1
|
Cevik AB, Yıldız H, Sert H. Preventive Behaviors, Knowledge of Esophageal Cancer and Relationships Socio-Demographic Characteristics Among Older Adults: A Cross Sectional Study. Am J Health Promot 2025:8901171251326322. [PMID: 40156548 DOI: 10.1177/08901171251326322] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/01/2025]
Abstract
PurposeThis study aimed to investigate the risks of esophageal cancer (EC), protective behaviors, and risk awareness among Turkish adults aged 50 years and older, as well as their relationships with sociodemographic characteristics.DesignCross-sectional. Recruitment took place between February-April 2022, via online google forms and WhatsApp.SettingParticipants completed the survey online throught WhatsApp.SampleAnalyses included 214 participants (≥50 years from three provinces of Turkey); 73.8% female (n:158), 62.6% between 50-60 years (n:134 ), 55.6% had a university education (n:47).MeasuresStudy-specific survey items included questions about demographics, risky conditions and diseases, preventive health behaviors and Esophageal Cancer Knowledge Test (ECRKT).AnalysesFrequency or Mean, Percentage, Independent Samples t-Test, one-way analysis of variance (ANOVA), post hoc analysis (Tukey, LSD), and Cohen's effect size and linear regression analysis.ResultsThe participants had low ECRKT scores (x:13.50 ± 9.33, Min:0, Max:31) and reported the most consumed hot beverages (36.9%) among negative EC preventive behaviors. Male gender, not working, not having previously received training on EC, low education and income status cause low knowledge and awareness about EC (P < 0.05). Most of the participants had oral herpes (54.7%), vitamin deficiency (49.1%), and obesity (47.2%) in the past or now. Regarding the participants' EC risk knowledge and, female gender, being between 50-60 years old, and being employed had a weak effect (η2<0.01), while having previous EC training had a moderate effect (η2 = 0.006), and having university or higher education had a large positive effect (η2 = 0.14) (P < 0.05). Participants' level of education and previous educational experiences contribute significantly to esophageal cancer risk knowledge (P < 0.01).ConclusionThese findings highlight adults aged ≥50 years are exposed to significant EC risks, and have poor knowledge of EC risks. These results highlight the urgent need for educational campaigns to improve EC awareness.
Collapse
Affiliation(s)
| | - Hicran Yıldız
- Nursing Department, Uludag University Health Science Faculty, Bursa, Turkey
| | - Havva Sert
- Nursing Department, Sakarya University Health Science Faculty, Sakarya, Turkey
| |
Collapse
|
2
|
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. [PMID: 40112834 DOI: 10.1055/a-2529-5025] [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: 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
|
3
|
Jia YP, Liu DC, Cao TL, Jiang HZ, Li T, Li Y, Ding X. Advances and global trends of precancerous lesions of gastric cancer: A bibliometric analysis. World J Gastrointest Oncol 2025; 17:102111. [PMID: 40092937 PMCID: PMC11866257 DOI: 10.4251/wjgo.v17.i3.102111] [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: 10/09/2024] [Revised: 11/22/2024] [Accepted: 12/30/2024] [Indexed: 02/14/2025] Open
Abstract
BACKGROUND Precancerous lesions of gastric cancer (PLGC) represent a critical pathological stage in the development of intestinal gastric cancer. Early detection and diagnosis are key to reducing the incidence of gastric cancer. Substantial advancements have been made in PLGC research in recent years, making it necessary to provide updated reviews using bibliometric methods. We hypothesize that this review will identify emerging trends, key research areas, and gaps in PLGC research, providing insights that could guide future studies and enhance prevention strategies. AIM To comprehensively review the current state of research on PLGC, examining development trends and research hotspots. METHODS We conducted a bibliometric analysis of PLGC-related studies published between 2004 and 2023 using the Web of Science Core Collection database. We employed Software, including VOSviewer, CiteSpace, R software, and SCImago Graphica, to map scientific networks and visualize knowledge trends in terms of publication volume, countries/regions, institutions, journals, authors, and keywords. RESULTS A total of 4097 articles were included, and overall publication volume showed an increasing trend. Over the past two decades, China published the most articles, followed by the United States, Japan, South Korea, and Italy. Among the top 10 contributors, the United States ranked highest in institutions, authors, and citations and demonstrated the strongest international collaboration. Research keywords in this field were clustered into three main categories: Risk factors, pathogenesis, and diagnosis and treatment. Pathogenesis and molecular biomarkers remain key areas of focus. Future research should explore the mechanisms of gut microbiota, immune microenvironment, metabolic reprogramming, and epigenetics. Advanced technologies, including single-cell sequencing, spatially resolved analysis, multi-omics approaches, artificial intelligence, and machine learning, will likely accelerate in-depth investigations of PLGC. CONCLUSION PLGC research has rapidly developed in recent years, gaining considerable attention. This bibliometric analysis reveals research state and emerging trends over the past 20 years, providing insights for future studies.
Collapse
Affiliation(s)
- Yuan-Ping Jia
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing 100700, China
| | - Dian-Chun Liu
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Ting-Lan Cao
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Hui-Zhong Jiang
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing 100700, China
| | - Tao Li
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Yuan Li
- National Institute of Traditional Chinese Medicine Constitution and Preventive Treatment of Diseases, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Xia Ding
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| |
Collapse
|
4
|
Morgan DR, Corral JE, Li D, Montgomery EA, Riquelme A, Kim JJ, Sauer B, Shah SC. ACG Clinical Guideline: Diagnosis and Management of Gastric Premalignant Conditions. Am J Gastroenterol 2025:00000434-990000000-01623. [PMID: 40072510 DOI: 10.14309/ajg.0000000000003350] [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: 01/02/2024] [Accepted: 12/13/2024] [Indexed: 03/14/2025]
Abstract
Gastric premalignant conditions (GPMC) are common and include atrophic gastritis, gastric intestinal metaplasia, dysplasia, and certain gastric epithelial polyps. GPMC have an increased risk of progression to gastric adenocarcinoma. Gastric cancer (GC) in the United States represents an important cancer disparity because incidence rates are 2- to 13-fold greater in non-White individuals, particularly early-generation immigrants from regions of high GC incidence. The US 5-year survival rate for GC is 36%, which falls short of global standards and is driven by the fact that only a small percentage of GC in the US is diagnosed in the early, curable stage. This document represents the first iteration of American College of Gastroenterology guidelines on this topic and encompasses endoscopic surveillance for high-risk patients with GPMC, the performance of high-quality endoscopy and image-enhanced endoscopy for diagnosis and surveillance, GPMC histology criteria and reporting, endoscopic treatment of dysplasia, the role of Helicobacter pylori eradication, general risk reduction measures, and the management of autoimmune gastritis and gastric epithelial polyps. There is insufficient evidence to make a recommendation on upper endoscopic screening for GC/GPMC detection in US populations deemed high-risk for GC. Surveillance endoscopy is recommended for individuals at high risk for GPMC progression, as defined by endoscopic, histologic, and demographic factors, typically every 3 years, but an individualized interval may be warranted. H. pylori testing, treatment, and eradication confirmation are recommended in all individuals with GPMC. Extensive high-quality data from US populations regarding GPMC management are lacking, but continue to accrue, and the quality of evidence for the recommendations presented herein should be interpreted with this dynamic context in mind. The GPMC research and education agendas are broad and include high-quality prospective studies evaluating opportunistic endoscopic screening for GC/GPMC, refined delineation of what constitutes "high-risk" populations, development of novel biomarkers, alignment of best practices, implementation of training programs for improved GPMC/GC detection, and evaluation of the impact of these interventions on GC incidence and mortality in the US.
Collapse
Affiliation(s)
- Douglas R Morgan
- Division of Gastroenterology, The University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Juan E Corral
- Division of Gastroenterology, Prisma Health, Greenville, South Carolina, USA
| | - Dan Li
- Department of Gastroenterology, Kaiser Permanente Medical Center, Santa Clara, California, USA
- Kaiser Permanente Northern California Division of Research, Oakland, California, USA
| | - Elizabeth A Montgomery
- Department of Pathology, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Arnoldo Riquelme
- Department of Gastroenterology, Faculty of Medicine, Pontificia Universidad Católica de Chile, Center for Control and Prevention of Cancer (CECAN), Santiago, Chile
| | - John J Kim
- Division of Gastroenterology, Los Angeles General Medical Center, Los Angeles, California, USA
| | - Bryan Sauer
- Division of Gastroenterology, University of Virginia, Charlottesville, Virginia, USA
| | - Shailja C Shah
- Division of Gastroenterology, University of California, San Diego, La Jolla, California, USA
- Gastroenterology Section, Jennifer Moreno Veterans Affairs Medical Center, La Jolla, California, USA
| |
Collapse
|
5
|
Karhunen P, Tuomisto S, Goebeler S, Martiskainen M, Kok E. Common occurrence of atrophic gastritis in an ageing non-hospitalised population: an autopsy study. Age Ageing 2025; 54:afaf047. [PMID: 40037561 PMCID: PMC11879357 DOI: 10.1093/ageing/afaf047] [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: 06/17/2024] [Accepted: 02/12/2025] [Indexed: 03/06/2025] Open
Abstract
BACKGROUND Atrophic gastritis-the end stage of chronic gastritis-is an asymptomatic disease due to Helicobacter pylori infection causing decreased vitamin B12 and folate absorption, which may lead to severe haematological and neuropsychological disorders including Alzheimer's disease. The diagnosis requires endoscopy and biopsies from symptomatic patients, explaining why its true prevalence in the population is not well-known. OBJECTIVE We aimed to evaluate the prevalence of various stages of chronic gastritis in an autopsy series most closely representing the general population. SUBJECTS AND METHODS Gastric mucosa samples were collected prospectively from out-of-hospital deaths included in the Tampere Sudden Death Study (n = 70, mean age 63, age range 22-91 years). Antrum and corpus samples were stained with a H. pylori antibody and staged histopathologically. RESULTS Chronic gastritis with or without atrophic changes was detected in 40% of the cases. The proportion of healthy mucosa decreased age-dependently from 71.4% among individuals aged <50 years to 43.5% among the oldest individuals (>70 years), and that of chronic non-atrophic gastritis from 21.4% to 8.7%. In contrast, the prevalence of atrophic gastritis was 27.1% and increased in the age groups from 7.1% to 47.8% (P = .019) among the oldest individuals, showing a strong association (P < .0001) with H. pylori immunopositivity. CONCLUSIONS Atrophic gastritis is a common feature of the ageing stomach, which is observed in every second individual aged 70+ years, showing a strong association with H. pylori immunopositivity. Atrophic gastritis may be a more common risk factor in old age for diseases associated with low serum B12 and folate levels than has been previously known.
Collapse
Affiliation(s)
- Pekka Karhunen
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Fimlab Laboratories Ltd, Tampere, Pirkanmaa, Finland
| | - Sari Tuomisto
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Fimlab Laboratories Ltd, Tampere, Pirkanmaa, Finland
| | - Sirkka Goebeler
- Finnish Institute for Health and Welfare, Helsinki, Uusimaa, Finland
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Pirkanmaa, Finland
| | - Mika Martiskainen
- Finnish Institute for Health and Welfare, Helsinki, Uusimaa, Finland
- Department of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Eloise Kok
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Pirkanmaa, Finland
- Faculty of Medicine, Department of Pathology, University of Helsinki, Helsinki, Uusimaa, Finland
| |
Collapse
|
6
|
Tieck MP, Single C, Poli S, Kowarik MC, Ziemann U, Mengel A, Feil K. Screening tools for malignancy in patients with cryptogenic stroke: Systematic review. Eur Stroke J 2025:23969873241310760. [PMID: 40008556 DOI: 10.1177/23969873241310760] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/27/2025] Open
Abstract
Up to 20% of patients with cryptogenic ischemic stroke have an occult malignancy at the time of stroke presentation, providing an opportunity for early cancer detection. Despite this significant association, there is currently no consensus in international guidelines on how to systematically screen for malignancy in ischemic stroke patients. This review aims to summarize recent evidence on clinical features and scores, and predictive laboratory tests, that can guide malignancy screening in ischemic stroke patients. Our systemic search included PubMed, MEDLINE and Cochrane databases and yielded a total of 12 studies meeting the inclusion criteria for review. Elevated D-dimer levels and multiple infarcts in different cerebral circulations emerged as key markers. Based on the summarized data, we propose a flowchart for clinical decision-making regarding malignancy screening in patients with ischemic stroke. As the initial steps, we recommend using D-dimers cut-offs and stroke pattern on brain imaging to classify patients according to their risk profile. Based on the identified risk, we recommend a subsequent diagnostic workup addressing the most prevalent cancer types, including gastrointestinal tract, lung adenocarcinoma and gender-related cancer. The clinical implications of early malignancy screening and the need for evidence-based guidelines in cryptogenic stroke are discussed.
Collapse
Affiliation(s)
- Maria P Tieck
- Department of Neurology & Stroke, University Hospital Tübingen, Tübingen, Germany
- Hertie-Institute for clinical Brain Research, University Hospital Tübingen, Tübingen, Germany
| | - Constanze Single
- Department of Neurology & Stroke, University Hospital Tübingen, Tübingen, Germany
| | - Sven Poli
- Department of Neurology & Stroke, University Hospital Tübingen, Tübingen, Germany
- Hertie-Institute for clinical Brain Research, University Hospital Tübingen, Tübingen, Germany
| | - Markus C Kowarik
- Department of Neurology & Stroke, University Hospital Tübingen, Tübingen, Germany
- Hertie-Institute for clinical Brain Research, University Hospital Tübingen, Tübingen, Germany
| | - Ulf Ziemann
- Department of Neurology & Stroke, University Hospital Tübingen, Tübingen, Germany
- Hertie-Institute for clinical Brain Research, University Hospital Tübingen, Tübingen, Germany
| | - Annerose Mengel
- Department of Neurology & Stroke, University Hospital Tübingen, Tübingen, Germany
- Hertie-Institute for clinical Brain Research, University Hospital Tübingen, Tübingen, Germany
| | - Katharina Feil
- Department of Neurology & Stroke, University Hospital Tübingen, Tübingen, Germany
| |
Collapse
|
7
|
Huang RJ, Wichmann IA, Su A, Sathe A, Shum MV, Grimes SM, Meka R, Almeda A, Bai X, Shen J, Nguyen Q, Luo I, Han SS, Amieva MR, Hwang JH, Ji HP. A spatial transcriptomic signature of 26 genes resolved at single-cell resolution characterizes high-risk gastric cancer precursors. NPJ Precis Oncol 2025; 9:52. [PMID: 40000871 PMCID: PMC11861308 DOI: 10.1038/s41698-025-00816-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Accepted: 01/17/2025] [Indexed: 02/27/2025] Open
Abstract
Gastric cancer precursors demonstrate highly-variable rates of progression toward neoplasia. Certain high-risk precursors, such as gastric intestinal metaplasia with advanced histologic features, may be at up to 30-fold increased risk for progression compared to lower-risk intestinal metaplasia. The biological differences between high- and low-risk lesions have been incompletely explored. In this study, we use several clinical cohorts to characterize the microenvironment of advanced gastric cancer precursors relative to low-risk lesions using bulk, spatial, and single-cell gene expression assays. We identified a 26-gene panel which is associated with advanced lesions, localizes to metaplastic glands on histopathology, and is expressed in aberrant mature and immature intestinal cells not normally present in the healthy stomach. This gene expression signature suggests an important role of the immature intestinal lineages in promoting carcinogenesis in the metaplastic microenvironment. These findings may help to inform future biomarker development and strategies of gastric cancer prevention.
