms are urgently needed to build more accurate and robust AI models for GC.
Collapse
Affiliation(s)
| | | | - Mengjie Fang
- School of Artificial Intelligence, University of Chinese Academy of Sciences, Beijing, P. R. China
- CAS Key Laboratory of Molecular Imaging, Beijing Key Laboratory of Molecular Imaging, the State Key Laboratory of Management and Control for Complex Systems, Institute of Automation, Chinese Academy of Sciences, Beijing, P. R. China
- Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, School of Engineering Medicine, Beihang University, Beijing, P. R. China
| | - Lianzhen Zhong
- School of Artificial Intelligence, University of Chinese Academy of Sciences, Beijing, P. R. China
- CAS Key Laboratory of Molecular Imaging, Beijing Key Laboratory of Molecular Imaging, the State Key Laboratory of Management and Control for Complex Systems, Institute of Automation, Chinese Academy of Sciences, Beijing, P. R. China
| | - Siwen Wang
- School of Artificial Intelligence, University of Chinese Academy of Sciences, Beijing, P. R. China
- CAS Key Laboratory of Molecular Imaging, Beijing Key Laboratory of Molecular Imaging, the State Key Laboratory of Management and Control for Complex Systems, Institute of Automation, Chinese Academy of Sciences, Beijing, P. R. China
| | - Lixin Gong
- College of Medicine and Biological Information Engineering School, Northeastern University, Shenyang, Liaoning, P. R. China
| | - Jiazheng Li
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Radiology Department, Peking University Cancer Hospital & Institute, Beijing, P. R. China
| | - Di Dong
- Corresponding authors. Di Dong, CAS Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, 95 Zhongguancun East Road, Beijing 100190, P. R. China. Tel: +86-13811833760; ; Jie Tian, Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, School of Engineering Medicine, Beihang University, Beijing, P. R. China. Tel: +86-10-82618465;
| | - Jie Tian
- Corresponding authors. Di Dong, CAS Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, 95 Zhongguancun East Road, Beijing 100190, P. R. China. Tel: +86-13811833760; ; Jie Tian, Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, School of Engineering Medicine, Beihang University, Beijing, P. R. China. Tel: +86-10-82618465;
| |
Collapse
|
36
|
A deep learning method to assist with chronic atrophic gastritis diagnosis using white light images. Dig Liver Dis 2022; 54:1513-1519. [PMID: 35610166 DOI: 10.1016/j.dld.2022.04.025] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 03/29/2022] [Accepted: 04/20/2022] [Indexed: 12/30/2022]
Abstract
BACKGROUND Chronic atrophic gastritis is a common preneoplastic condition of the stomach with a low detection rate during endoscopy. AIMS This study aimed to develop two deep learning models to improve the diagnostic rate. METHODS We collected 10,593 images from 4005 patients including 2280 patients with chronic atrophic gastritis and 1725 patients with chronic non-atrophic gastritis from two tertiary hospitals. Two deep learning models were developed to detect chronic atrophic gastritis using ResNet50. The detection ability of the deep learning model was compared with that of three expert endoscopists. RESULTS In the external test set, the diagnostic accuracy of model 1 for detecting gastric antrum atrophy was 0.890. The identification accuracies for the severity of gastric antrum atrophy were 0.773 and 0.590 in the internal and external test sets, respectively. In the other two external sets, the detection accuracies of model 2 for chronic atrophic gastritis were 0.854 and 0.916, respectively. Deep learning model 1's ability to identify gastric antrum atrophy was comparable to that of human experts. CONCLUSION Deep-learning-based models can detect chronic atrophic gastritis with good performance, which may greatly reduce the burden on endoscopists, relieve patient suffering, and improve the disease's detection rate in primary hospitals.
Collapse
|
37
|
Yang H, Wu Y, Yang B, Wu M, Zhou J, Liu Q, Lin Y, Li S, Li X, Zhang J, Wang R, Xie Q, Li J, Luo Y, Tu M, Wang X, Lan H, Bai X, Wu H, Zeng F, Zhao H, Yi Z, Zeng F. Identification of upper GI diseases during screening gastroscopy using a deep convolutional neural network algorithm. Gastrointest Endosc 2022; 96:787-795.e6. [PMID: 35718070 DOI: 10.1016/j.gie.2022.06.011] [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] [Received: 01/30/2022] [Revised: 06/09/2022] [Accepted: 06/11/2022] [Indexed: 02/05/2023]
Abstract
BACKGROUND AND AIMS The clinical application of GI endoscopy for the diagnosis of multiple diseases using artificial intelligence (AI) has been limited by its high false-positive rates. There is an unmet need to develop a GI endoscopy AI-assisted diagnosis system (GEADS) to improve diagnostic accuracy and clinical utility. METHODS In this retrospective, multicenter study, a convolutional neural network was trained to assess upper GI diseases based on 26,228 endoscopic images from Dazhou Central Hospital that were randomly assigned (3:1:1) to a training dataset, validation dataset, and test dataset, respectively. To validate the model, 6 external independent datasets comprising 51,372 images of upper GI diseases were collected. In addition, 1 prospective dataset comprising 27,975 images was collected. The performance of GEADS was compared with endoscopists with 2 professional degrees of expertise: expert and novice. Eight endoscopists were in the expert group with >5 years of experience, whereas 3 endoscopists were in the novice group with 1 to 5 years of experience. RESULTS The GEADS model achieved an accuracy of .918 (95% confidence interval [CI], .914-.922), with an F1 score of .884 (95% CI, .879-.889), recall of .873 (95% CI, .868-.878), and precision of .890 (95% CI, .885-.895) in the internal validation dataset. In the external validation datasets and 1 prospective validation dataset, the diagnostic accuracy of the GEADS ranged from .841 (95% CI, .834-.848) to .949 (95% CI, .935-.963). With the help of the GEADS, the diagnosing accuracies of novice and expert endoscopists were significantly improved (P < .001). CONCLUSIONS The AI system can assist endoscopists in improving the accuracy of diagnosing upper GI diseases.