Collapse
Affiliation(s)
- Robert J Huang
- Division of Gastroenterology, Department of Medicine, Stanford School of Medicine, Stanford, CA, 94305, USA
| | - Ignacio A Wichmann
- Division of Oncology, Department of Medicine, Stanford School of Medicine, Stanford, CA, 94305, USA
- Division of Obstetrics and Gynecology, Department of Obstetrics, Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago, 8331150, Chile
- Advanced Center for Chronic Diseases (ACCDiS), Pontificia Universidad Católica de Chile, Santiago, 8331150, Chile
| | - Andrew Su
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Anuja Sathe
- Division of Oncology, Department of Medicine, Stanford School of Medicine, Stanford, CA, 94305, USA
| | - Miranda V Shum
- Division of Gastroenterology, Department of Medicine, Stanford School of Medicine, Stanford, CA, 94305, USA
| | - Susan M Grimes
- Division of Oncology, Department of Medicine, Stanford School of Medicine, Stanford, CA, 94305, USA
| | - Rithika Meka
- Division of Oncology, Department of Medicine, Stanford School of Medicine, Stanford, CA, 94305, USA
| | - Alison Almeda
- Division of Oncology, Department of Medicine, Stanford School of Medicine, Stanford, CA, 94305, USA
| | - Xiangqi Bai
- Division of Oncology, Department of Medicine, Stanford School of Medicine, Stanford, CA, 94305, USA
| | - Jeanne Shen
- Department of Pathology, Stanford School of Medicine, Stanford, CA, 94305, USA
| | - Quan Nguyen
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Ingrid Luo
- Quantitative Sciences Unit, Department of Medicine, Stanford School of Medicine, Stanford, CA, 94305, USA
| | - Summer S Han
- Quantitative Sciences Unit, Department of Medicine, Stanford School of Medicine, Stanford, CA, 94305, USA
- Department of Neurosurgery, Stanford School of Medicine, Stanford, CA, 94305, USA
- Stanford Cancer Institute, Stanford, CA, 94305, USA
| | - Manuel R Amieva
- Department of Microbiology and Immunology, Stanford University, Stanford, CA, 94305, USA
- Department of Pediatrics, Stanford University, Stanford, CA, 94305, USA
| | - Joo Ha Hwang
- Division of Gastroenterology, Department of Medicine, Stanford School of Medicine, Stanford, CA, 94305, USA.
| | - Hanlee P Ji
- Division of Oncology, Department of Medicine, Stanford School of Medicine, Stanford, CA, 94305, USA.
| |
Collapse
|
8
|
Ou JY, Liu Y, Zhang L, Luo TQ, Li JY, Lu LM, Wang L, He QR, Liu X, Pan HF. Assessing the quality and integrating the evidence and strength of recommendations in the guidelines for gastric precancerous lesions. BMC Cancer 2025; 25:272. [PMID: 39955530 PMCID: PMC11830177 DOI: 10.1186/s12885-025-13687-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Accepted: 02/07/2025] [Indexed: 02/17/2025] Open
Abstract
BACKGROUND Clinical practice guidelines (CPGs) are intended to offer appropriate recommendations for clinical practice based on the available evidence while acknowledging existing gaps and uncertainties. The quality of CPGs for gastric precancerous lesions (GPL), distribution of evidence quality, and strength of recommendations are unknown. OBJECTIVE Systematically evaluate the quality of CPGs for GPL and identify areas for improvement in the development process. METHODS PubMed, Embase, Cochrane Library, Cumulative Index to Nursing and Allied Health Literature, and six online CPG repositories were systematically searched for CPGs related to GPL. Three researchers independently assessed the methodological quality of the included CPGs by using the AGREE II tool. The reporting and recommendation quality of the CPGs were evaluated using the RIGHT and AGREE-REX tools through consensus. Evidence-based CPGs were analyzed using the Grading of Recommendation Assessment, Development, and Evaluation system to determine the distribution of quality of evidence and strength of recommendations. RESULTS A total of 4046 records were identified; nine CPGs met the eligibility criteria for this study. The mean overall score for the methodological quality of the CPGs was 46.22%. Among the six domains, the mean score for clarity of presentation was the highest (71.67%), while the mean score for applicability was the lowest (24.56%). Among the nine CPGs, only one was considered high quality. Regarding reporting quality, domains 1, 3, and 4 had mean reporting rates equal to or higher than 60%. The mean overall score for the recommendation quality was 19.11%. In total, 235 recommendations were identified through the screening process, of which 64.4% were classified as strong. However, only 17.5% of the strong recommendations were supported by high-quality evidence. CONCLUSION The overall quality of CPGs for GPL was poor, with uneven quality across domains. In addition, the consistency between the strength of recommendations and the quality of evidence was poor.
Collapse
Affiliation(s)
- Jia-Yin Ou
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province, 510405, China
| | - Yang Liu
- The Second Clinical Medical College of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province, 510405, China
| | - Lang Zhang
- The Second Clinical Medical College of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province, 510405, China
| | - Tian-Qi Luo
- The First Clinical Medical College of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province, 510405, China
| | - Jia-Yu Li
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province, 510405, China
| | - Li-Ming Lu
- South China Research Center for Acupuncture and Moxibustion, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province, 510006, China
- Medical College of Acu-Moxi and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province, 510006, China
| | - Lin Wang
- South China Research Center for Acupuncture and Moxibustion, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province, 510006, China
- Medical College of Acu-Moxi and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province, 510006, China
| | - Qiu-Rong He
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province, 510006, China
| | - Xin Liu
- The First Clinical Medical College of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province, 510405, China
| | - Hua-Feng Pan
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province, 510405, China.
| |
Collapse
|
9
|
Zheng S, Wang Y, Ni C, Guo R, Qiu X, Chen J, Wang L, Sun X, Chen M, Liu Y, Yuan Y, Gong Y. Helicobacter pylori SlyD stabilizes TPT1 via hnRNPK and enhances OCT1-mediated CDX2 transcriptional activation to drive gastric intestinal metaplasia. BMC Med 2025; 23:71. [PMID: 39915880 PMCID: PMC11803974 DOI: 10.1186/s12916-025-03911-8] [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: 09/13/2024] [Accepted: 01/24/2025] [Indexed: 02/09/2025] Open
Abstract
BACKGROUND Gastric intestinal metaplasia (GIM) represents an important precancerous lesion in intestinal-type gastric cancer, triggered by persistent Helicobacter pylori (H. pylori) infection. In a previous study, we unveiled SlyD as a novel virulence factor of H. pylori, establishing its role in GIM induction through TPT1. However, the underlying mechanism remains undetermined. METHODS Gastric epithelial cells were stimulated with H. pylori 26695, a SlyD inactivated mutant (ΔSlyD), and purified HpSlyD protein, respectively. Real-time qPCR and western blot were subsequently used to assess the expression levels of hnRNPK, TPT1, OCT1, and GIM markers. RNA sequencing was employed to identify differentially expressed genes associated with H. pylori SlyD infection. Protein stability was evaluated using cycloheximide. Molecular interactions were investigated through co-immunoprecipitation, chromatin immunoprecipitation, and dual-luciferase reporter assays. Additionally, molecular docking was utilized to predict TPT1 inhibitors. Immunohistochemistry staining was conducted to validate hnRNPK, TPT1, OCT1, and CDX2 expression in gastric tissue samples from both human and Mongolian gerbils. RESULTS H. pylori SlyD upregulates TPT1 and induces the expression of GIM markers through hnRNPK. The interaction between hnRNPK and TPT1 enhances TPT1 protein stability, with H. pylori SlyD intensifying this association. TPT1 promotes the expression of GIM markers mediated via OCT1, which binds to CDX2 promoter region, thereby modulating its transcriptional activity. Dihydroartemisinin has the potential to target TPT1, inhibiting the H. pylori SlyD-induced expression of GIM markers. CONCLUSIONS In vitro and in vivo experiments verified that H. pylori SlyD enhances TPT1 stability through hnRNPK, leading to OCT1-mediated transcriptional activation of CDX2 and the initiation of the GIM process. Our study offers novel perspectives on the pathogenesis of H. pylori-related gastric precancerous conditions.
Collapse
Affiliation(s)
- Shuwen Zheng
- Tumor Etiology and Screening Department of Cancer Institute and General Surgery, the First Hospital of China Medical University, Shenyang, 110001, China
- Key Laboratory of Cancer Etiology and Prevention in Liaoning Education Department, the First Hospital of China Medical University, Shenyang, 110001, China
- Key Laboratory of GI Cancer Etiology and Prevention in Liaoning Province, the First Hospital of China Medical University, Shenyang, 110001, China
| | - Yingying Wang
- Tumor Etiology and Screening Department of Cancer Institute and General Surgery, the First Hospital of China Medical University, Shenyang, 110001, China
- Key Laboratory of Cancer Etiology and Prevention in Liaoning Education Department, the First Hospital of China Medical University, Shenyang, 110001, China
- Key Laboratory of GI Cancer Etiology and Prevention in Liaoning Province, the First Hospital of China Medical University, Shenyang, 110001, China
| | - Chuxuan Ni
- Tumor Etiology and Screening Department of Cancer Institute and General Surgery, the First Hospital of China Medical University, Shenyang, 110001, China
- Key Laboratory of Cancer Etiology and Prevention in Liaoning Education Department, the First Hospital of China Medical University, Shenyang, 110001, China
- Key Laboratory of GI Cancer Etiology and Prevention in Liaoning Province, the First Hospital of China Medical University, Shenyang, 110001, China
| | - Rui Guo
- Tumor Etiology and Screening Department of Cancer Institute and General Surgery, the First Hospital of China Medical University, Shenyang, 110001, China
- Key Laboratory of Cancer Etiology and Prevention in Liaoning Education Department, the First Hospital of China Medical University, Shenyang, 110001, China
- Key Laboratory of GI Cancer Etiology and Prevention in Liaoning Province, the First Hospital of China Medical University, Shenyang, 110001, China
| | - Xunan Qiu
- Tumor Etiology and Screening Department of Cancer Institute and General Surgery, the First Hospital of China Medical University, Shenyang, 110001, China
- Key Laboratory of Cancer Etiology and Prevention in Liaoning Education Department, the First Hospital of China Medical University, Shenyang, 110001, China
- Key Laboratory of GI Cancer Etiology and Prevention in Liaoning Province, the First Hospital of China Medical University, Shenyang, 110001, China
| | - Jijun Chen
- Tumor Etiology and Screening Department of Cancer Institute and General Surgery, the First Hospital of China Medical University, Shenyang, 110001, China
- Key Laboratory of Cancer Etiology and Prevention in Liaoning Education Department, the First Hospital of China Medical University, Shenyang, 110001, China
- Key Laboratory of GI Cancer Etiology and Prevention in Liaoning Province, the First Hospital of China Medical University, Shenyang, 110001, China
| | - Lu Wang
- Tumor Etiology and Screening Department of Cancer Institute and General Surgery, the First Hospital of China Medical University, Shenyang, 110001, China
- Key Laboratory of Cancer Etiology and Prevention in Liaoning Education Department, the First Hospital of China Medical University, Shenyang, 110001, China
- Key Laboratory of GI Cancer Etiology and Prevention in Liaoning Province, the First Hospital of China Medical University, Shenyang, 110001, China
| | - Xiaohu Sun
- Tumor Etiology and Screening Department of Cancer Institute and General Surgery, the First Hospital of China Medical University, Shenyang, 110001, China
- Key Laboratory of Cancer Etiology and Prevention in Liaoning Education Department, the First Hospital of China Medical University, Shenyang, 110001, China
- Key Laboratory of GI Cancer Etiology and Prevention in Liaoning Province, the First Hospital of China Medical University, Shenyang, 110001, China
| | - Moye Chen
- Tumor Etiology and Screening Department of Cancer Institute and General Surgery, the First Hospital of China Medical University, Shenyang, 110001, China
- Key Laboratory of Cancer Etiology and Prevention in Liaoning Education Department, the First Hospital of China Medical University, Shenyang, 110001, China
- Key Laboratory of GI Cancer Etiology and Prevention in Liaoning Province, the First Hospital of China Medical University, Shenyang, 110001, China
| | - Yunen Liu
- Tumor Etiology and Screening Department of Cancer Institute and General Surgery, the First Hospital of China Medical University, Shenyang, 110001, China
- Key Laboratory of Cancer Etiology and Prevention in Liaoning Education Department, the First Hospital of China Medical University, Shenyang, 110001, China
- Key Laboratory of GI Cancer Etiology and Prevention in Liaoning Province, the First Hospital of China Medical University, Shenyang, 110001, China
| | - Yuan Yuan
- Tumor Etiology and Screening Department of Cancer Institute and General Surgery, the First Hospital of China Medical University, Shenyang, 110001, China.
- Key Laboratory of Cancer Etiology and Prevention in Liaoning Education Department, the First Hospital of China Medical University, Shenyang, 110001, China.
- Key Laboratory of GI Cancer Etiology and Prevention in Liaoning Province, the First Hospital of China Medical University, Shenyang, 110001, China.
| | - Yuehua Gong
- Tumor Etiology and Screening Department of Cancer Institute and General Surgery, the First Hospital of China Medical University, Shenyang, 110001, China.
- Key Laboratory of Cancer Etiology and Prevention in Liaoning Education Department, the First Hospital of China Medical University, Shenyang, 110001, China.
- Key Laboratory of GI Cancer Etiology and Prevention in Liaoning Province, the First Hospital of China Medical University, Shenyang, 110001, China.
| |
Collapse
|
10
|
He L, Zhou J, Ding D, Jiang Y, Yang R, Li Z. MiR-99a-3p downregulates TRIM21 to promote gastric cancer development. Mol Cell Biochem 2025; 480:1001-1012. [PMID: 38720056 PMCID: PMC11835897 DOI: 10.1007/s11010-024-05005-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Accepted: 04/03/2024] [Indexed: 02/19/2025]
Abstract
Gastric cancer (GC) stands as one of the most formidable malignancies worldwide. It is well-established that miRNAs play a crucial role in the initiation and progression of various human cancers. Among these, miR-99a-3p has been implicated in the pathogenesis of GC. In the context of our study, we embarked on the comprehensive examination of miR-99a-3p expression in GC cells. Additionally, we sought to establish a correlation between miR-99a-3p expression levels and the overall survival (OS) of GC patients, and our findings hinted at its potential role in predicting an unfavorable prognosis. To further investigate the functional implications of miR-99a-3p in GC, we conducted a series of cell-based experiments after successfully knocking down miR-99a-3p. These investigations uncovered a substantial inhibition of cellular events associated with tumor progression. Moreover, employing TargetScan, we identified Tripartite motif-containing protein 21 (TRIM21) as a putative target with a binding site for miR-99a-3p. Subsequent dual-luciferase reporter gene assay confirmed the direct interaction between miR-99a-3p and TRIM21. Western blot analysis validated the alteration in TRIM21 expression levels, revealing an upregulation upon miR-99a-3p knockdown. Building on these molecular findings, we extended our investigations to human GC tissues, where we observed a downregulation of TRIM21, which, notably, correlated with shorter overall survival. Lastly, to further solidify our conclusions, we conducted a series of in vitro and in vivo rescue experiments, collectively suggesting that miR-99a-3p promoted the progression of GC cells through the downregulation of TRIM21. In summary, our study comprehensively explored the role of miR-99a-3p in GC, revealing its association with unfavorable patient outcomes, functional implications in tumor progression, and a direct regulatory relationship with TRIM21. These findings collectively underscore the significance of miR-99a-3p in the pathogenesis of GC and present a potential therapeutic avenue for further investigation.