Collapse
Affiliation(s)
- Hang Yang
- Department of Clinical Research Center, Dazhou Central Hospital, Dazhou, Sichuan, China
| | - Yu Wu
- Center of Intelligent Medicine, Computer Science, Sichuan University, Chengdu, Sichuan, China
| | - Bo Yang
- Digestive Endoscopy Center, Dazhou Central Hospital, Dazhou, Sichuan, China
| | - Min Wu
- Huaxi MR Research Center (HMRRC), Department of Radiology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Jun Zhou
- Department of Clinical Research Center, Dazhou Central Hospital, Dazhou, Sichuan, China
| | - Qin Liu
- Department of Clinical Research Center, Dazhou Central Hospital, Dazhou, Sichuan, China
| | - Yifei Lin
- Precision Medicine Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Shilin Li
- Department of Clinical Research Center, Dazhou Central Hospital, Dazhou, Sichuan, China
| | - Xue Li
- Department of Clinical Research Center, Dazhou Central Hospital, Dazhou, Sichuan, China
| | - Jie Zhang
- Department of Clinical Research Center, Dazhou Central Hospital, Dazhou, Sichuan, China
| | - Rui Wang
- Department of Clinical Research Center, Dazhou Central Hospital, Dazhou, Sichuan, China
| | - Qianrong Xie
- Department of Clinical Research Center, Dazhou Central Hospital, Dazhou, Sichuan, China
| | - Jingqi Li
- College of Aulin, Northeast Forestry University, Harbin, Heilongjiang, China
| | - Yue Luo
- College of Basic Medical Sciences, North Sichuan Medical College, Nanchong, Sichuan, China
| | - Mengjie Tu
- Department of Clinical Research Center, Dazhou Central Hospital, Dazhou, Sichuan, China; Department of Surgery, Shantou University Medical College, Shantou, Guangdong, China
| | - Xiao Wang
- Digestive Endoscopy Center, Dazhou Central Hospital, Dazhou, Sichuan, China
| | - Haitao Lan
- Department of Sichuan, Academy of Medical Sciences, Sichuan Provincial People's Hospital, Chengdu, Sichuan, China
| | - Xuesong Bai
- Digestive Endoscopy Center, Dazhou Central Hospital, Dazhou, Sichuan, China
| | - Huaping Wu
- Department of Cardiac &Vascular Surgery, Dazhou Central Hospital, Dazhou, Sichuan, China
| | - Fanwei Zeng
- Department of Spinal Surgery, Sichuan Province Orthopedic Hospital, Chengdu, Sichuan, China
| | - Hong Zhao
- Department of Hepatobiliary Surgery, State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Zhang Yi
- Center of Intelligent Medicine, Computer Science, Sichuan University, Chengdu, Sichuan, China
| | - Fanxin Zeng
- Department of Clinical Research Center, Dazhou Central Hospital, Dazhou, Sichuan, China; Center of Intelligent Medicine, Computer Science, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
38
|
Ortiz Zúñiga O, Fernández Esparrach MG, Daca M, Pellisé M. Artificial intelligence in gastrointestinal endoscopy - Evolution to a new era. REVISTA ESPANOLA DE ENFERMEDADES DIGESTIVAS 2022; 114:605-615. [PMID: 35770604 DOI: 10.17235/reed.2022.8961/2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Artificial intelligence (AI) systems based on machine learning have evolved in the last few years with an increasing applicability in gastrointestinal endoscopy. Thanks to AI, an image (input) can be transformed into a clinical decision (output). Although AI systems have been initially studied to improve detection (CADe) and characterization of colorectal lesions (CADx), other indications are being currently investigated as detection of blind spots, scope guidance, or delineation/measurement of lesions. The objective of these review is to summarize the current evidence on applicability of AI systems in gastrointestinal endoscopy, highlight strengths and limitations of the technology and review regulatory and ethical aspects for its general implementation in gastrointestinal endoscopy.
Collapse
Affiliation(s)
| | | | - María Daca
- Gastroenterología, Hospital Clínic Barcelona, España
| | - María Pellisé
- Gastroenterología, Hospital Clínic Barcelona, España
| |
Collapse
|
39
|
Zhao Q, Jia Q, Chi T. Deep learning as a novel method for endoscopic diagnosis of chronic atrophic gastritis: a prospective nested case-control study. BMC Gastroenterol 2022; 22:352. [PMID: 35879649 PMCID: PMC9310473 DOI: 10.1186/s12876-022-02427-2] [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: 05/15/2022] [Accepted: 07/15/2022] [Indexed: 11/28/2022] Open
Abstract
Background and aims Chronic atrophic gastritis (CAG) is a precancerous disease that often leads to the development of gastric cancer (GC) and is positively correlated with GC morbidity. However, the sensitivity of the endoscopic diagnosis of CAG is only 42%. Therefore, we developed a real-time video monitoring model for endoscopic diagnosis of CAG based on U-Net deep learning (DL) and conducted a prospective nested case–control study to evaluate the diagnostic evaluation indices of the model and its consistency with pathological diagnosis.