Collapse
Affiliation(s)
- Ling He
- Department of Gastroenterology, Affiliated Hospital of Jiangxi University of TCM, No.445, Bayi Road, Nanchang, 330006, Jiangxi, China
| | - Jiaoli Zhou
- Jiangxi University of TCM, No.56, Yangming Road, Nanchang, 330006, Jiangxi, China
| | - Doukun Ding
- Jiangxi University of TCM, No.56, Yangming Road, Nanchang, 330006, Jiangxi, China
| | - Yongjing Jiang
- Jiangxi University of TCM, No.56, Yangming Road, Nanchang, 330006, Jiangxi, China
| | - Rui Yang
- Jiangxi University of TCM, No.56, Yangming Road, Nanchang, 330006, Jiangxi, China
| | - Zhiming Li
- Department of Oncology, Affiliated Hospital of Jiangxi University of TCM, No.445, Bayi Road, Nanchang, 330006, Jiangxi, China.
| |
Collapse
|
11
|
Shah SC, Wang AY, Wallace MB, Hwang JH. AGA Clinical Practice Update on Screening and Surveillance in Individuals at Increased Risk for Gastric Cancer in the United States: Expert Review. Gastroenterology 2025; 168:405-416.e1. [PMID: 39718517 DOI: 10.1053/j.gastro.2024.11.001] [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: 09/19/2024] [Revised: 11/01/2024] [Accepted: 11/02/2024] [Indexed: 12/25/2024]
Abstract
DESCRIPTION Gastric cancer (GC) is a leading cause of preventable cancer and mortality in certain US populations. The most impactful way to reduce GC mortality is via primary prevention, namely Helicobacter pylori eradication, and secondary prevention, namely endoscopic screening and surveillance of precancerous conditions, such as gastric intestinal metaplasia (GIM). An emerging body of evidence supports the possible impact of these strategies on GC incidence and mortality in identifiable high-risk populations in the United States. Accordingly, the primary objective of this American Gastroenterological Association (AGA) Clinical Practice Update (CPU) Expert Review is to provide best practice advice for primary and secondary prevention of GC in the context of current clinical practice and evidence in the United States. METHODS This CPU Expert Review was commissioned and approved by the AGA Institute CPU Committee and the AGA Governing Board to provide timely guidance on a topic of high clinical importance to the AGA membership, and underwent internal peer review by the CPU Committee and external peer review through standard procedures of Gastroenterology. These best practice advice statements were drawn from a review of the published literature and expert opinion. Because systematic reviews were not performed, these best practice advice statements do not carry formal ratings regarding the quality of evidence or strength of the presented considerations. Best Practice Advice Statements BEST PRACTICE ADVICE 1: There are identifiable high-risk groups in the United States who should be considered for GC screening. These include first-generation immigrants from high-incidence GC regions and possibly other non-White racial and ethnic groups, those with a family history of GC in a first-degree relative, and individuals with certain hereditary gastrointestinal polyposis or hereditary cancer syndromes. BEST PRACTICE ADVICE 2: Endoscopy is the best test for screening or surveillance in individuals at increased risk for GC. Endoscopy enables direct visualization to endoscopically stage the mucosa and identify areas concerning for neoplasia, as well as enables biopsies for further histologic examination and mucosal staging. Both endoscopic and histologic staging are key for risk stratification and determining whether ongoing surveillance is indicated and at what interval. BEST PRACTICE ADVICE 3: High-quality upper endoscopy for the detection of premalignant and malignant gastric lesions should include the use of a high-definition white-light endoscopy system with image enhancement, gastric mucosal cleansing, and insufflation to achieve optimal mucosal visualization, in addition to adequate visual inspection time, photodocumentation, and use of a systematic biopsy protocol for mucosal staging when appropriate. BEST PRACTICE ADVICE 4: H pylori eradication is essential and serves as an adjunct to endoscopic screening and surveillance for primary and secondary prevention of GC. Opportunistic screening for H pylori infection should be considered in individuals deemed to be at increased risk for GC (refer to Best Practice Advice 1). Screening for H pylori infection in adult household members of individuals who test positive for H pylori (so-called "familial-based testing") should also be considered. BEST PRACTICE ADVICE 5: In individuals with suspected gastric atrophy with or without intestinal metaplasia, gastric biopsies should be obtained according to a systematic protocol (eg, updated Sydney System) to enable histologic confirmation and staging. A minimum of 5 total biopsies should be obtained, with samples from the antrum/incisura and corpus placed in separately labeled jars (eg, jar 1, "antrum/incisura" and jar 2, "corpus"). Any suspicious areas should be described and biopsied separately. BEST PRACTICE ADVICE 6: GIM and dysplasia are endoscopically detectable. However, these findings often go undiagnosed when endoscopists are unfamiliar with the characteristic visual features; accordingly, there is an unmet need for improved training, especially in the United States. Artificial intelligence tools appear promising for the detection of early gastric neoplasia in the adequately visualized stomach, but data are too preliminary to recommend routine use. BEST PRACTICE ADVICE 7: Endoscopists should work with their local pathologists to achieve consensus for consistent documentation of histologic risk-stratification parameters when atrophic gastritis with or without metaplasia is diagnosed. At a minimum, the presence or absence of H pylori infection, severity of atrophy and/or metaplasia, and histologic subtyping of GIM, if applicable, should be documented to inform clinical decision making. BEST PRACTICE ADVICE 8: If the index screening endoscopy performed in an individual at increased risk for GC (refer to Best Practice Advice 1) does not identify atrophy, GIM, or neoplasia, then the decision to continue screening should be based on that individual's risk factors and preferences. If the individual has a family history of GC or multiple risk factors for GC, then ongoing screening should be considered. The optimal screening intervals in such scenarios are not well defined. BEST PRACTICE ADVICE 9: Endoscopists should ensure that all individuals with confirmed gastric atrophy with or without GIM undergo risk stratification. Individuals with severe atrophic gastritis and/or multifocal or incomplete GIM are likely to benefit from endoscopic surveillance, particularly if they have other risk factors for GC (eg, family history). Endoscopic surveillance should be considered every 3 years; however, intervals are not well defined and shorter intervals may be advisable in those with multiple risk factors, such as severe GIM that is anatomically extensive. BEST PRACTICE ADVICE 10: Indefinite and low-grade dysplasia can be difficult to reproducibly identify by endoscopy and accurately diagnose on histopathology. Accordingly, all dysplasia should be confirmed by an experienced gastrointestinal pathologist, and clinicians should refer patients with visible or nonvisible dysplasia to an endoscopist or center with expertise in the diagnosis and management of gastric neoplasia. Individuals with indefinite or low-grade dysplasia who are infected with H pylori should be treated and have eradication confirmed, followed by repeat endoscopy and biopsies by an experienced endoscopist, as visual and histologic discernment may improve once inflammation subsides. BEST PRACTICE ADVICE 11: Individuals with suspected high-grade dysplasia or early GC should undergo endoscopic submucosal dissection with the goal of en bloc, R0 resection to enable accurate pathologic staging with curative intent. Eradication of active H pylori infection is essential, but should not delay endoscopic intervention. Endoscopic submucosal dissection should be performed at a center with endoscopic and pathologic expertise. BEST PRACTICE ADVICE 12: Individuals with a history of successfully resected gastric dysplasia or cancer require ongoing endoscopic surveillance. Suggested surveillance intervals exist, but additional data are required to refine surveillance recommendations, particularly in the United States. BEST PRACTICE ADVICE 13: Type I gastric carcinoids in individuals with atrophic gastritis are typically indolent, especially if <1 cm. Endoscopists may consider resecting gastric carcinoids <1 cm and should endoscopically resect lesions measuring 1-2 cm. Individuals with type I gastric carcinoids >2 cm should undergo cross-sectional imaging and be referred for surgical resection, given the risk of metastasis. Individuals with type I gastric carcinoids should undergo surveillance, but the intervals are not well defined. BEST PRACTICE ADVICE 14: In general, only individuals who are fit for endoscopic or potentially surgical treatment should be screened for GC and continued surveillance of premalignant gastric conditions. If a person is no longer fit for endoscopic or surgical treatment, then screening and surveillance should be stopped. BEST PRACTICE ADVICE 15: To achieve health equity, a personalized approach should be taken to assess an individual's risk for GC to determine whether screening and surveillance should be pursued. In conjunction, modifiable risk factors for GC should be distinctly addressed, as most of these risk factors disproportionately impact people at high risk for GC and represent health care disparities.
Collapse
Affiliation(s)
- Shailja C Shah
- Division of Gastroenterology and Hepatology, University of California San Diego, San Diego, California; Gastroenterology Section, Jennifer Moreno Department of Veterans Affairs Medical Center, San Diego, California.
| | - Andrew Y Wang
- Division of Gastroenterology and Hepatology, University of Virginia, Charlottesville, Virginia
| | - Michael B Wallace
- Division of Gastroenterology and Hepatology, Mayo Clinic, Jacksonville, Florida
| | - Joo Ha Hwang
- Division of Gastroenterology and Hepatology, Stanford University School of Medicine, Palo Alto, California
| |
Collapse
|
12
|
Barauskaite E, Raciunas A, Vaicekauskas R. Endoscopic Screening and Surveillance of Gastrointestinal Cancer. Cureus 2025; 17:e79274. [PMID: 40125194 PMCID: PMC11926922 DOI: 10.7759/cureus.79274] [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] [Accepted: 02/19/2025] [Indexed: 03/25/2025] Open
Abstract
Gastrointestinal (GI) cancer is a major health concern, contributing significantly to mortality rates in many regions, including Europe. It affects millions of people worldwide and leads to hundreds of thousands of deaths each year. Early detection and treatment through endoscopic methods play a vital role, providing less invasive and more affordable options compared to traditional surgical procedures. Targeted screening is vital for conditions such as Barrett's esophagus (BE), esophageal adenocarcinoma (EAC), gastric cancer (GC), ampullary carcinoma (AC), and colorectal cancer (CRC), particularly in high-risk populations. Endoscopic surveillance significantly reduces cancer incidence and improves survival rates, highlighting the importance of continuous advancements and updated guidelines to enhance screening efficacy and patient outcomes.
Collapse
Affiliation(s)
- Emilija Barauskaite
- Department of Family Medicine Center, Vilnius University Hospital Santaros Clinics, Vilnius, LTU
| | - Andrius Raciunas
- Department of Family Medicine Center, Vilnius University Hospital Santaros Clinics, Vilnius, LTU
| | - Rolandas Vaicekauskas
- Department of Gastroenterology, Nephrourology, and Surgery, Vilnius University Hospital Santaros Clinics, Vilnius, LTU
| |
Collapse
|
13
|
Ligato I, Dilaghi E, Cozza G, Scalamonti S, Pilozzi E, Panzuto F, Lahner E, Esposito G. Endoscopic and histological assessment in first-degree relatives of gastric cancer patients undergoing gastroscopy: a cross-sectional study. Eur J Gastroenterol Hepatol 2025:00042737-990000000-00467. [PMID: 39975999 DOI: 10.1097/meg.0000000000002925] [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: 02/21/2025]
Abstract
BACKGROUND First-degree relatives of gastric cancer (GC) patients are known to have an increased risk of developing GC. However, guidelines in low-intermediate incidence regions often lack specific recommendations for managing both asymptomatic and symptomatic relatives at risk. AIM This study aimed to evaluate the differences in relevant histological findings (e.g. Helicobacter pylori infection, gastric precancerous and neoplastic conditions) between asymptomatic patients undergoing esophagogastroduodenoscopy due to first-degree relatives with GC and patients with symptoms or other clinical indications and presence of first-degree relatives with GC. The secondary aim was to identify the patient's risk factors of relevant histological findings. METHODS This single-center retrospective study included patients undergoing esophagogastroduodenoscopy with biopsies with the indication for first-degree relatives with GC from January 2008 to September 2022. They were analyzed in two groups based on whether they had additional symptoms or clinical indications for esophagogastroduodenoscopy. RESULTS Overall, 283 patients were included (54.5% asymptomatic vs. 45.5% symptomatic). Histological findings that led to changes in patient management were identified in 32% of cases. No significant differences in histological findings between the two groups were observed (P = 0.077). A subanalysis revealed that patients with male relatives affected by GC had a higher incidence of relevant histological findings than those with female family members with GC (P = 0.013) with an odds ratio of 3.10. CONCLUSION First-degree relatives of GC patients may be at risk for H. pylori infection and gastric precancerous conditions regardless of symptoms or other indications, and a proactive endoscopic screening could be considered even in countries with low GC incidence.
Collapse
Affiliation(s)
- Irene Ligato
- Department of Medical-Surgical Sciences and Translational Medicine, Sant'Andrea Hospital
| | - Emanuele Dilaghi
- Department of Medical-Surgical Sciences and Translational Medicine, Sant'Andrea Hospital
| | - Giulio Cozza
- Department of Medical-Surgical Sciences and Translational Medicine, Sant'Andrea Hospital
| | - Silvia Scalamonti
- Department of Medical-Surgical Sciences and Translational Medicine, Sant'Andrea Hospital
| | - Emanuela Pilozzi
- Department of Clinical and Molecular Medicine, Pathologic Morphological and Molecular Anatomy Unit, Sant'Andrea University Hospital, Sapienza University of Rome, Rome, Italy
| | - Francesco Panzuto
- Department of Medical-Surgical Sciences and Translational Medicine, Sant'Andrea Hospital
| | - Edith Lahner
- Department of Medical-Surgical Sciences and Translational Medicine, Sant'Andrea Hospital
| | - Gianluca Esposito
- Department of Medical-Surgical Sciences and Translational Medicine, Sant'Andrea Hospital
| |
Collapse
|
14
|
Khalaf K, Fujiyoshi Y, Bechara R. Endoscopic and clinical characteristics of autoimmune atrophic gastritis: Retrospective study. Endosc Int Open 2025; 13:a24774666. [PMID: 40012571 PMCID: PMC11863545 DOI: 10.1055/a-2477-4666] [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: 01/26/2024] [Accepted: 11/19/2024] [Indexed: 02/28/2025] Open
Abstract
Background and study aims Autoimmune atrophic gastritis (AIG) is a rare chronic autoimmune disease characterized by gastric mucosa inflammation and atrophy. Limited clinical data exist about AIG, especially in western populations. In addition, there are no western series on the magnifying endoscopic features in AIG. This study presents a cohort of 63 patients with AIG, reporting their clinical, laboratory, and endoscopic findings. Patients and methods A retrospective analysis was conducted on patients diagnosed with AIG at Kingston Health Sciences Centre, Canada, between January 2016 and December 2023. Data collected from medical records included age, sex, presenting symptoms, laboratory findings, endoscopic features, histopathology reports, and concomitant autoimmune diseases. Results The study included 63 patients with autoimmune gastritis. Positive anti-parietal cell antibodies were found in the majority of patients (84.13%), whereas positive anti-intrinsic factor antibodies were less prevalent (25.40%). Deficiencies in vitamin B12 (49.21%) and iron (76.19%) were observed, along with a high prevalence of anemia (71.43%) and concomitant autoimmune diseases (58.73%). The dominant magnification pattern of atrophy in the body was oval/slit in 57.14% of patients (n=36), followed by tubular in 30.16% (n=19) and foveolar in 12.70% (n=8). Prevalence of neoplasia in our study was 42.86% (n=27). Conclusion This study offers insights into the clinical, laboratory, and magnifying endoscopic features of patients with AIG. It demonstrates the three main magnifying endoscopic appearances of AIG and highlights the significant prevalence of gastric neoplasia, even in the low-risk Western population. These findings emphasize the importance of the endoscopic exam in identifying AIG and notably present the key magnifying endoscopy findings in a Western setting for the first time.