Methods Our cohort consisted of 1539 patients undergoing gastroscopy from December 1, 2020, to July 1, 2021. Based on pathological diagnosis, patients in the cohort were divided into the CAG group or the chronic nonatrophic gastritis (CNAG) group, and we assessed the diagnostic evaluation indices of this model and its consistency with pathological diagnosis after propensity score matching (PSM) to minimize selection bias in the study. Results After matching, the diagnostic evaluation indices and consistency evaluation of the model were better than those of endoscopists [sensitivity (84.02% vs. 62.72%), specificity (97.04% vs. 81.95%), positive predictive value (96.60% vs. 77.66%), negative predictive value (85.86% vs. 68.73%), accuracy rate (90.53% vs. 72.34%), Youden index (81.06% vs. 44.67%), odd product (172.5 vs. 7.64), positive likelihood ratio (28.39 vs. 3.47), negative likelihood ratio (0.16 vs. 0.45), AUC (95% CI) [0.909 (0.884–0.934) vs. 0.740 (0.702–0.778)] and Kappa (0.852 vs. 0.558)]. Conclusions Our prospective nested case–control study proved that the diagnostic evaluation indices and consistency evaluation of the real-time video monitoring model for endoscopic diagnosis of CAG based on U-Net DL were superior to those of endoscopists. Trial registrationChiCTR2100044458, 18/03/2020.
Collapse
Affiliation(s)
- Quchuan Zhao
- Department of Gastroenterology, Xuanwu Hospital of Capital Medical University, 45 Chang-chun Street, Beijing, 100053, China
| | - Qing Jia
- Department of Anesthesiology, Guang'anmen Hospital China Academy of Chinese Medical Sciences, 5 North Court Street, Beijing, 100053, China.
| | - Tianyu Chi
- Department of Gastroenterology, Xuanwu Hospital of Capital Medical University, 45 Chang-chun Street, Beijing, 100053, China.
| |
Collapse
|
40
|
Gong Y, Kang J, Wu R, Ge F, Zheng YS, Zeng Q. Gastroscopic results for the asymptomatic, average-risk population in Northern China: a cross-sectional study of 60,519 adults. Scand J Gastroenterol 2022; 57:748-756. [PMID: 35132926 DOI: 10.1080/00365521.2022.2035810] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2021] [Revised: 01/19/2022] [Accepted: 01/26/2022] [Indexed: 02/08/2023]
Abstract
BACKGROUND Studies on average-risk individuals undergoing gastroscopy screening in China are scarce. OBJECTIVE To determine and compare the prevalence of lesions found by gastroscopy and the association between sex, age, Helicobacter pylori infection and gastric premalignant lesions. METHODS Gastroscopy results were analysed for 60,519 individuals enrolled from January 2013 to December 2019. RESULTS The median age was 49.84 years (SD, 9.47 years) for women and 48.90 years (SD, 8.82 years) for men, and the ratio of females to males was 35.10% (n = 21,240) to 64.90% (n = 39,279). The most common lesions detected by endoscopy were chronic gastritis, reflux oesophagitis, duodenitis and gastric polyps, detected in 24.48%, 10.28%, 3.96% and 3.61%, respectively. Oesophageal cancer and gastric cancer were detected in 0.33% and 0.47% of patients, respectively. The prevalence of chronic gastritis increased with age and was higher in males than in females (26.47% [n = 10396] versus 20.80% [n = 4417], p < .001). The prevalence of gastric ulcers was highest in the elderly group, and the H. pylori infection rate of gastric ulcer patients was 47.28%. The prevalence of gastric polyps was higher in females than in males (5.47% [n = 1161] versus 2.61% [n = 1024], p < .001), and the H. pylori infection rate in inflammatory polyp patients was higher than that in fundic gland polyp patients (28.32% [n = 442] versus 7.29% [n = 29], p < .001). CONCLUSION The prevalence of upper gastrointestinal endoscopic lesions is high in the asymptomatic population undergoing physical examination and is associated with sex, age, and H. pylori infection.
Collapse
Affiliation(s)
- Yan Gong
- Health Management Institute, The Second Medical Center & National Clinical Research Center for Geriatric Disease, Chinese PLA General Hospital, Beijing, China
| | - Juan Kang
- Department of Emergency, The Second Medical Center & National Clinical Research Center for Geriatric Disease, Chinese PLA General Hospital, Beijing, China
| | - Rilige Wu
- Medical Big Data Research Center, Medical Innovation Research Division of Chinese, PLA General Hospital, Beijing, China
| | - Fulin Ge
- Department of Gastroenterology, The Second Medical Center & National Clinical Research Center for Geriatric Disease, Chinese PLA General Hospital, Beijing, China
| | - Yan-Song Zheng
- Health Management Institute, The Second Medical Center & National Clinical Research Center for Geriatric Disease, Chinese PLA General Hospital, Beijing, China
| | - Qiang Zeng
- Health Management Institute, The Second Medical Center & National Clinical Research Center for Geriatric Disease, Chinese PLA General Hospital, Beijing, China
| |
Collapse
|
41
|
Renna F, Martins M, Neto A, Cunha A, Libânio D, Dinis-Ribeiro M, Coimbra M. Artificial Intelligence for Upper Gastrointestinal Endoscopy: A Roadmap from Technology Development to Clinical Practice. Diagnostics (Basel) 2022; 12:1278. [PMID: 35626433 PMCID: PMC9141387 DOI: 10.3390/diagnostics12051278] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 05/14/2022] [Accepted: 05/18/2022] [Indexed: 02/05/2023] Open
Abstract
Stomach cancer is the third deadliest type of cancer in the world (0.86 million deaths in 2017). In 2035, a 20% increase will be observed both in incidence and mortality due to demographic effects if no interventions are foreseen. Upper GI endoscopy (UGIE) plays a paramount role in early diagnosis and, therefore, improved survival rates. On the other hand, human and technical factors can contribute to misdiagnosis while performing UGIE. In this scenario, artificial intelligence (AI) has recently shown its potential in compensating for the pitfalls of UGIE, by leveraging deep learning architectures able to efficiently recognize endoscopic patterns from UGIE video data. This work presents a review of the current state-of-the-art algorithms in the application of AI to gastroscopy. It focuses specifically on the threefold tasks of assuring exam completeness (i.e., detecting the presence of blind spots) and assisting in the detection and characterization of clinical findings, both gastric precancerous conditions and neoplastic lesion changes. Early and promising results have already been obtained using well-known deep learning architectures for computer vision, but many algorithmic challenges remain in achieving the vision of AI-assisted UGIE. Future challenges in the roadmap for the effective integration of AI tools within the UGIE clinical practice are discussed, namely the adoption of more robust deep learning architectures and methods able to embed domain knowledge into image/video classifiers as well as the availability of large, annotated datasets.