Collapse
Affiliation(s)
- Kareem Khalaf
- Division of Gastroenterology, St Michael's Hospital, Toronto, Canada
| | - Yusuke Fujiyoshi
- Division of Gastroenterology, The Ottawa Hospital, University of Ottawa, The Ottawa Hospital Foundation, Ottawa, Canada
| | - Robert Bechara
- Gastroenterology, Kingston Health Sciences Centre, Kingston, Canada
| |
Collapse
|
15
|
Papadia C, Marelli L, Wood E, Novelli M, Feakins R, Syrjänen KJ, Shidrawi R. Can GastroPanel be used as a triage tool to select patients with advanced atrophic gastritis for gastroscopy? A prospective clinical validation study. BMJ Open Gastroenterol 2025; 12:e001559. [PMID: 39762070 PMCID: PMC11749878 DOI: 10.1136/bmjgast-2024-001559] [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: 08/08/2024] [Accepted: 12/06/2024] [Indexed: 01/23/2025] Open
Abstract
OBJECTIVE Gastric adenocarcinoma (GAC) is the 17th most common cancer in the UK with a 5-year survival rate of 22%. GastroPanel (Biohit Oyj; Helsinki, Finland) is an ELISA kit that measures pepsinogen I (PGI); pepsinogen II (PGII); gastrin-17 (G-17); and Helicobacter pylori IgG antibodies (Hp IgG). PGI and the PGI/PGII ratio correlate inversely with the severity of chronic atrophic gastritis (AG). The aim of this study was to assess GastroPanel performance in the identification of moderate to severe AG in dyspepsia. METHODS In this UK, single-centre, prospective diagnostic accuracy study, 324 patients [56.8% (n=184) female; median age 57 years (range 39-92 years)] were recruited for gastroscopy with biopsy and histology according to the updated Sydney System (USS). Blood (plasma) samples were collected for GastroPanel analysis. Paired samples were obtained from 268 patients [56.3% (n=151) female; median age=57 (range 39-92 years)]. GastroPanel results were interpreted using the GastroSoft app (Biohit). RESULTS Overall agreement between GastroPanel and the USS classification was 90% (95% CI=86.7 to 93.8%), with a weighted kappa (κw) of 0.828 (95% CI=0.781 to 0.865). In receiver operating characteristics (ROC) curve analysis, using moderate/severe atrophic gastritis of the corpus (AGC2+) as the endpoint, AUC=0.840 (95% CI 0.630 to 1.000) and 0.960 (95% CI 0.907 to 1.000) for PGI and the PGI/PGII ratio, respectively. CONCLUSION GastroPanel is a reliable dyspepsia triage test distinguishing patients who can be safely treated conservatively from those with moderate to severe corpus atrophic gastritis at high risk of developing GAC.
Collapse
Affiliation(s)
- Cinzia Papadia
- Gastroenterology, Barts Health Trust, London, UK
- Immunology, Barts and the London School of Medicine and Dentistry, London, UK
| | - Laura Marelli
- Homerton University Hospital NHS Foundation Trust, London, UK
| | - Eleanor Wood
- Homerton University Hospital NHS Foundation Trust, London, UK
| | - Marco Novelli
- Department of Pathology, University College London, London, UK
- Department of Pathology, University College London, London, UK
| | - Roger Feakins
- Department of Pathology, University College London, London, UK
- Department of Pathology, Royal Free Hampstead NHS Trust, London, UK
| | - Kari Juhani Syrjänen
- Medicine, University of Oulu Cancer and Translational Medicine Research Unit, Oulu, Finland
| | - Ray Shidrawi
- Gastroenterology, Homerton University Hospital, London, UK
| |
Collapse
|
16
|
He S, Zhang Z, Song G, Wang Z, Dai C, Yan S, Jiang K, Song B, Li H, Cao M, Sun D, Yang F, Yan X, Zhang S, Teng Y, Li Q, Xia C, Chen W. Can patients with mild non-neoplastic lesions diagnosed at baseline screening be safely exempt from surveillance: evidence from multicenter community-based cohorts. SCIENCE CHINA. LIFE SCIENCES 2025; 68:263-271. [PMID: 39254888 DOI: 10.1007/s11427-023-2558-x] [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: 12/04/2023] [Accepted: 01/25/2024] [Indexed: 09/11/2024]
Abstract
Surveillance recommendations for gastric cancer (GC) in current guidelines focused on advanced precancerous lesions and were based on precise diagnosis of severity/extent of baseline lesions. We aimed to develop a less endoscopy-related equipment-dependent risk-stratification tool, and assessed whether mild-precursor-lesion patients can be safely exempt from surveillance. In the multicenter community-based cohort, 75,051 participants receiving baseline endoscopy were enrolled during 2015-2017 and followed-up until 2021. Cumulative incidence rates (CIRs) of GC for precancerous-conditions were calculated by Kaplan-Meier method and compared by Log-rank tests. Mixed-effects Cox regression models were used to detect potential factors for progression towards GC. A risk score was calculated as counts of selected factors. An independent cohort, including 26,586 participants was used for external validation. During a median follow-up of 6.25 years, CIRs of GC were 0.302%, 0.436%, and 4.756% for normal group, non-neoplastic (atrophic gastritis/intestinal metaplasia) and neoplastic lesions (low-grade/high-grade dysplasia), respectively (Ptrend<0.001). Four predictors, including male, ⩾60 years, smoking, and limited vegetable consumption, were selected for risk-stratification. High-risk patients (⩾3 risk factors) with non-neoplastic lesions showed higher GC risks (adjusted HR=7.73, 95%CI: 4.29-13.92), and their four-year CIR reached the one-year CIR of neoplastic lesions. Further categorizing non-neoplastic lesions by histological grade, both patients with moderate-to-severe lesions (aHR=3.07, 95%CI: 1.67-5.64) and high-risk patients with mild lesions (aHR=7.29, 95%CI: 3.58-14.86) showed higher risks. Consistent trends were observed in validation cohort. High-risk mild-precursor-lesion patients should receive surveillance within 3-5 years after baseline screening. Our study provides evidence on supplementing current guideline recommendations.
Collapse
Affiliation(s)
- Siyi He
- Office of Cancer Screening, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Zhiyi Zhang
- Department of Gastroenterology, Gansu Wuwei Tumor Hospital, Wuwei, 730000, China
| | - Guohui Song
- Cixian Cancer Institute, Handan, 056500, China
| | | | - Chunyun Dai
- Center for Disease Control and Prevention of Sheyang County, Yancheng, 224499, China
| | - Shipeng Yan
- Department of Cancer Prevention and Control, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410029, China
| | - Kun Jiang
- Luoshan Center for Disease Control and Prevention, Xinyang, 464299, China
| | - Bingbing Song
- Office for Cancer Control and Research, Affiliated Cancer Hospital of Harbin Medical University, Harbin, 150081, China
| | - He Li
- Office of Cancer Screening, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Maomao Cao
- Office of Cancer Screening, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Dianqin Sun
- Office of Cancer Screening, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Fan Yang
- Office of Cancer Screening, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Xinxin Yan
- Office of Cancer Screening, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Shaoli Zhang
- Office of Cancer Screening, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Yi Teng
- Office of Cancer Screening, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Qianru Li
- Office of Cancer Screening, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Changfa Xia
- Office of Cancer Screening, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Wanqing Chen
- Office of Cancer Screening, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| |
Collapse
|
17
|
Rouphael C, Elkin B, El Dahdah J, Moufawad M, Yang Q, Bena J, Shah S, Kim MK. Practice Trends among US Gastroenterologists following the 2020 American Gastroenterological Association Guidelines on Gastric Intestinal Metaplasia: Data from a Tertiary Care Center. J Clin Gastroenterol 2025; 59:62-69. [PMID: 38502036 DOI: 10.1097/mcg.0000000000001991] [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: 10/10/2023] [Accepted: 02/09/2024] [Indexed: 03/20/2024]
Abstract
BACKGROUND AND AIMS Studies show variability in gastroenterologists' management of gastric intestinal metaplasia (GIM) in the United States. In 2020, the American Gastroenterological Association published GIM guidelines, recommending physician-patient shared decision-making on GIM surveillance based on risk factors. We compared gastroenterologists' communication trends of a GIM finding and surveillance recommendations before and after 2020 and evaluated patient and provider factors associated with a surveillance recommendation. METHODS A sample of patients diagnosed with GIM on biopsies from upper endoscopies performed in 2018 (cohort A) and 2021 (cohort B) were included. Logistic regression analysis assessed the association between patient/provider characteristics and surveillance recommendations in the overall cohort and over time. MATERIALS In all, 347 patients were included: 175 in cohort A and 172 in B. Median age was 65.7 (56.0, 73.4), and 54.5% were females. Communication to patients about GIM findings and surveillance recommendations increased from 24.6% <2020 to 50% >2020 ( P <0.001) and 20% <2020 to 41.3% >2020 ( P <0.001), respectively. Overall, endoscopy >2020, family history of gastric cancer, autoimmune gastritis, female providers, and gastroenterologists with 10 to 20 years of experience were associated with a surveillance recommendation. The effect of family history of gastric cancer and the effect of the patient's female sex on surveillance was significantly different between both cohorts [Odds ratio (OR): 0.13, 95% (Confidence interval) CI: 0.02, 0.97 and OR 3.39, 95% CI: 1.12, 10.2, respectively). CONCLUSIONS Despite a 2-fold increase in surveillance recommendations after 2020, there was no meaningful effect of any of the patients' factors on a recommendation for surveillance over time, which raises the question as to whether surveillance is being offered to both average and high-risk patients without thorough risk stratification.
Collapse
Affiliation(s)
- Carol Rouphael
- Department of Gastroenterology, Hepatology and Nutrition, Digestive Disease Institute
| | | | | | | | - Qijun Yang
- Department of Qualitative Health Sciences, Cleveland Clinic, Cleveland, OH
| | - James Bena
- Department of Qualitative Health Sciences, Cleveland Clinic, Cleveland, OH
| | - Shailja Shah
- Department of Gastroenterology, University of California San Diego
- Jennifer Moreno Department of Veterans Affairs Medical Center, Gastroenterology Section, San Diego, CA
| | - Michelle K Kim
- Department of Gastroenterology, Hepatology and Nutrition, Digestive Disease Institute
| |
Collapse
|
18
|
Fansiwala K, Lewis MS, Pisegna JR. Gastritis: Pathophysiology and Clinical Management. Am J Gastroenterol 2025; 120:5-8. [PMID: 39569875 PMCID: PMC11695143 DOI: 10.14309/ajg.0000000000003216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Accepted: 10/25/2024] [Indexed: 11/22/2024]
Affiliation(s)
- Kush Fansiwala
- Department of Internal Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095, USA
| | - Michael S. Lewis
- Departments of Medicine and Pathology, Cedars Sinai Medical Center, Los Angeles, CA, 90048, USA
- Department of Pathology, VA Greater Los Angeles Medical Center, Los Angeles, CA, 90073, USA
| | - Joseph R. Pisegna
- Division of Gastroenterology, Hepatology and Parenteral Nutrition, Dept of Medicine VA Greater Los Angeles Healthcare System, Los Angeles, CA, 90073, USA
- Department of Human Nutrition David Geffen School of Medicine at UCLA, Los Angeles, CA, 90095, USA
| |
Collapse
|
19
|
Schauer C, Teng A, Signal V, Stanley J, Mules TC, Koea J, Inns SJ. Translating evidence into action: overcoming barriers to gastric cancer prevention in Aotearoa. J R Soc N Z 2024:1-19. [DOI: 10.1080/03036758.2024.2427818] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Accepted: 11/06/2024] [Indexed: 01/06/2025]
Affiliation(s)
- Cameron Schauer
- Department of Gastroenterology, Health New Zealand Te Whatu Ora, Waitematā, University of Auckland, Auckland, New Zealand
| | - Andrea Teng
- Department of Public Health, University of Otago, Wellington, New Zealand
| | - Virgina Signal
- Department of Public Health, University of Otago, Wellington, New Zealand
| | - James Stanley
- Department of Public Health, University of Otago, Wellington, New Zealand
| | - Thomas C Mules
- Malaghan Institute of Medical Research, Wellington, New Zealand
| | - Jonathan Koea
- Department of Surgery, University of Auckland, Auckland, New Zealand
| | - Stephen J. Inns
- Wellington, Health New Zealand Te Whatu Ora, Capital Coast and Hutt Valley, University of Otago, Dunedin, New Zealand
| |
Collapse
|
20
|
Liu A, Zhang X, Zhong J, Wang Z, Ge Z, Wang Z, Fan X, Zhang J. A deep learning approach for gastroscopic manifestation recognition based on Kyoto Gastritis Score. Ann Med 2024; 56:2418963. [PMID: 39498518 PMCID: PMC11539395 DOI: 10.1080/07853890.2024.2418963] [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: 04/24/2024] [Revised: 06/29/2024] [Accepted: 07/13/2024] [Indexed: 11/07/2024] Open
Abstract
OBJECTIVE The risk of gastric cancer can be predicted by gastroscopic manifestation recognition and the Kyoto Gastritis Score. This study aims to validate the applicability of AI approaches for recognizing gastroscopic manifestations according to the definition of Kyoto Gastritis Score, with the goal of improving early gastric cancer detection and reducing gastric cancer mortality. METHODS In this retrospective study, 29013 gastric endoscopy images were collected and carefully annotated into five categories according to the Kyoto Gastritis Score, i.e. atrophy (A), diffuse redness (DR), enlarged folds (H), intestinal metaplasia (IM), and nodularity (N). As a multi-label recognition task, we propose a deep learning approach composed of five GAM-EfficientNet models, each performing a multiple classification to quantify gastroscopic manifestations, i.e. no presentation or the severity score 0-2. This approach was compared with endoscopists of varying years of experience in terms of accuracy, specificity, precision, recall, and F1 score. RESULTS The approach demonstrated good performance in identifying the five manifestations of the Kyoto Gastritis Score, with an average accuracy, specificity, precision, recall, and F1 score of 78.70%, 91.92%, 80.23%, 78.70%, and 0.78, respectively. The average performance of five experienced endoscopists was 72.63%, 90.00%, 77.68%, 72.63%, and 0.73, while that of five less experienced endoscopists was 66.60%, 87.44%, 70.88%, 66.60%, and 0.66, respectively. The sample t-test indicates that the approach's average accuracy, specificity, precision, recall, and F1 score for identifying the five manifestations were significantly higher than those of less experienced endoscopists, experienced endoscopists, and all endoscopists on average (p < 0.05). CONCLUSION Our study demonstrates the potential of deep learning approaches on gastric manifestation recognition over junior, even senior endoscopists. Thus, the deep learning approach holds potential as an auxiliary tool, although prospective validation is still needed to assess its clinical applicability.