Collapse
Affiliation(s)
- Francesco Renna
- Instituto de Engenharia de Sistemas e Computadores, Tecnologia e Ciência, 3200-465 Porto, Portugal; (M.M.); (A.N.); (A.C.); (M.C.)
- Faculdade de Ciências, Universidade do Porto, 4169-007 Porto, Portugal
| | - Miguel Martins
- Instituto de Engenharia de Sistemas e Computadores, Tecnologia e Ciência, 3200-465 Porto, Portugal; (M.M.); (A.N.); (A.C.); (M.C.)
- Faculdade de Ciências, Universidade do Porto, 4169-007 Porto, Portugal
| | - Alexandre Neto
- Instituto de Engenharia de Sistemas e Computadores, Tecnologia e Ciência, 3200-465 Porto, Portugal; (M.M.); (A.N.); (A.C.); (M.C.)
- Escola de Ciências e Tecnologia, Universidade de Trás-os-Montes e Alto Douro, Quinta de Prados, 5001-801 Vila Real, Portugal
| | - António Cunha
- Instituto de Engenharia de Sistemas e Computadores, Tecnologia e Ciência, 3200-465 Porto, Portugal; (M.M.); (A.N.); (A.C.); (M.C.)
- Escola de Ciências e Tecnologia, Universidade de Trás-os-Montes e Alto Douro, Quinta de Prados, 5001-801 Vila Real, Portugal
| | - Diogo Libânio
- Departamento de Ciências da Informação e da Decisão em Saúde/Centro de Investigação em Tecnologias e Serviços de Saúde (CIDES/CINTESIS), Faculdade de Medicina, Universidade do Porto, 4200-319 Porto, Portugal; (D.L.); (M.D.-R.)
| | - Mário Dinis-Ribeiro
- Departamento de Ciências da Informação e da Decisão em Saúde/Centro de Investigação em Tecnologias e Serviços de Saúde (CIDES/CINTESIS), Faculdade de Medicina, Universidade do Porto, 4200-319 Porto, Portugal; (D.L.); (M.D.-R.)
| | - Miguel Coimbra
- Instituto de Engenharia de Sistemas e Computadores, Tecnologia e Ciência, 3200-465 Porto, Portugal; (M.M.); (A.N.); (A.C.); (M.C.)
- Faculdade de Ciências, Universidade do Porto, 4169-007 Porto, Portugal
| |
Collapse
|
42
|
Yu T, Lin N, Zhong X, Zhang X, Zhang X, Chen Y, Liu J, Hu W, Duan H, Si J. Multi-label recognition of cancer-related lesions with clinical priors on white-light endoscopy. Comput Biol Med 2022; 143:105255. [PMID: 35151153 DOI: 10.1016/j.compbiomed.2022.105255] [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: 11/16/2021] [Revised: 01/07/2022] [Accepted: 01/20/2022] [Indexed: 11/28/2022]
Abstract
Deep learning-based computer-aided diagnosis techniques have demonstrated encouraging performance in endoscopic lesion identification and detection, and have reduced the rate of missed and false detections of disease during endoscopy. However, the interpretability of the model-based results has not been adequately addressed by existing methods. This phenomenon is directly manifested by a significant bias in the representation of feature localization. Good recognition models experience severe feature localization errors, particularly for lesions with subtle morphological features, and such unsatisfactory performance hinders the clinical deployment of models. To effectively alleviate this problem, we proposed a solution to optimize the localization bias in feature representations of cancer-related recognition models that is difficult to accurately label and identify in clinical practice. Optimization was performed in the training phase of the model through the proposed data augmentation method and auxiliary loss function based on clinical priors. The data augmentation method, called partial jigsaw, can "break" the spatial structure of lesion-independent image blocks and enrich the data feature space to decouple the interference of background features on the space and focus on fine-grained lesion features. The annotation-based auxiliary loss function used class activation maps for sample distribution correction and led the model to present localization representation converging on the gold standard annotation of visualization maps. The results show that with the improvement of our method, the precision of model recognition reached an average of 92.79%, an F1-score of 92.61%, and accuracy of 95.56% based on a dataset constructed from 23 hospitals. In addition, we quantified the evaluation representation of visualization feature maps. The improved model yielded significant offset correction results for visualized feature maps compared with the baseline model. The average visualization-weighted positive coverage improved from 51.85% to 83.76%. The proposed approach did not change the deployment capability and inference speed of the original model and can be incorporated into any state-of-the-art neural network. It also shows the potential to provide more accurate localization inference results and assist in clinical examinations during endoscopies.
Collapse
Affiliation(s)
- Tao Yu
- Key Laboratory for Biomedical Engineering of Ministry of Education, College of Biomedical Engineering and Instrument Science, Zhejiang University, Hangzhou, China
| | - Ne Lin
- Department of Gastroenterology, Sir Run Run Shaw Hospital, Medical School, Zhejiang University, Hangzhou, China
| | - Xingwei Zhong
- Department of Gastroenterology, Sir Run Run Shaw Hospital, Medical School, Zhejiang University, Hangzhou, China
| | - Xiaoyan Zhang
- Key Laboratory for Biomedical Engineering of Ministry of Education, College of Biomedical Engineering and Instrument Science, Zhejiang University, Hangzhou, China
| | - Xinsen Zhang
- Key Laboratory for Biomedical Engineering of Ministry of Education, College of Biomedical Engineering and Instrument Science, Zhejiang University, Hangzhou, China
| | - Yihe Chen
- Key Laboratory for Biomedical Engineering of Ministry of Education, College of Biomedical Engineering and Instrument Science, Zhejiang University, Hangzhou, China
| | - Jiquan Liu
- Key Laboratory for Biomedical Engineering of Ministry of Education, College of Biomedical Engineering and Instrument Science, Zhejiang University, Hangzhou, China.
| | - Weiling Hu
- Department of Gastroenterology, Sir Run Run Shaw Hospital, Medical School, Zhejiang University, Hangzhou, China; Institute of Gastroenterology, Zhejiang University, Hangzhou, China
| | - Huilong Duan
- Key Laboratory for Biomedical Engineering of Ministry of Education, College of Biomedical Engineering and Instrument Science, Zhejiang University, Hangzhou, China
| | - Jianmin Si
- Department of Gastroenterology, Sir Run Run Shaw Hospital, Medical School, Zhejiang University, Hangzhou, China; Institute of Gastroenterology, Zhejiang University, Hangzhou, China
| |
Collapse
|
43
|
Nagao S, Tani Y, Shibata J, Tsuji Y, Tada T, Ishihara R, Fujishiro M. Implementation of artificial intelligence in upper gastrointestinal endoscopy. DEN OPEN 2022; 2:e72. [PMID: 35873509 PMCID: PMC9302271 DOI: 10.1002/deo2.72] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 10/11/2021] [Accepted: 10/16/2021] [Indexed: 12/24/2022]
Abstract
The application of artificial intelligence (AI) using deep learning has significantly expanded in the field of esophagogastric endoscopy. Recent studies have shown promising results in detecting and differentiating early gastric cancer using AI tools built using white light, magnified, or image-enhanced endoscopic images. Some studies have reported the use of AI tools to predict the depth of early gastric cancer based on endoscopic images. Similarly, studies based on using AI for detecting early esophageal cancer have also been reported, with an accuracy comparable to that of endoscopy specialists. Moreover, an AI system, developed to diagnose pharyngeal cancer, has shown promising performance with high sensitivity. These reports suggest that, if introduced for regular use in clinical settings, AI systems can significantly reduce the burden on physicians. This review summarizes the current status of AI applications in the upper gastrointestinal tract and presents directions for clinical practice implementation and future research.
Collapse
Affiliation(s)
- Sayaka Nagao
- Department of GastroenterologyGraduate School of Medicinethe University of TokyoTokyoJapan
- Department of Endoscopy and Endoscopic SurgeryGraduate School of Medicinethe University of TokyoTokyoJapan
| | - Yasuhiro Tani
- Department of Gastrointestinal OncologyOsaka International Cancer InstituteOsakaJapan
| | - Junichi Shibata
- Tada Tomohiro Institute of Gastroenterology and ProctologySaitamaJapan
| | - Yosuke Tsuji
- Department of GastroenterologyGraduate School of Medicinethe University of TokyoTokyoJapan
| | - Tomohiro Tada
- Tada Tomohiro Institute of Gastroenterology and ProctologySaitamaJapan
- AI Medical Service Inc.TokyoJapan
- Department of Surgical OncologyGraduate School of Medicinethe University of TokyoTokyoJapan
| | - Ryu Ishihara
- Department of Gastrointestinal OncologyOsaka International Cancer InstituteOsakaJapan
| | - Mitsuhiro Fujishiro
- Department of GastroenterologyGraduate School of Medicinethe University of TokyoTokyoJapan
| |
Collapse
|
44
|
Zhao Q, Chi T. Deep learning model can improve the diagnosis rate of endoscopic chronic atrophic gastritis: a prospective cohort study. BMC Gastroenterol 2022; 22:133. [PMID: 35321641 PMCID: PMC8941797 DOI: 10.1186/s12876-022-02212-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Accepted: 03/15/2022] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND AND AIMS Chronic atrophic gastritis (CAG) is a precancerous form of gastric cancer. However, with pathological diagnosis as the gold standard, the sensitivity of endoscopic diagnosis of atrophy is only 42%. We developed a deep learning (DL)-based real-time video monitoring diagnostic model for endoscopic CAG and conducted a prospective cohort study to verify whether this diagnostic model could improve the diagnosis rate of endoscopic CAG compared with that of endoscopists. METHODS A U-NET network was used to build a real-time video monitoring diagnostic model for endoscopic CAG based on DL. We enrolled 431 patients who underwent gastroscopy from October 1, 2020, to December 1, 2020. To keep the baseline data of enrolled patient uniform and control for confounding factors, we applied a paired design and included the same patients in both the DL and the endoscopist group. RESULTS The DL model improved the diagnosis rate of endoscopic CAG compared with that of endoscopists. Compared with diagnoses by endoscopists, the proportions of moderate and severe CAG in the atrophy patients diagnosed by the DL model were significantly larger, the proportion of "type O" CAG was significantly larger, the number of atrophy sites found was significantly increased, and the number of biopsies was significantly decreased. Compared with diagnoses by endoscopists, in the atrophic lesions diagnosed by the DL model, the proportions of severe atrophy and severe intestinal metaplasia were significantly increased. CONCLUSIONS Our study suggested the DL model could improve the diagnosis rate of endoscopic CAG compared with that of endoscopists. TRIAL REGISTRATION ChiCTR2100044458, 18/03/2020.