Collapse
Affiliation(s)
- Ao Liu
- School of Software Technology, Dalian University of Technology, Dalian, China
| | - Xilin Zhang
- Department of Digestive Endoscopy, Central Hospital of Dalian University of Technology, Dalian, China
- China Medical University, Shenyang, China
| | - Jiaxin Zhong
- School of Software Technology, Dalian University of Technology, Dalian, China
| | - Zilu Wang
- School of Software Technology, Dalian University of Technology, Dalian, China
| | - Zhenyang Ge
- Department of Digestive Endoscopy, Central Hospital of Dalian University of Technology, Dalian, China
| | - Zhong Wang
- School of Software Technology, Dalian University of Technology, Dalian, China
| | - Xiaoya Fan
- School of Software Technology, Dalian University of Technology, Dalian, China
| | - Jing Zhang
- Department of Digestive Endoscopy, Central Hospital of Dalian University of Technology, Dalian, China
| |
Collapse
|
21
|
De Prado Á, Cal-Sabater P, Fiz-López A, Izquierdo S, Corrales D, Pérez-Cózar F, H-Vázquez J, Arribas-Rodríguez E, Perez-Segurado C, Muñoz ÁM, Garrote JA, Arranz E, Marañón C, Cuesta-Sancho S, Fernández-Salazar L, Bernardo D. Complex immune network and regional consistency in the human gastric mucosa revealed by high-resolution spectral cytometry. Sci Rep 2024; 14:28685. [PMID: 39562636 PMCID: PMC11577052 DOI: 10.1038/s41598-024-78908-z] [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: 08/17/2023] [Accepted: 11/05/2024] [Indexed: 11/21/2024] Open
Abstract
The immune cellular landscape from the gastric mucosa remains largely unknown despite its relevance in several inflammatory conditions. Human gastric biopsies were obtained from the antrum, body and incisura from 10 individuals to obtain lamina propria mononuclear cells that were further characterized by spectral cytometry. Phenotypic hierarchical analyses identified a total of 52 different immune cell subsets within the human gastric mucosa revealing that T-cells (> 60%) and NK cells (> 20%) were the main populations. Within T-cells, CD4+ and CD8+ were equally represented with both subsets displaying mainly a memory and effector phenotype. NK cells, on the contrary, were largely of the early phenotype. No regional differences were observed for any subsets among the 3 locations. Following unsupervised analysis, a total of 82 clusters were found. Again, no differences were observed amongst locations although a great degree of inter-individual variability was found, largely influenced by the presence of H. pylori infection and dyspepsia. We have unraveled the human gastric immune cellular subset composition and a unique interindividual immune fingerprint with no inter-regional variations.
Collapse
Affiliation(s)
- Ángel De Prado
- Mucosal Immunology Lab, Unit of Excellence, Institute of Biomedicine and Molecular Genetics (IBGM), University of Valladolid-CSIC, Sanz y Forés 3., 47003, Valladolid, Spain
| | - Paloma Cal-Sabater
- Mucosal Immunology Lab, Unit of Excellence, Institute of Biomedicine and Molecular Genetics (IBGM), University of Valladolid-CSIC, Sanz y Forés 3., 47003, Valladolid, Spain
| | - Aida Fiz-López
- Mucosal Immunology Lab, Unit of Excellence, Institute of Biomedicine and Molecular Genetics (IBGM), University of Valladolid-CSIC, Sanz y Forés 3., 47003, Valladolid, Spain
| | - Sandra Izquierdo
- Gastroenterology Department, Hospital Clínico, Universitario (HCUV-SACYL), University of Valladolid, Valladolid, Spain
| | - Daniel Corrales
- Pathology Department, Hospital Clínico, Universitario (HCUV-SACYL). Valladolid, Valladolid, Spain
| | - Francisco Pérez-Cózar
- Pfizer-University of Granada-Junta de Andalucía Centre for Genomics and Oncological Research (GENYO), PTS, Granada, Spain
| | - Juan H-Vázquez
- Mucosal Immunology Lab, Unit of Excellence, Institute of Biomedicine and Molecular Genetics (IBGM), University of Valladolid-CSIC, Sanz y Forés 3., 47003, Valladolid, Spain
| | - Elisa Arribas-Rodríguez
- Mucosal Immunology Lab, Unit of Excellence, Institute of Biomedicine and Molecular Genetics (IBGM), University of Valladolid-CSIC, Sanz y Forés 3., 47003, Valladolid, Spain
| | - Cándido Perez-Segurado
- Mucosal Immunology Lab, Unit of Excellence, Institute of Biomedicine and Molecular Genetics (IBGM), University of Valladolid-CSIC, Sanz y Forés 3., 47003, Valladolid, Spain
| | - Álvaro Martín Muñoz
- Flow Cytometry Facility. Unit of Excellence Instituto de Biología y Genética Molecular (IBGM), University of Valladolid-CSIC, Valladolid, Spain
| | - José A Garrote
- Mucosal Immunology Lab, Unit of Excellence, Institute of Biomedicine and Molecular Genetics (IBGM), University of Valladolid-CSIC, Sanz y Forés 3., 47003, Valladolid, Spain
| | - Eduardo Arranz
- Mucosal Immunology Lab, Unit of Excellence, Institute of Biomedicine and Molecular Genetics (IBGM), University of Valladolid-CSIC, Sanz y Forés 3., 47003, Valladolid, Spain
| | - Concepción Marañón
- Pfizer-University of Granada-Junta de Andalucía Centre for Genomics and Oncological Research (GENYO), PTS, Granada, Spain
| | - Sara Cuesta-Sancho
- Mucosal Immunology Lab, Unit of Excellence, Institute of Biomedicine and Molecular Genetics (IBGM), University of Valladolid-CSIC, Sanz y Forés 3., 47003, Valladolid, Spain
| | - Luis Fernández-Salazar
- Gastroenterology Department, Hospital Clínico, Universitario (HCUV-SACYL), University of Valladolid, Valladolid, Spain
| | - David Bernardo
- Mucosal Immunology Lab, Unit of Excellence, Institute of Biomedicine and Molecular Genetics (IBGM), University of Valladolid-CSIC, Sanz y Forés 3., 47003, Valladolid, Spain.
- Centro de Investigaciones Biomédicas en Red de Enfermedades Infecciosas (CIBERINFEC), Madrid, Spain.
| |
Collapse
|
22
|
Reyes-Placencia D, Cantú-Germano E, Latorre G, Espino A, Fernández-Esparrach G, Moreira L. Gastric Epithelial Polyps: Current Diagnosis, Management, and Endoscopic Frontiers. Cancers (Basel) 2024; 16:3771. [PMID: 39594726 PMCID: PMC11591925 DOI: 10.3390/cancers16223771] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Revised: 11/02/2024] [Accepted: 11/07/2024] [Indexed: 11/28/2024] Open
Abstract
Polyps are defined as luminal lesions that project into the mucosal surface of the gastrointestinal tract and are characterized according to their morphological and histological features [...].
Collapse
Affiliation(s)
- Diego Reyes-Placencia
- Department of Gastroenterology, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago 8320165, Chile
| | - Elisa Cantú-Germano
- Department of Gastroenterology, Fundació de Recerca Clínic Barcelona-Institut d’Investigacions Biomèdiques August Pi i Sunyer (FRCB-IDIBAPS), CIBEREHD, 08036 Barcelona, Spain
- Facultat de Medicina, Universitat de Barcelona, 08036 Barcelona, Spain
| | - Gonzalo Latorre
- Department of Gastroenterology, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago 8320165, Chile
| | - Alberto Espino
- Department of Gastroenterology, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago 8320165, Chile
| | - Glòria Fernández-Esparrach
- Department of Gastroenterology, Fundació de Recerca Clínic Barcelona-Institut d’Investigacions Biomèdiques August Pi i Sunyer (FRCB-IDIBAPS), CIBEREHD, 08036 Barcelona, Spain
- Facultat de Medicina, Universitat de Barcelona, 08036 Barcelona, Spain
| | - Leticia Moreira
- Department of Gastroenterology, Fundació de Recerca Clínic Barcelona-Institut d’Investigacions Biomèdiques August Pi i Sunyer (FRCB-IDIBAPS), CIBEREHD, 08036 Barcelona, Spain
- Facultat de Medicina, Universitat de Barcelona, 08036 Barcelona, Spain
| |
Collapse
|
23
|
Andersen GT, Ianevski A, Resell M, Pojskic N, Rabben HL, Geithus S, Kodama Y, Hiroyuki T, Kainov D, Grønbech JE, Hayakawa Y, Wang TC, Zhao CM, Chen D. Multi-bioinformatics revealed potential biomarkers and repurposed drugs for gastric adenocarcinoma-related gastric intestinal metaplasia. NPJ Syst Biol Appl 2024; 10:127. [PMID: 39496635 PMCID: PMC11535201 DOI: 10.1038/s41540-024-00455-0] [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: 06/05/2024] [Accepted: 10/13/2024] [Indexed: 11/06/2024] Open
Abstract
Biomarkers associated with the progression from gastric intestinal metaplasia (GIM) to gastric adenocarcinoma (GA), i.e., GA-related GIM, could provide valuable insights into identifying patients with increased risk for GA. The aim of this study was to utilize multi-bioinformatics to reveal potential biomarkers for the GA-related GIM and predict potential drug repurposing for GA prevention in patients. The multi-bioinformatics included gene expression matrix (GEM) by microarray gene expression (MGE), ScType (a fully automated and ultra-fast cell-type identification based solely on a given scRNA-seq data), Ingenuity Pathway Analysis, PageRank centrality, GO and MSigDB enrichments, Cytoscape, Human Protein Atlas and molecular docking analysis in combination with immunohistochemistry. To identify GA-related GIM, paired surgical biopsies were collected from 16 GIM-GA patients who underwent gastrectomy, yielding 64 samples (4 biopsies per stomach x 16 patients) for MGE. Co-analysis was performed by including scRNAseq and immunohistochemistry datasets of endoscopic biopsies of 37 patients. The results of the present study showed potential biomarkers for GA-related GIM, including GEM of individual patients, individual genes (such as RBP2 and CD44), signaling pathways, network of molecules, and network of signaling pathways with key topological nodes. Accordingly, potential treatment targets with repurposed drugs were identified including epidermal growth factor receptor, proto-oncogene tyrosine-protein kinase Src, paxillin, transcription factor Jun, breast cancer type 1 susceptibility protein, cellular tumor antigen p53, mouse double minute 2, and CD44.
Collapse
Affiliation(s)
- Gøran Troseth Andersen
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
- Department of Surgery, St. Olav's Hospital, Trondheim, Norway
- Department of Surgery, Namsos Hospital, Namsos, Norway
| | - Aleksandr Ianevski
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Mathilde Resell
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Naris Pojskic
- Laboratory for Bioinformatics and Biostatistics, University of Sarajevo - Institute for Genetic Engineering and Biotechnology, Sarajevo, Bosnia and Herzegovina
| | - Hanne-Line Rabben
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Synne Geithus
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Yosuke Kodama
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Tomita Hiroyuki
- Department of Tumor Pathology, Gifu University Graduate School of Medicine, Gifu, Japan
| | - Denis Kainov
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Jon Erik Grønbech
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
- Department of Surgery, St. Olav's Hospital, Trondheim, Norway
| | - Yoku Hayakawa
- Department of Gastroenterology, Tokyo University Hospital, Tokyo, Japan
| | - Timothy C Wang
- Department of Digestive and Liver Diseases and Herbert Iring Comprehensive Cancer Center, Columbia University Medical Center, New York, USA
| | - Chun-Mei Zhao
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Duan Chen
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology (NTNU), Trondheim, Norway.
| |
Collapse
|
24
|
Zhang T, Chen L, Li S, Shen C. Upregulation of CDC25B by transcription factor TEAD4 drives invasion and inhibits cisplatin sensitivity through cell adhesion in stomach adenocarcinoma. Anticancer Drugs 2024; 35:922-931. [PMID: 39079173 DOI: 10.1097/cad.0000000000001645] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2024]
Abstract
Cisplatin is crucial in management of advanced stomach adenocarcinoma, whereas development of chemotherapy resistance hinders overall efficacy of cisplatin. This work aims to explore role of CDC25B in cisplatin sensitivity in stomach adenocarcinoma and offer a possible mechanism for explaining its function. By using bioinformatics approaches, CDC25B and TEAD4 expression levels in stomach adenocarcinoma tissues and enriched pathways of CDC25B were analyzed. qRT-PCR of CDC25B and TEAD4 expression in stomach adenocarcinoma cells, CCK-8 detection of cell viability and IC 50 values, and colony formation assay on cell proliferation were performed. Cell adhesion experiment detected cell adhesion ability. Western blot detected expression of proteins related to cell adhesion, specifically Muc-1, ICAM-1, VCAM-1. Dual luciferase assay and ChIP experiment verified binding relationship between TEAD4 and CDC25B. CDC25B was upregulated in stomach adenocarcinoma tissues and cells, enriched in focal adhesion pathway. Treatment with cell adhesion inhibitors revealed that CDC25B overexpression inhibits the sensitivity of stomach adenocarcinoma to cisplatin through the cell adhesion pathway. CDC25B has an upstream transcription factor TEAD4, which targeted and bound to CDC25B and was highly expressed in stomach adenocarcinoma. Rescue experiment revealed that knocking down TEAD4 weakened suppressive impact of CDC25B overexpression on sensitivity of stomach adenocarcinoma cells to cisplatin. Transcription factor TEAD4 could activate the transcription of CDC25B through cell adhesion to drive cell invasion and reduce sensitivity of stomach adenocarcinoma to cisplatin. TEAD4 and CDC25B may become new targets for management of stomach adenocarcinoma.
Collapse
Affiliation(s)
- Tao Zhang
- Department of General Surgery, The Fourth Hospital of Changsha City
| | - Lijian Chen
- Department of General Surgery, Hunan Children's Hospital
| | - Shuang Li
- Department of Geriatrics, The Second Xiangya Hospital, Central South University
| | - Chao Shen
- Disinfection Supply Room, The Fourth Hospital of Changsha City, Changsha, China
| |
Collapse
|
25
|
Ang TL. Climate Sustainability as a Catalyst for Quality and Excellence in Gastrointestinal Endoscopy: A Narrative Review. J Dig Dis 2024; 25:638-644. [PMID: 39909063 DOI: 10.1111/1751-2980.13330] [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: 12/29/2024] [Accepted: 12/31/2024] [Indexed: 02/07/2025]
Abstract
The significant contribution of greenhouse gas (GHG) emissions to global warming and the resultant negative impact on health is well established. Within the hospital setting, the endoscopy center has been ranked third-after the operating theater complex and intensive care unit-in terms of the volume of hazardous medical waste generated. Such regulated medical waste cannot be recycled, and the disposal process results in higher costs and a larger carbon footprint. There have been clarion calls to reduce the number of endoscopic procedures as a means of reducing GHG emissions. Previous studies have demonstrated the carbon footprint of inappropriate endoscopic procedures. However, endoscopy is an important diagnostic and therapeutic tool that has been shown to be cost-effective. A focus simply on reduction in procedural case volume alone raises the unintended consequences of inequitable access to endoscopy for those in need. A more nuanced approach that is aligned with the innate sense of healthcare professional aspirations and pride, in terms of seeking quality and clinical excellence, should be considered. An evidence-based approach, with a focus on quality and efficiency, as well as appropriate and timely use of new technology, will serve as a catalyst for positive behavioral change toward quality procedures and excellence. This will improve patient satisfaction, increase professional expertise and pride, and reduce carbon footprint through minimization of inappropriate and avoidable repeat procedures.