Collapse
Affiliation(s)
- Quchuan Zhao
- Department of Gastroenterology, Xuanwu Hospital of Capital Medical University, 45 Chang-chun Street, Beijing, 100053, China
| | - Tianyu Chi
- Department of Gastroenterology, Xuanwu Hospital of Capital Medical University, 45 Chang-chun Street, Beijing, 100053, China.
| |
Collapse
|
45
|
Yang H, Yang WJ, Hu B. Gastric epithelial histology and precancerous conditions. World J Gastrointest Oncol 2022; 14:396-412. [PMID: 35317321 PMCID: PMC8919001 DOI: 10.4251/wjgo.v14.i2.396] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Revised: 11/08/2021] [Accepted: 01/05/2022] [Indexed: 02/06/2023] Open
Abstract
The most common histological type of gastric cancer (GC) is gastric adenocarcinoma arising from the gastric epithelium. Less common variants include mesenchymal, lymphoproliferative and neuroendocrine neoplasms. The Lauren scheme classifies GC into intestinal type, diffuse type and mixed type. The WHO classification includes papillary, tubular, mucinous, poorly cohesive and mixed GC. Chronic atrophic gastritis (CAG) and intestinal metaplasia are recommended as common precancerous conditions. No definite precancerous condition of diffuse/poorly/undifferentiated type is recommended. Chronic superficial inflammation and hyperplasia of foveolar cells may be the focus. Presently, the management of early GC and precancerous conditions mainly relies on endoscopy including diagnosis, treatment and surveillance. Management of precancerous conditions promotes the early detection and treatment of early GC, and even prevent the occurrence of GC. In the review, precancerous conditions including CAG, metaplasia, foveolar hyperplasia and gastric hyperplastic polyps derived from the gastric epithelium have been concluded, based on the overview of gastric epithelial histological organization and its renewal.
Collapse
Affiliation(s)
- Hang Yang
- Department of Gastroenterology, West China Hospital, Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Wen-Juan Yang
- Department of Gastroenterology, West China Hospital, Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Bing Hu
- Department of Gastroenterology, West China Hospital, Sichuan University, Chengdu 610041, Sichuan Province, China
| |
Collapse
|
46
|
Jin Z, Gan T, Wang P, Fu Z, Zhang C, Yan Q, Zheng X, Liang X, Ye X. Deep learning for gastroscopic images: computer-aided techniques for clinicians. Biomed Eng Online 2022; 21:12. [PMID: 35148764 PMCID: PMC8832738 DOI: 10.1186/s12938-022-00979-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Accepted: 01/21/2022] [Indexed: 12/13/2022] Open
Abstract
Gastric disease is a major health problem worldwide. Gastroscopy is the main method and the gold standard used to screen and diagnose many gastric diseases. However, several factors, such as the experience and fatigue of endoscopists, limit its performance. With recent advancements in deep learning, an increasing number of studies have used this technology to provide on-site assistance during real-time gastroscopy. This review summarizes the latest publications on deep learning applications in overcoming disease-related and nondisease-related gastroscopy challenges. The former aims to help endoscopists find lesions and characterize them when they appear in the view shed of the gastroscope. The purpose of the latter is to avoid missing lesions due to poor-quality frames, incomplete inspection coverage of gastroscopy, etc., thus improving the quality of gastroscopy. This study aims to provide technical guidance and a comprehensive perspective for physicians to understand deep learning technology in gastroscopy. Some key issues to be handled before the clinical application of deep learning technology and the future direction of disease-related and nondisease-related applications of deep learning to gastroscopy are discussed herein.
Collapse
Affiliation(s)
- Ziyi Jin
- Biosensor National Special Laboratory, Key Laboratory of Biomedical Engineering of Ministry of Education, College of Biomedical Engineering and Instrument Science, Zhejiang University, Hangzhou, 310027, People's Republic of China
| | - Tianyuan Gan
- Biosensor National Special Laboratory, Key Laboratory of Biomedical Engineering of Ministry of Education, College of Biomedical Engineering and Instrument Science, Zhejiang University, Hangzhou, 310027, People's Republic of China
| | - Peng Wang
- Biosensor National Special Laboratory, Key Laboratory of Biomedical Engineering of Ministry of Education, College of Biomedical Engineering and Instrument Science, Zhejiang University, Hangzhou, 310027, People's Republic of China
| | - Zuoming Fu
- Biosensor National Special Laboratory, Key Laboratory of Biomedical Engineering of Ministry of Education, College of Biomedical Engineering and Instrument Science, Zhejiang University, Hangzhou, 310027, People's Republic of China
| | - Chongan Zhang
- Biosensor National Special Laboratory, Key Laboratory of Biomedical Engineering of Ministry of Education, College of Biomedical Engineering and Instrument Science, Zhejiang University, Hangzhou, 310027, People's Republic of China
| | - Qinglai Yan
- Hangzhou Center for Medical Device Quality Supervision and Testing, CFDA, Hangzhou, 310000, People's Republic of China
| | - Xueyong Zheng
- Department of General Surgery, Sir Run-Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 310016, People's Republic of China
| | - Xiao Liang
- Department of General Surgery, Sir Run-Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 310016, People's Republic of China
| | - Xuesong Ye
- Biosensor National Special Laboratory, Key Laboratory of Biomedical Engineering of Ministry of Education, College of Biomedical Engineering and Instrument Science, Zhejiang University, Hangzhou, 310027, People's Republic of China.