Collapse
Affiliation(s)
- Tiing Leong Ang
- Department of Gastroenterology and Hepatology, Changi General Hospital, Singapore
- Duke-NUS Medical School, Singapore
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore
| |
Collapse
|
26
|
Zacharakis G, Dahale A, Elbary ERA, Babikir RR, Alla MA, Mustafa MO. Factors associated with precancerous stomach lesions and progresion: A 7-year multi-center prospective cohort study on the low incidence of gastric cancer in central Saudi Arabia. Saudi J Gastroenterol 2024; 30:389-398. [PMID: 39118443 PMCID: PMC11630487 DOI: 10.4103/sjg.sjg_172_24] [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: 05/14/2024] [Revised: 06/26/2024] [Accepted: 06/29/2024] [Indexed: 08/10/2024] Open
Abstract
BACKGROUND In Saudi Arabia (SA) no data are available on precancerous stomach lesions (PSLs) or the associated risk factors. We aimed to identify PSLs and investigate factors associated with PSLs and their progression. METHODS This 7-year prospective study screened for PSLs in asymptomatic Saudi patients aged 45-75 years in central SA ( n = 35,640). Those who had high-sensitivity guaiac fecal occult blood tests (HSgFOBT+) and negative colonoscopy results ( n = 1242) were subjected to upper GI endoscopy to identify PSLs and were followed up every 3 years or earlier, depending on the type of PSL. Factors associated with PSLs were investigated. RESULTS The 7-year participation rate was 86.9% (1080/1242). The 7-year prevalence of PSLs was 30.9% (334/1080). The incidence rate of PSLs was 134 new cases/100,000 population/year, total population at risk - 35,640 and 44.3 new cases/1,000 persons/year among the 1080 participants with HSgFOBT+ and negative colonoscopy results. Among the 334 participants with PSLs, 8 (2.4%) had neoplastic progression to GC during the surveillance period. Age, Helicobacter pylori infection, smoking status, a diet with preserved salty foods, low income, and a family history of GC were associated with PSLs. CONCLUSIONS The incidence of GC is low in central SA, but screening for PSLs among participants with HSgFOBT+ and negative colonoscopy findings may contribute to the early detection and subsequent treatment of GC. HP eradication, not smoking, normal body weight, and adhering to a healthy diet seem to be potential factors associated with the development of PSLs. Further studies are needed to search if such interventions would decrease the incidence of PSLs and progression to early GC.
Collapse
Affiliation(s)
- Georgios Zacharakis
- Endosocpy Unit, Department of Internal Medicine, College of Medicine, Prince Sattam bin, Abdulaziz University, Prince Sattam bin Abdulaziz University Hospital, Al-Kharj, Saudi Arabia
| | - Amol Dahale
- Endosocpy Unit, Department of Internal Medicine, College of Medicine, Prince Sattam bin, Abdulaziz University, Prince Sattam bin Abdulaziz University Hospital, Al-Kharj, Saudi Arabia
- Department of Gastroenterology, Dr. D.Y. Patil Medical College and Hospital and Research Centre, Pune, Maharashtra, India
| | - Elsayed R.A. Abd Elbary
- Endosocpy Unit, Department of Internal Medicine, College of Medicine, Prince Sattam bin, Abdulaziz University, Prince Sattam bin Abdulaziz University Hospital, Al-Kharj, Saudi Arabia
- Department of Oncosurgery, Faculty of Medicine, Al-Azhar University, Cairo, Egypt
| | - Rawan R.E. Babikir
- Endosocpy Unit, Department of Internal Medicine, College of Medicine, Prince Sattam bin, Abdulaziz University, Prince Sattam bin Abdulaziz University Hospital, Al-Kharj, Saudi Arabia
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Khartoum, Khartoum, Sudan
| | - Motaz A.N. Alla
- Endosocpy Unit, Department of Internal Medicine, College of Medicine, Prince Sattam bin, Abdulaziz University, Prince Sattam bin Abdulaziz University Hospital, Al-Kharj, Saudi Arabia
- Department of Anatomy, Faculty of Medicine, University of Al-Butana, Rafaa, Northern State, Sudan
| | - Mohamed O. Mustafa
- Endosocpy Unit, Department of Internal Medicine, College of Medicine, Prince Sattam bin, Abdulaziz University, Prince Sattam bin Abdulaziz University Hospital, Al-Kharj, Saudi Arabia
- Department of Anatomy, Faculty of Medicine, University of Gadarif, Al Qadarif, Sudan
| |
Collapse
|
27
|
Judge C, Halder A, Pateria P, Khor T, Muwanwella N, Chin M, Ragunath K. Outcomes and validity of risk stratification tools for endoscopic submucosal dissection of early gastric cancer in Western Australia. JGH Open 2024; 8:e70034. [PMID: 39554984 PMCID: PMC11567118 DOI: 10.1002/jgh3.70034] [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: 04/29/2024] [Revised: 07/25/2024] [Accepted: 09/16/2024] [Indexed: 11/19/2024]
Abstract
Background and Aim Endoscopic submucosal dissection (ESD) has become the treatment of choice for many superficial gastric neoplasms. Clinical outcomes are increasingly comparable between Japanese and Western series; however, data are lacking on the validity of risk stratification tools in Western cohorts. We aimed to evaluate clinical outcomes, explore risk stratification, and compare our data with published Western series. Methods We conducted a retrospective, observational cohort study in a single tertiary referral center over a 13-year period. Primary outcomes were rates of en bloc, complete (R0) and curative resection. Secondary outcomes included adverse events, recurrence, metachronous lesions, eCura grades, and ESGE criteria. A comparative analysis was performed with existing published series from Western centers. Results Totally 112 patients were included in the study cohort. 50.9% were male, 87.5% Caucasian, and median age was 75.5 years (IQR 14.3 years). Lesions were predominantly antral (36.6%) or body (35.7%); median size 20 mm (IQR 15 mm). Rates of en bloc, R0 resection, and curative resection were 96.4%, 89.3%, and 78.6% (identical between eCura and ESGE), respectively. Adverse events occurred in 5.8%, recurrence in 0%, and metachronous lesions in 9.9%. Our data compared favorably with a review existing Western series, which illustrates increasing adoption of ESD and stable outcomes over time. Conclusion ESD represents a safe and effective method of treatment for gastric neoplasia in the Western setting. This study highlights the potential for excellent outcomes in a single center with a heterogeneous patient cohort and supports the use of eCura in guiding post procedural management.
Collapse
Affiliation(s)
- Ciaran Judge
- Department of GastroenterologyRoyal Perth HospitalPerthWestern AustraliaAustralia
| | - Abir Halder
- Department of GastroenterologyRoyal Perth HospitalPerthWestern AustraliaAustralia
| | - Puraskar Pateria
- Department of GastroenterologyRoyal Perth HospitalPerthWestern AustraliaAustralia
| | - Tzeng Khor
- Department of Anatomical PathologyRoyal Perth HospitalPerthWestern AustraliaAustralia
| | - Niroshan Muwanwella
- Department of GastroenterologyRoyal Perth HospitalPerthWestern AustraliaAustralia
| | - Marcus Chin
- Department of GastroenterologyRoyal Perth HospitalPerthWestern AustraliaAustralia
| | - Krish Ragunath
- Department of GastroenterologyRoyal Perth HospitalPerthWestern AustraliaAustralia
- Faculty of Health Sciences, Curtin Medical SchoolCurtin UniversityPerthWestern AustraliaAustralia
| |
Collapse
|
28
|
Latorre G, Espino A, Orr CE, Bechara R. Endoscopic Submucosal Dissection of Gastric High-Grade Foveolar Dysplasia With Normal Background Mucosa. GASTRO HEP ADVANCES 2024; 4:100565. [PMID: 39866717 PMCID: PMC11758418 DOI: 10.1016/j.gastha.2024.10.005] [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: 09/22/2024] [Accepted: 10/05/2024] [Indexed: 01/28/2025]
Abstract
Most gastric neoplastic lesions appear in patients with gastric premalignant conditions. Here, we present the case of a 75-year-old woman with no prior history of Helicobacter pylori infection, with a big gastric adenoma resected by endoscopic submucosal dissection. Histopathological examination revealed high-grade foveolar dysplasia. Interestingly, surrounding mucosa was normal, without signs of H. pylori infection or gastric preneoplastic conditions. The presented case emphasizes that high-risk gastric lesions may be present within a normal stomach without endoscopic signs of H. pylori infection or premalignant conditions. This underscores the importance of careful examination in regular practice of esophagogastroduodenoscopy, even in low-risk patients.
Collapse
Affiliation(s)
- Gonzalo Latorre
- Department of Gastroenterology, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Alberto Espino
- Department of Gastroenterology, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Christine E. Orr
- Department of Pathology and Molecular Medicine, Queen’s University, Kingston Health Sciences Centre, Kingston, Ontario, Canada
| | - Robert Bechara
- Division of Gastroenterology, Kingston Health Sciences Centre, Queen's University, Kingston General Hospital, Kingston, Ontario, Canada
| |
Collapse
|
29
|
Dottori L, Palumbo C, Dilaghi E, Pivetta G, Ligato I, Esposito G, Pilozzi E, Annibale B, Lahner E. Antral mucosa healing at long-term follow-up in patients with corpus atrophic gastritis and concomitant antral gastritis may mimic autoimmune gastritis. Dig Liver Dis 2024:S1590-8658(24)01015-6. [PMID: 39379226 DOI: 10.1016/j.dld.2024.09.017] [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: 08/05/2024] [Revised: 09/09/2024] [Accepted: 09/15/2024] [Indexed: 10/10/2024]
Abstract
BACKGROUND AND AIM Corpus atrophic gastritis (CAG) is defined as autoimmune when the antrum is spared, representing this element a crucial diagnostic criterium of autoimmune gastritis. In contrast, CAG with concomitant antral gastritis (AG), atrophic or non-atrophic, is generally attributed to H. pylori infection. During the natural history of CAG, possible antrum healing has been supposed. The current study aimed to assess the antral mucosa histopathological changes at long-term follow-up (FU) with respect to baseline in patients with CAG and concomitant atrophic or non-atrophic gastritis AG. METHODS Retrospective study on 130 patients with histologically diagnosed CAG with atrophic or non-atrophic AG. Mean FU gastroscopy was at 40.6 (range 4-192) months. Patients with confirmed CAG (n = 117; median age 66, range 20-87 years; 67.5 % F) were finally included. At baseline, 47 (40.2 %) had non-atrophic and 70 (59.8 %) atrophic AG. Helicobacter pylori (Hp) infection was present at histology in 27.3 % of patients, all treated. RESULTS At FU, 30/117(25.6 %) patients showed a complete antral healing; 11/29(37.9 %) were Hp positive at baseline, cured in all but one. Atrophic AG regressed in 16/70(22.8 %) patients. Both, antral healing and regression of antral AG, were found to be similar in Hp-cured and not-cured/ naïve-negatives patients (p > 0.05). CONCLUSION In a subset of CAG patients, AG may regress at long-term FU irrespective of Hp cure, thus mimicking autoimmune atrophic gastritis and raising concerns about its current histopathological diagnostic criteria.
Collapse
Affiliation(s)
- Ludovica Dottori
- Department of Medical-Surgical Sciences and Translational Medicine, Sant'Andrea Hospital, Sapienza University of Rome, Italy
| | - Carla Palumbo
- Department of Medical-Surgical Sciences and Translational Medicine, Sant'Andrea Hospital, Sapienza University of Rome, Italy
| | - Emanuele Dilaghi
- Department of Medical-Surgical Sciences and Translational Medicine, Sant'Andrea Hospital, Sapienza University of Rome, Italy
| | - Giulia Pivetta
- Department of Medical-Surgical Sciences and Translational Medicine, Sant'Andrea Hospital, Sapienza University of Rome, Italy
| | - Irene Ligato
- Department of Medical-Surgical Sciences and Translational Medicine, Sant'Andrea Hospital, Sapienza University of Rome, Italy
| | - Gianluca Esposito
- Department of Medical-Surgical Sciences and Translational Medicine, Sant'Andrea Hospital, Sapienza University of Rome, Italy
| | - Emanuela Pilozzi
- Department of Clinical and Molecular Medicine, Sant'Andrea Hospital, Sapienza University of Rome, Italy
| | - Bruno Annibale
- Department of Medical-Surgical Sciences and Translational Medicine, Sant'Andrea Hospital, Sapienza University of Rome, Italy
| | - Edith Lahner
- Department of Medical-Surgical Sciences and Translational Medicine, Sant'Andrea Hospital, Sapienza University of Rome, Italy.
| |
Collapse
|
30
|
Farinati F, Pelizzaro F. Gastric cancer screening in Western countries: A call to action. Dig Liver Dis 2024; 56:1653-1662. [PMID: 38403513 DOI: 10.1016/j.dld.2024.02.008] [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: 01/25/2024] [Revised: 02/11/2024] [Accepted: 02/12/2024] [Indexed: 02/27/2024]
Abstract
Gastric cancer is a major cause of cancer-related death worldwide, despite the reduction in its incidence. The disease is still burdened with a poor prognosis, particularly in Western countries. The main risk factor is the infection by Helicobacter pylori, classified as a class I carcinogen by the IARC, and It is well-known that primary prevention of gastric cancer can be achieved with the eradication of the infection. Moreover, non-invasive measurement of pepsinogens (PGI and PGI/PGII ratio) allows the identification of patients that should undergo upper gastrointestinal (GI) endoscopy. Gastric non-cardia adenocarcinoma is indeed preceded by a well-defined precancerous process that involves consecutive stages, described for the first time by Correa et al. more than 40 years ago, and patients with advance stages of gastric atrophy/intestinal metaplasia and with dysplastic changes should be followed-up periodically with upper GI endoscopies. Despite these effective screening and surveillance methods, national-level screening campaigns have been adopted only in few countries in eastern Asia (Japan and South Korea). In this review, we describe primary and secondary preventive measures for gastric cancer, discussing the need to introduce screening also in Western countries. Moreover, we propose a simple algorithm for screening that could be easily applied in clinical practice.
Collapse
Affiliation(s)
- Fabio Farinati
- Department of Surgery, Oncology and Gastroenterology, University of Padova, Via Giustiniani 2, Padova 35128, Italy; Gastroenterology Unit, Azienda Ospedale-Università di Padova, Via Giustiniani 2, Padova 35128, Italy.
| | - Filippo Pelizzaro
- Department of Surgery, Oncology and Gastroenterology, University of Padova, Via Giustiniani 2, Padova 35128, Italy; Gastroenterology Unit, Azienda Ospedale-Università di Padova, Via Giustiniani 2, Padova 35128, Italy
| |
Collapse
|
31
|
Tang Q, Wang R, Niu H, Li Y, Li Y, Hu Z, Liu X, Tao Y. Mapping network connection and direction among symptoms of depression and anxiety in patients with chronic gastritis. Psych J 2024; 13:824-834. [PMID: 38616130 PMCID: PMC11444727 DOI: 10.1002/pchj.757] [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: 12/27/2023] [Accepted: 03/15/2024] [Indexed: 04/16/2024]
Abstract
Regarding neurophysiological and developmental findings, anxiety and depression are usual comorbidities of gastritis patients. However, research related to anxiety and depression among chronic gastritis patients was conducted on the disease level while ignoring symptoms. Hence, we rendered the network approach to reveal the symptoms of anxiety and depression among chronic gastritis patients. Three hundred and sixty-nine chronic gastritis patients (female = 139, Mage = 55.87 years) were asked to complete the Self-Rating Anxiety Scale and Self-Rating Depression Scale. Three symptom networks and one directed acyclic graph (DAG) network were formed. First, in the anxiety network of chronic gastritis patients, dizziness was the most influential symptom. In the depression network of chronic gastritis patients, depressed affect and psychomotor retardation were the influential symptoms. Second, panic, easy fatiguability, weakness, palpitation, depressed affect, tachycardia, fatigue, and psychomotor agitation bridged the anxiety-depression network of chronic gastritis patients. Third, DAG networks showed that anxiousness and hopelessness could trigger other symptoms in the anxiety-depression networks of chronic gastritis patients. The current study provided insightful information on patients with chronic gastritis by examining the structures of symptoms.
Collapse
Affiliation(s)
- Qihui Tang
- Faculty of Psychology, Beijing Normal University, Beijing, China
- Beijing Key Laboratory of Applied Experimental Psychology, National Demonstration Center for Experimental Psychology Education, Beijing Normal University, Beijing, China
| | - Rui Wang
- Department of Gastroenterology, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, China
| | - Haiqun Niu
- School of Psychology, Nanjing Normal University, Nanjing, China
| | - Yifang Li
- Department of Chinese Medicine Nursing, School of Nursing, Anhui University of Chinese Medicine, Hefei, China
| | - Yuting Li
- Department of Chinese Medicine Nursing, School of Nursing, Anhui University of Chinese Medicine, Hefei, China
- School of Psychology, Shanghai University of Sport, Shanghai, China
| | - Zichao Hu
- Department of Gastroenterology, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, China
| | - Xiangping Liu
- Faculty of Psychology, Beijing Normal University, Beijing, China
- Beijing Key Laboratory of Applied Experimental Psychology, National Demonstration Center for Experimental Psychology Education, Beijing Normal University, Beijing, China
| | - Yanqiang Tao
- Faculty of Psychology, Beijing Normal University, Beijing, China
- Beijing Key Laboratory of Applied Experimental Psychology, National Demonstration Center for Experimental Psychology Education, Beijing Normal University, Beijing, China
| |
Collapse
|
32
|
Burke E, Harkins P, Arumugasamy M. Incidence of Gastric Adenocarcinoma in Those With Gastric Atrophy: A Systematic Review. Cureus 2024; 16:e71768. [PMID: 39429990 PMCID: PMC11488155 DOI: 10.7759/cureus.71768] [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] [Accepted: 10/17/2024] [Indexed: 10/22/2024] Open
Abstract
Gastric atrophy (GA), or atrophic gastritis, is a pre-neoplastic lesion of gastric cancer (GC). It is part of the Correa cascade, which culminates in intestinal-type gastric adenocarcinoma. The cascade posits that intestinal-type gastric adenocarcinoma develops along a defined pathway of pre-neoplastic stages. The cascade begins with chronic gastritis, most commonly caused by Helicobacter pylori (H. pylori) infection, and proceeds through GA, gastric intestinal metaplasia (GIM), both complete and incomplete, dysplasia, both low and high-grade, and culminating in intestinal-type gastric adenocarcinoma. Attempts in Europe have been made to identify patients at risk of developing GC and target them with surveillance oesophagogastroduodenoscopy (OGD). However, there remains uncertainty about GA's risk of developing into GC. This poses issues in terms of guiding the need for and determining intervals for surveillance OGDs, which are a costly form of surveillance. As such, we attempted to gather all available studies assessing the risk of GC developing from GA, which is the first step in the Correa cascade. This study was a comprehensive systematic review of published papers, reported per the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) statement. This systematic review, which included a substantial 25,455 patients across 18 studies, found that the relative risk (RR) of GC in those with GA, using standardised incidence ratios as a measure of RR, was 15.1, with a 95% confidence interval ranging from 13.5 to 16.9. We conclude that GA does increase the risk of developing GC, and this risk may be higher than previously appreciated. Further large-scale studies are needed in Western cohorts of patients to precisely define this risk and guide the need for surveillance programs. These future studies must be standardised to account for H. pylori status, the topographical distribution of the GA, and the methods for assessing the degree of GA.
Collapse
Affiliation(s)
- Eoghan Burke
- Surgery, Royal College of Surgeons in Ireland (RCSI), Dublin, IRL
| | - Patricia Harkins
- Medicine, Royal College of Physicians of Ireland (RCPI), Dublin, IRL
| | | |
Collapse
|
33
|
Hahn AI, Mülder DT, Huang RJ, Zhou MJ, Blake B, Omofuma O, Murphy JD, Gutiérrez-Torres DS, Zauber AG, O'Mahony JF, Camargo MC, Ladabaum U, Yeh JM, Hur C, Lansdorp-Vogelaar I, Meester R, Laszkowska M. Global Progression Rates of Precursor Lesions for Gastric Cancer: A Systematic Review and Meta-Analysis. Clin Gastroenterol Hepatol 2024:S1542-3565(24)00864-4. [PMID: 39362617 PMCID: PMC11958785 DOI: 10.1016/j.cgh.2024.09.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Revised: 08/27/2024] [Accepted: 09/04/2024] [Indexed: 10/05/2024]
Abstract
BACKGROUND & AIMS Whether gastric cancer (GC) precursor lesions progress to invasive cancer at similar rates globally remains unknown. We conducted a systematic review and meta-analysis to determine the progression of precursor lesions to GC in countries with low versus medium/high incidence. METHODS We searched relevant databases for studies reporting the progression of endoscopically confirmed precursor lesions to GC. Studies were stratified by low (<6 per 100,000) or medium/high (≥6 per 100,000) GC incidence countries. Random-effects models were used to estimate the progression rates of atrophic gastritis (AG), intestinal metaplasia (IM), and dysplasia to GC per 1000 person-years. RESULTS Among the 5829 studies identified, 44 met our inclusion criteria. The global pooled estimates of the progression rate per 1000 person-years were 2.09 (95% confidence interval, 1.46-2.99), 2.89 (2.03-4.11), and 10.09 (5.23-19.49) for AG, IM, and dysplasia, respectively. The estimated progression rates per 1000 person-years for low versus medium/high GC incidence countries, respectively, were 0.97 (0.86-1.10) versus 2.47 (1.70-2.99) for AG (P < .01), 2.37 (1.43-3.92) versus 3.47 (2.13-5.65) for IM (P = .29), and 5.51 (2.92-10.39) versus 14.80 (5.87-37.28) for dysplasia (P = .08). There were no differences for progression of AG between groups when high-quality studies were compared. CONCLUSIONS Similar progression rates of IM and dysplasia were observed among low and medium/high GC incidence countries. This suggests that the potential benefits of surveillance for these lesions in low-risk regions may be comparable with those of population-wide interventions in high-risk regions. Further prospective studies are needed to confirm these findings and inform global screening and surveillance guidelines.
Collapse
Affiliation(s)
- Anne I Hahn
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York.
| | - Duco T Mülder
- Department of Public Health, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Robert J Huang
- Division of Gastroenterology and Hepatology, Stanford University School of Medicine, Stanford, California
| | - Margaret J Zhou
- Division of Gastroenterology and Hepatology, Stanford University School of Medicine, Stanford, California
| | - Benjamin Blake
- Weill Cornell Medical College of Cornell University, New York, New York
| | - Omonefe Omofuma
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, Maryland
| | - John D Murphy
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, Maryland
| | | | - Ann G Zauber
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - James F O'Mahony
- Department of Public Health, Erasmus Medical Center, Rotterdam, the Netherlands; School of Economics, University College Dublin, Dublin, Ireland
| | - M Constanza Camargo
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, Maryland
| | - Uri Ladabaum
- Division of Gastroenterology and Hepatology, Stanford University School of Medicine, Stanford, California
| | - Jennifer M Yeh
- Department of Pediatrics, Harvard Medical School, Boston Children's Hospital, Boston, Massachusetts
| | - Chin Hur
- Division of General Medicine, Department of Medicine, Columbia University Irving Medical Center, New York, New York; Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, New York
| | | | - Reinier Meester
- Department of Public Health, Erasmus Medical Center, Rotterdam, the Netherlands; Health Economics & Outcomes Research, Freenome Holdings Inc, San Francisco, California
| | - Monika Laszkowska
- Gastroenterology, Hepatology, and Nutrition Service, Department of Subspecialty Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| |
Collapse
|
34
|
Jin X, Zhou Q, Lyu B, Zhang C, Huang L. Ability of detection in different resolution endoscopy for upper gastrointestinal mucosal lesions. Surg Endosc 2024; 38:5903-5913. [PMID: 39168859 PMCID: PMC11458703 DOI: 10.1007/s00464-024-11186-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Accepted: 08/10/2024] [Indexed: 08/23/2024]
Abstract
BACKGROUND Most endoscopists believe that higher resolution improves lesion detection rates. However, existing studies primarily compared the detection rates of white light endoscopy (WLE) and other imaging modalities. Our previous study demonstrated the advantages of magnifying endoscopy from general endoscopy for lesion detection, prompting further investigation into the variations in lesion detection rates across endoscopes with different resolutions. METHODS Endoscopic and corresponding pathological data from our medical unit over the past 5 years were analyzed. We excluded specific-purpose endoscopic procedures to ensure the natural randomization of the data. Baseline adjustment and risk factor analyses used multi-group propensity score matching and logistic regression. RESULTS The overall lesion detection rate was significantly higher with high-quality endoscopy (Q-endoscopy) compared to high-definition endoscopy (H-endoscopy) and high definition and quality endoscopy (HQ-endoscopy) (34.4% vs. 30.2% vs. 29.6%, P = 0.001). Similar results were observed for elevated lesions (25.7% vs. 21.0% vs. 22.9%, P = 0.001) and depressed lesions (6.6% vs. 6.2% vs. 3.6%, P < 0.001). HQ-endoscopy had a superior detection rate for superficial lesions compared to both H- and Q-endoscopies (3.0% vs. 2.8% vs. 1.8%, P = 0.041). However, there were no significant differences in neoplastic detection rate or missed neoplastic lesion rate among the three groups. CONCLUSION Q-endoscopy is superior in detecting non-superficial lesions, while HQ-endoscopy is better at detecting superficial lesions. However, there were no statistically significant differences in detecting or omitting neoplastic lesions among the three endoscopic examinations.
Collapse
Affiliation(s)
- Xiaoliang Jin
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Qiujun Zhou
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Bin Lyu
- Department of Gastroenterology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, 310060, China
| | - Chunli Zhang
- Department of Pathology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, 310060, China
| | - Liang Huang
- Department of Endoscopy Center, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Youdian Street 54, Hangzhou, 310060, China.
| |
Collapse
|
35
|
Jannot AS, Girardeau Y, Chaussade S, Cerf-Bensussan N, Malamut G. Increased risk of gastric cancer in relation with pernicious anaemia in patients with primary antibody deficiency: A nationwide case control study. Dig Liver Dis 2024; 56:1760-1765. [PMID: 38853087 DOI: 10.1016/j.dld.2024.05.032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 05/27/2024] [Accepted: 05/28/2024] [Indexed: 06/11/2024]
Abstract
BACKGROUND/AIM We aimed to assess gastrointestinal cancers risks in a large cohort of individuals with primary antibody deficiency (PAD) and their association with risk of autoimmune and inflammatory gastrointestinal diseases. METHODS Investigating a French national database of inpatient admissions between 2010 and 2018, we identified 12,748 patients with PAD and 38,244 control non-exposed individuals. We performed multiple exposed-non-exposed studies using conditional logistic regression. RESULTS In comparison with non-exposed patients, PAD patients had increased risk of in situ gastric carcinoma (Odds Ratio (OR) =10.5 [95 % CI 2.2; 50.5]), malignant gastric tumor (OR=3.2 [95 % CI 2.2; 4.4]) and colorectal cancer (OR=1.2 [95 % CI 1; 1.5]). PAD patients had also increased risk of pernicious anaemia (OR=8 |95 % CI 5.6; 11.5]), Crohn's disease (OR= 4.4 [95 % CI 3.5; 5.6]), ulcerative colitis (OR=2.9 [95 % CI 2.4; 3.6]) and coeliac disease (OR=13.3 [95 % CI 9.1; 19.5]). Within patients with gastric cancer, those with PAD had increased risk of pernicious anaemia (OR=8.4 [95 % CI 1.5; 215]; p = 0.01) but not of H. pylori infection. CONCLUSIONS Risk of gastric cancer is particularly high in PAD patients and notably risk of in situ gastric carcinoma in association with pernicious anaemia. It supports indication of early endoscopic screening in these patients.
Collapse
Affiliation(s)
- Anne-Sophie Jannot
- French National Rare Disease Registry (BNDMR), Greater Paris University Hospitals (AP-HP), Université Paris Cité, Paris, France; Université Paris Cité, HeKA, INRIA Paris, Inserm, Centre de Recherche des Cordeliers- Université Paris Cité, Paris, France
| | - Yannick Girardeau
- Department of Clinical Investigation and Clinical Epidemiology, AP-HP-Centre-Université Paris Cité, Hôpital Européen Georges Pompidou, Paris, France
| | - Stanislas Chaussade
- Department of Gastroenterology, AP-HP. Centre- Université Paris Cité, Hôpital Cochin, Paris, France
| | - Nadine Cerf-Bensussan
- Université de Paris, INSERM UMR 1163 and Imagine Institute, Laboratory of Intestinal Immunity, Université Paris Cité, Paris, France
| | - Georgia Malamut
- Department of Gastroenterology, AP-HP. Centre- Université Paris Cité, Hôpital Cochin, Paris, France; Université de Paris, INSERM UMR 1163 and Imagine Institute, Laboratory of Intestinal Immunity, Université Paris Cité, Paris, France.
| |
Collapse
|
36
|
Vogli S, Markopoulos P, Filippakou A, Stanc G, Tsironi E, Telakis E. Spontaneous Regression of a Large Gastric Adenoma Following Gynecologic Surgery. Cureus 2024; 16:e71020. [PMID: 39507201 PMCID: PMC11540251 DOI: 10.7759/cureus.71020] [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] [Accepted: 10/05/2024] [Indexed: 11/08/2024] Open
Abstract
Gastric adenomas are defined as polypoid lesions of neoplastic epithelium in the stomach. They are rare, occurring much less frequently than fundic gland polyps and hyperplastic polyps, and are typically associated with mucosal atrophy and intestinal metaplasia. Gastric adenomas also carry a risk of malignant transformation. We report a case of a 66-year-old woman with a gastric adenoma of the corpus found during a preoperative esophagogastroduodenoscopy before abdominal surgery for an ovarian tumor. The patient demonstrated spontaneous regression of her gastric adenoma 14 months after her initial endoscopy and subsequent hysterectomy with salpingo-oophorectomy without undergoing endoscopic resection or any other intervention. To our knowledge, this is the first well-documented case of spontaneous regression of a gastric adenoma.
Collapse
Affiliation(s)
- Stamatina Vogli
- Department of Gastroenterology, Metaxa Cancer Hospital of Piraeus, Piraeus, GRC
| | | | | | - Gabriela Stanc
- Department of Pathology, Metaxa Cancer Hospital of Piraeus, Piraeus, GRC
| | - Eftychia Tsironi
- Department of Gastroenterology, Metaxa Cancer Hospital of Piraeus, Piraeus, GRC
| | - Emmanouil Telakis
- Department of Gastroenterology, Metaxa Cancer Hospital of Piraeus, Piraeus, GRC
| |
Collapse
|
37
|
Kővári B, Carneiro F, Lauwers GY. Epithelial tumours of the stomach. MORSON AND DAWSON'S GASTROINTESTINAL PATHOLOGY 2024:227-286. [DOI: 10.1002/9781119423195.ch13] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
38
|
Xu B, Shi Y, Yuan C, Wang Z, Chen Q, Wang C, Chai J. Integrated gene-metabolite association network analysis reveals key metabolic pathways in gastric adenocarcinoma. Heliyon 2024; 10:e37156. [PMID: 39319160 PMCID: PMC11419903 DOI: 10.1016/j.heliyon.2024.e37156] [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: 03/05/2024] [Revised: 07/22/2024] [Accepted: 08/28/2024] [Indexed: 09/26/2024] Open
Abstract
Gastric adenocarcinoma is one of the most death cause cancers worldwide. Metabolomics is an effective approach for investigating the occurrence and progression of cancer and detecting prognostic biomarkers by studying the profiles of small bioactive molecules. To fully decipher the functional roles of the disrupted metabolites that modulate the cellular mechanism of gastric cancer, integrated gene-metabolite association network methods are critical to map the associations between metabolites and genes. In this study, we constructed a knowledge-based gene-metabolite association network of gastric cancer using the dysregulated metabolites and genes between gastric cancer patients and control group. The topological pathway analysis and gene-protein-metabolite-disease association analysis revealed four key gene-metabolite pathways which include eleven metabolites associated with modulated genes. The integrated gene-metabolite association network enables mechanistic investigation and provides a comprehensive overview regarding the investigation of molecular mechanisms of gastric cancer, which facilitates the in-depth understanding of metabolic biomarker roles in gastric cancer.