| |
Collapse
|
47
|
Luo J, Chen Y, Yang Y, Zhang K, Liu Y, Zhao H, Dong L, Xu J, Li Y, Wei W. Prognosis Prediction of Uveal Melanoma After Plaque Brachytherapy Based on Ultrasound With Machine Learning. Front Med (Lausanne) 2022; 8:777142. [PMID: 35127747 PMCID: PMC8816318 DOI: 10.3389/fmed.2021.777142] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Accepted: 12/22/2021] [Indexed: 12/29/2022] Open
Abstract
INTRODUCTION Uveal melanoma (UM) is the most common intraocular malignancy in adults. Plaque brachytherapy remains the dominant eyeball-conserving therapy for UM. Tumor regression in UM after plaque brachytherapy has been reported as a valuable prognostic factor. The present study aimed to develop an accurate machine-learning model to predict the 4-year risk of metastasis and death in UM based on ocular ultrasound data. MATERIAL AND METHODS A total of 454 patients with UM were enrolled in this retrospective, single-center study. All patients were followed up for at least 4 years after plaque brachytherapy and underwent ophthalmologic evaluations before the therapy. B-scan ultrasonography was used to measure the basal diameters and thickness of tumors preoperatively and postoperatively. Random Forest (RF) algorithm was used to construct two prediction models: whether a patient will survive for more than 4 years and whether the tumor will develop metastasis within 4 years after treatment. RESULTS Our predictive model achieved an area under the receiver operating characteristic curve (AUC) of 0.708 for predicting death using only a one-time follow-up record. Including the data from two additional follow-ups increased the AUC of the model to 0.883. We attained AUCs of 0.730 and 0.846 with data from one and three-time follow-up, respectively, for predicting metastasis. The model found that the amount of postoperative follow-up data significantly improved death and metastasis prediction accuracy. Furthermore, we divided tumor treatment response into four patterns. The D(decrease)/S(stable) patterns are associated with a significantly better prognosis than the I(increase)/O(other) patterns. CONCLUSIONS The present study developed an RF model to predict the risk of metastasis and death from UM within 4 years based on ultrasound follow-up records following plaque brachytherapy. We intend to further validate our model in prospective datasets, enabling us to implement timely and efficient treatments.
Collapse
Affiliation(s)
- Jingting Luo
- Beijing Tongren Eye Center, Beijing key Laboratory of Intraocular Tumor Diagnosis and Treatment, Beijing Ophthalmology & Visual Sciences Key Lab, Medical Artificial Intelligence Research and Verification Key Laboratory of the Ministry of Industry and Information Technology, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Yuning Chen
- Beijing Tongren Eye Center, Beijing key Laboratory of Intraocular Tumor Diagnosis and Treatment, Beijing Ophthalmology & Visual Sciences Key Lab, Medical Artificial Intelligence Research and Verification Key Laboratory of the Ministry of Industry and Information Technology, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Yuhang Yang
- Beijing Tongren Eye Center, Beijing key Laboratory of Intraocular Tumor Diagnosis and Treatment, Beijing Ophthalmology & Visual Sciences Key Lab, Medical Artificial Intelligence Research and Verification Key Laboratory of the Ministry of Industry and Information Technology, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Kai Zhang
- InferVision Healthcare Science and Technology Limited Company, Shanghai, China
| | - Yueming Liu
- Beijing Tongren Eye Center, Beijing key Laboratory of Intraocular Tumor Diagnosis and Treatment, Beijing Ophthalmology & Visual Sciences Key Lab, Medical Artificial Intelligence Research and Verification Key Laboratory of the Ministry of Industry and Information Technology, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Hanqing Zhao
- Beijing Tongren Eye Center, Beijing key Laboratory of Intraocular Tumor Diagnosis and Treatment, Beijing Ophthalmology & Visual Sciences Key Lab, Medical Artificial Intelligence Research and Verification Key Laboratory of the Ministry of Industry and Information Technology, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Li Dong
- Beijing Tongren Eye Center, Beijing key Laboratory of Intraocular Tumor Diagnosis and Treatment, Beijing Ophthalmology & Visual Sciences Key Lab, Medical Artificial Intelligence Research and Verification Key Laboratory of the Ministry of Industry and Information Technology, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Jie Xu
- Beijing Tongren Eye Center, Beijing key Laboratory of Intraocular Tumor Diagnosis and Treatment, Beijing Ophthalmology & Visual Sciences Key Lab, Medical Artificial Intelligence Research and Verification Key Laboratory of the Ministry of Industry and Information Technology, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Yang Li
- Beijing Tongren Eye Center, Beijing key Laboratory of Intraocular Tumor Diagnosis and Treatment, Beijing Ophthalmology & Visual Sciences Key Lab, Medical Artificial Intelligence Research and Verification Key Laboratory of the Ministry of Industry and Information Technology, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Wenbin Wei
- Beijing Tongren Eye Center, Beijing key Laboratory of Intraocular Tumor Diagnosis and Treatment, Beijing Ophthalmology & Visual Sciences Key Lab, Medical Artificial Intelligence Research and Verification Key Laboratory of the Ministry of Industry and Information Technology, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
48
|
Zhuang H, Bao A, Tan Y, Wang H, Xie Q, Qiu M, Xiong W, Liao F. Application and prospect of artificial intelligence in digestive endoscopy. Expert Rev Gastroenterol Hepatol 2022; 16:21-31. [PMID: 34937459 DOI: 10.1080/17474124.2022.2020646] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
INTRODUCTION With the progress of science and technology, artificial intelligence represented by deep learning has gradually begun to be applied in the medical field. Artificial intelligence has been applied to benign gastrointestinal lesions, tumors, early cancer, inflammatory bowel disease, gallbladder, pancreas, and other diseases. This review summarizes the latest research results on artificial intelligence in digestive endoscopy and discusses the prospect of artificial intelligence in digestive system diseases. AREAS COVERED We retrieved relevant documents on artificial intelligence in digestive tract diseases from PubMed and Medline. This review elaborates on the knowledge of computer-aided diagnosis in digestive endoscopy. EXPERT OPINION Artificial intelligence significantly improves diagnostic accuracy, reduces physicians' workload, and provides a shred of evidence for clinical diagnosis and treatment. Shortly, artificial intelligence will have high application value in the field of medicine.
Collapse
Affiliation(s)
- Huangming Zhuang
- Gastroenterology Department, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Anyu Bao
- Clinical Laboratory, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Yulin Tan
- Gastroenterology Department, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Hanyu Wang
- Gastroenterology Department, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Qingfang Xie
- Gastroenterology Department, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Meiqi Qiu
- Gastroenterology Department, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Wanli Xiong
- Gastroenterology Department, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Fei Liao
- Gastroenterology Department, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| |
Collapse
|
49
|
Visaggi P, de Bortoli N, Barberio B, Savarino V, Oleas R, Rosi EM, Marchi S, Ribolsi M, Savarino E. Artificial Intelligence in the Diagnosis of Upper Gastrointestinal Diseases. J Clin Gastroenterol 2022; 56:23-35. [PMID: 34739406 PMCID: PMC9988236 DOI: 10.1097/mcg.0000000000001629] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Artificial intelligence (AI) has enormous potential to support clinical routine workflows and therefore is gaining increasing popularity among medical professionals. In the field of gastroenterology, investigations on AI and computer-aided diagnosis (CAD) systems have mainly focused on the lower gastrointestinal (GI) tract. However, numerous CAD tools have been tested also in upper GI disorders showing encouraging results. The main application of AI in the upper GI tract is endoscopy; however, the need to analyze increasing loads of numerical and categorical data in short times has pushed researchers to investigate applications of AI systems in other upper GI settings, including gastroesophageal reflux disease, eosinophilic esophagitis, and motility disorders. AI and CAD systems will be increasingly incorporated into daily clinical practice in the coming years, thus at least basic notions will be soon required among physicians. For noninsiders, the working principles and potential of AI may be as fascinating as obscure. Accordingly, we reviewed systematic reviews, meta-analyses, randomized controlled trials, and original research articles regarding the performance of AI in the diagnosis of both malignant and benign esophageal and gastric diseases, also discussing essential characteristics of AI.
Collapse
Affiliation(s)
- Pierfrancesco Visaggi
- Gastroenterology Unit, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa
| | - Nicola de Bortoli
- Gastroenterology Unit, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa
| | - Brigida Barberio
- Department of Surgery, Oncology, and Gastroenterology, Division of Gastroenterology, University of Padua, Padua
| | - Vincenzo Savarino
- Gastroenterology Unit, Department of Internal Medicine, University of Genoa, Genoa
| | - Roberto Oleas
- Ecuadorean Institute of Digestive Diseases, Guayaquil, Ecuador
| | - Emma M. Rosi
- Gastroenterology Unit, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa
| | - Santino Marchi
- Gastroenterology Unit, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa
| | - Mentore Ribolsi
- Department of Digestive Diseases, Campus Bio Medico University of Rome, Roma, Italy
| | - Edoardo Savarino
- Department of Surgery, Oncology, and Gastroenterology, Division of Gastroenterology, University of Padua, Padua
| |
Collapse
|
50
|
Tong Y, Jing M, Zhao X, Liu H, Wei S, Li R, Liu X, Wang M, Song H, Zhao Y. Metabolomic Study of Zuojin Pill in Relieving 1-Methyl-3-nitro-1-nitrosoguanidine-Induced Chronic Atrophic Gastritis. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2021; 2021:7004798. [PMID: 34956382 PMCID: PMC8709764 DOI: 10.1155/2021/7004798] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Accepted: 11/27/2021] [Indexed: 11/29/2022]
Abstract
The classic prescription Zuojin Pill (ZJP) shows a good therapeutic effect on chronic atrophic gastritis (CAG); it is of great significance to clarify its specific mechanism. Therefore, we explore the mechanism of ZJP on MNNG-induced CAG by integrating approaches. First of all, through the pathological changes of gastric tissue and the expression level of PGI and PGI/II in serum, the expression of inflammation-related factors was determined by RT-PCR to determine the efficacy. Then, UPLC-Q-TOF/MS was used for plasma and urine metabolomic analysis to screen the specific potential biomarkers and metabolic pathway of ZJP in ameliorating CAG and to explore its possible mechanism. ZJP significantly ameliorate the pathological injury of gastric tissue, increase levels of PGI and PGI/II, and reduce the expression level of proinflammatory factors. Through metabolomic analysis, 9 potential metabolic differences were identified and 6 related metabolic pathways were enriched. These findings indicate for the first time the potential mechanism of ZJP in improving CAG induced by MNNG and are of great significance to the clinical development and application of ZJP-related drugs.
Collapse
Affiliation(s)
- Yuling Tong
- Department of Pharmacy, Chinese PLA General Hospital, Beijing, China
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Manyi Jing
- Department of Pharmacy, Chinese PLA General Hospital, Beijing, China
| | - Xu Zhao
- Senior Department of Hepatology, The Fifth Medical Center of PLA General Hospital, Beijing, China
| | - Honghong Liu
- Senior Department of Hepatology, The Fifth Medical Center of PLA General Hospital, Beijing, China
| | - Shizhang Wei
- Department of Pharmacy, Chinese PLA General Hospital, Beijing, China
| | - Ruisheng Li
- Research Center for Clinical and Translational Medicine, Fifth Medical Center of PLA General Hospital of Chinese, Beijing, China
| | - Xia Liu
- Department of Pharmacy, Chinese PLA General Hospital, Beijing, China
| | - Min Wang
- Department of Pharmacy, Chinese PLA General Hospital, Beijing, China
| | - Hongtao Song
- Department of Pharmacy, 900 Hospital of the Joint Logistics Team, Fuzhou, China
| | - Yanling Zhao
- Department of Pharmacy, Chinese PLA General Hospital, Beijing, China
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|