Collapse
Affiliation(s)
- Botao Xu
- Department of Gastrointestinal Surgery, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Science, Jinan, China
| | - Yuying Shi
- Department of Biostatistics, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
- National Institute of Health Data Science of China, Shandong University, Jinan, 250000, China
- National Science Library (Chengdu), Chinese Academy of Sciences, Chengdu, 610299, China
| | - Chuang Yuan
- Department of Biochemistry and Biophysics, School of Basic Medical Sciences, Peking University, Beijing, 100191, China
| | - Zhe Wang
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Science, Jinan, Shandong, China
| | - Qitao Chen
- Department of Biostatistics, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
- National Institute of Health Data Science of China, Shandong University, Jinan, 250000, China
| | - Cheng Wang
- Department of Biostatistics, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
- National Institute of Health Data Science of China, Shandong University, Jinan, 250000, China
| | - Jie Chai
- Department of Gastrointestinal Surgery, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Science, Jinan, China
| |
Collapse
|
39
|
Dinis-Ribeiro M, Shah S, El-Serag H, Banks M, Uedo N, Tajiri H, Coelho LG, Libanio D, Lahner E, Rollan A, Fang JY, Moreira L, Bornschein J, Malfertheiner P, Kuipers EJ, El-Omar EM. The road to a world-unified approach to the management of patients with gastric intestinal metaplasia: a review of current guidelines. Gut 2024; 73:1607-1617. [PMID: 39122364 DOI: 10.1136/gutjnl-2024-333029] [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: 06/05/2024] [Accepted: 07/14/2024] [Indexed: 08/12/2024]
Abstract
OBJECTIVE During the last decade, the management of gastric intestinal metaplasia (GIM) has been addressed by several distinct international evidence-based guidelines. In this review, we aimed to synthesise these guidelines and provide clinicians with a global perspective of the current recommendations for managing patients with GIM, as well as highlight evidence gaps that need to be addressed with future research. DESIGN We conducted a systematic review of the literature for guidelines and consensus statements published between January 2010 and February 2023 that address the diagnosis and management of GIM. RESULTS From 426 manuscripts identified, 16 guidelines were assessed. There was consistency across guidelines regarding the purpose of endoscopic surveillance of GIM, which is to identify prevalent neoplastic lesions and stage gastric preneoplastic conditions. The guidelines also agreed that only patients with high-risk GIM phenotypes (eg, corpus-extended GIM, OLGIM stages III/IV, incomplete GIM subtype), persistent refractory Helicobacter pylori infection or first-degree family history of gastric cancer should undergo regular-interval endoscopic surveillance. In contrast, low-risk phenotypes, which comprise most patients with GIM, do not require surveillance. Not all guidelines are aligned on histological staging systems. If surveillance is indicated, most guidelines recommend a 3-year interval, but there is some variability. All guidelines recommend H. pylori eradication as the only non-endoscopic intervention for gastric cancer prevention, while some offer additional recommendations regarding lifestyle modifications. While most guidelines allude to the importance of high-quality endoscopy for endoscopic surveillance, few detail important metrics apart from stating that a systematic gastric biopsy protocol should be followed. Notably, most guidelines comment on the role of endoscopy for gastric cancer screening and detection of gastric precancerous conditions, but with high heterogeneity, limited guidance regarding implementation, and lack of robust evidence. CONCLUSION Despite heterogeneous populations and practices, international guidelines are generally aligned on the importance of GIM as a precancerous condition and the need for a risk-stratified approach to endoscopic surveillance, as well as H. pylori eradication when present. There is room for harmonisation of guidelines regarding (1) which populations merit index endoscopic screening for gastric cancer and GIM detection/staging; (2) objective metrics for high-quality endoscopy; (3) consensus on the need for histological staging and (4) non-endoscopic interventions for gastric cancer prevention apart from H. pylori eradication alone. Robust studies, ideally in the form of randomised trials, are needed to bridge the ample evidence gaps that exist.
Collapse
Affiliation(s)
- Mario Dinis-Ribeiro
- Department of Gastroenterology, Porto Comprehensive Cancer Center & RISE@CI-IPO, University of Porto, Porto, Portugal
- MEDCIDS (Department of Community Medicine, Health Information, and Decision), University of Porto, Porto, Portugal
| | - Shailja Shah
- Division of Gastroenterology, University of California and Jennifer Moreno Veterans Affairs San Diego Healthcare System, San Diego, California, USA
| | - Hashem El-Serag
- Gastroenterology and Hepatology, Baylor College of Medicine, Houston, Texas, USA
| | - Matthew Banks
- University College London Hospital, University College London Hospitals NHS Foundation Trust, London, UK
| | - Noriya Uedo
- Gastrointestinal Oncology, Osaka International Cancer Institute, Osaka, Japan
| | - Hisao Tajiri
- Endoscopy, The Jikei University School of Medicine, Tokyo, Japan
| | - Luiz Gonzaga Coelho
- Instituto Alfa de Gastrenterologia, Hospital das Clínicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Diogo Libanio
- Department of Gastroenterology, Porto Comprehensive Cancer Center & RISE@CI-IPO, University of Porto, Porto, Portugal
- MEDCIDS (Department of Community Medicine, Health Information, and Decision), University of Porto, Porto, Portugal
| | - Edith Lahner
- Department of Medical-Surgical Sciences and Translational Medicine, Sant'Andrea Hospital, Rome, Italy
| | - Antonio Rollan
- Facultad de Medicina Clinica Alemana-Universidad del Desarrollo, Santiago, Chile
| | - Jing-Yuan Fang
- Division of Gastroenterology and Hepatology, Shanghai Institute of Digestive Disease, Shanghai, China
| | - Leticia Moreira
- Gastroenterology, Hospital Clinic de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Jan Bornschein
- MRC Translational Immune Discovery Unit, MRC Weatherall Institute of Molecular Medicine, John Radcliffe Hospital, University of Oxford, Oxford, UK
| | | | - Ernst J Kuipers
- Medical Department II, LMU University Clinic, München, Germany
| | - Emad M El-Omar
- UNSW Microbiome Research Centre, University of New South Wales, Sydney, New South Wales, Australia
| |
Collapse
|
40
|
Latorre G, Silva F, Montero I, Bustamante M, Dukes E, Uribe J, Corsi Sotelo O, Reyes D, Fuentes-López E, Pizarro M, Medel P, Torres J, Roa JC, Pizarro S, Achurra P, Donoso A, Wichmann I, Corvalán AH, Chahuan J, Candia R, Agüero C, Gonzalez R, Vargas JI, Espino A, Camargo MC, Shah SC, Riquelme A. Comparison of OLGA and OLGIM as predictors of gastric cancer in a Latin American population: the ECHOS Study. Gut 2024; 73:e18. [PMID: 38148138 DOI: 10.1136/gutjnl-2023-331059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2023] [Accepted: 12/15/2023] [Indexed: 12/28/2023]
Affiliation(s)
- Gonzalo Latorre
- Department of Gastroenterology, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Felipe Silva
- Department of Gastroenterology, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Isabella Montero
- Department of Gastroenterology, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Miguel Bustamante
- Department of Gastroenterology, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Eitan Dukes
- Department of Gastroenterology, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Javier Uribe
- Department of Gastroenterology, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Oscar Corsi Sotelo
- Department of Gastroenterology, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Diego Reyes
- Department of Gastroenterology, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Eduardo Fuentes-López
- Department of Health Sciences, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Margarita Pizarro
- Department of Gastroenterology, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Patricio Medel
- Pharmacology and Toxicology Program, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Javiera Torres
- Department of Pathology, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Juan Carlos Roa
- Department of Pathology, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
- Centro para la Prevención y el Control del Cáncer (CECAN), Santiago, Chile
| | - Sebastián Pizarro
- Department of Pathology, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Pablo Achurra
- Department of Digestive Surgery, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Andrés Donoso
- Department of Digestive Surgery, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Ignacio Wichmann
- Department of Obstetrics, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
- Division of Oncology, Department of Medicine, Stanford University School of Medicine, Stanford, California, USA
- Advanced Center for Chronic Diseases (ACCDiS), Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Alejandro H Corvalán
- Advanced Center for Chronic Diseases (ACCDiS), Pontificia Universidad Católica de Chile, Santiago, Chile
- Department of Hematology & Oncology, Pontificia Universidad Catolica de Chile, Santiago, Chile
| | - Javier Chahuan
- Department of Gastroenterology, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Roberto Candia
- Department of Gastroenterology, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Carlos Agüero
- Department of Gastroenterology, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Robinson Gonzalez
- Department of Gastroenterology, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Jose Ignacio Vargas
- Department of Gastroenterology, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Alberto Espino
- Department of Gastroenterology, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - M Constanza Camargo
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Shailja C Shah
- Gastroenterology Section, Veterans Affairs, San Diego Healthcare System, San Diego, California, USA
- Division of Gastroenterology, University of California San Diego, La Jolla, California, USA
| | - Arnoldo Riquelme
- Department of Gastroenterology, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
- Centro para la Prevención y el Control del Cáncer (CECAN), Santiago, Chile
| |
Collapse
|
41
|
Xu L, Lyu J, Zheng X, Wang A. Risk Prediction Models for Gastric Cancer: A Scoping Review. J Multidiscip Healthc 2024; 17:4337-4352. [PMID: 39257385 PMCID: PMC11385365 DOI: 10.2147/jmdh.s479699] [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: 05/24/2024] [Accepted: 08/27/2024] [Indexed: 09/12/2024] Open
Abstract
Background Gastric cancer is a significant contributor to the global cancer burden. Risk prediction models aim to estimate future risk based on current and past information, and can be utilized for risk stratification in population screening programs for gastric cancer. This review aims to explore the research design of existing models, as well as the methods, variables, and performance of model construction. Methods Six databases were searched through to November 4, 2023 to identify appropriate studies. PRISMA extension for scoping reviews and the Arksey and O'Malley framework were followed. Data sources included PubMed, Embase, Web of Science, CNKI, Wanfang, and VIP, focusing on gastric cancer risk prediction model studies. Results A total of 29 articles met the inclusion criteria, from which 28 original risk prediction models were identified that met the analysis criteria. The risk prediction model is screened, and the data extracted includes research characteristics, prediction variables selection, model construction methods and evaluation indicators. The area under the curve (AUC) of the models ranged from 0.560 to 0.989, while the C-statistics varied between 0.684 and 0.940. The number of predictor variables is mainly concentrated between 5 to 11. The top 5 most frequently included variables were age, helicobacter pylori (Hp), precancerous lesion, pepsinogen (PG), sex, and smoking. Age and Hp were the most consistently included variables. Conclusion This review enhances understanding of current gastric cancer risk prediction research and its future directions. The findings provide a strong scientific basis and technical support for developing more accurate gastric cancer risk models. We expect that these conclusions will point the way for future research and clinical practice in this area to assist in the early prevention and treatment of gastric cancer.
Collapse
Affiliation(s)
- Linyu Xu
- Department of Public Service, The First Affiliated Hospital of China Medical University, Shenyang, 110001, People’s Republic of China
| | - Jianxia Lyu
- Department of Public Service, The First Affiliated Hospital of China Medical University, Shenyang, 110001, People’s Republic of China
| | - Xutong Zheng
- Department of Public Service, The First Affiliated Hospital of China Medical University, Shenyang, 110001, People’s Republic of China
| | - Aiping Wang
- Department of Public Service, The First Affiliated Hospital of China Medical University, Shenyang, 110001, People’s Republic of China
| |
Collapse
|
42
|
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
|
43
|
Niu W, Liu L, Dong Z, Bu X, Yao F, Wang J, Wu X, Chen C, Mao T, Wu Y, Yuan L, Wan X, Zhou H. A deep learning model based on magnifying endoscopy with narrow-band imaging to evaluate intestinal metaplasia grading and OLGIM staging: A multicenter study. Dig Liver Dis 2024; 56:1565-1571. [PMID: 38402085 DOI: 10.1016/j.dld.2024.02.001] [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: 11/05/2023] [Revised: 02/04/2024] [Accepted: 02/05/2024] [Indexed: 02/26/2024]
Abstract
BACKGROUND AND PURPOSE Patients with stage III or IV of operative link for gastric intestinal metaplasia assessment (OLGIM) are at a higher risk of gastric cancer (GC). We aimed to construct a deep learning (DL) model based on magnifying endoscopy with narrow-band imaging (ME-NBI) to evaluate OLGIM staging. METHODS This study included 4473 ME-NBI images obtained from 803 patients at three endoscopy centres. The endoscopic expert marked intestinal metaplasia (IM) regions on endoscopic images of the target biopsy sites. Faster Region-Convolutional Neural Network model was used to grade IM lesions and predict OLGIM staging. RESULTS The diagnostic performance of the model for IM grading in internal and external validation sets, as measured by the area under the curve (AUC), was 0.872 and 0.803, respectively. The accuracy of this model in predicting the high-risk stage of OLGIM was 84.0%, which was not statistically different from that of three junior (71.3%, p = 0.148) and three senior endoscopists (75.3%, p = 0.317) specially trained in endoscopic images corresponding to pathological IM grade, but higher than that of three untrained junior endoscopists (64.0%, p = 0.023). CONCLUSION This DL model can assist endoscopists in predicting OLGIM staging using ME-NBI without biopsy, thereby facilitating screening high-risk patients for GC.
Collapse
Affiliation(s)
- Wenlu Niu
- Department of Gastroenterology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Leheng Liu
- Department of Gastroenterology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai Key Laboratory of Pancreatic Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhixia Dong
- Department of Gastroenterology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Xiongzhu Bu
- School of Mechanical Engineering, Nanjing University of Science and Technology, Nanjing, China
| | - Fanghao Yao
- School of Mechanical Engineering, Nanjing University of Science and Technology, Nanjing, China
| | - Jing Wang
- Department of Pathology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaowan Wu
- Department of Gastroenterology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Congying Chen
- Department of Gastroenterology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Tiancheng Mao
- Department of Gastroenterology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai Key Laboratory of Pancreatic Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yulun Wu
- Department of Gastroenterology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai Key Laboratory of Pancreatic Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lin Yuan
- Department of Pathology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xinjian Wan
- Department of Gastroenterology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China.
| | - Hui Zhou
- Department of Gastroenterology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai Key Laboratory of Pancreatic Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
44
|
Zhao J, Tian W, Zhang X, Dong S, Shen Y, Gao X, Yang M, Lv J, Hu F, Han J, Zhan Q, An F. The diagnostic value of serum trefoil factor 3 and pepsinogen combination in chronic atrophic gastritis: a retrospective study based on a gastric cancer screening cohort in the community population. Biomarkers 2024; 29:384-392. [PMID: 39234749 DOI: 10.1080/1354750x.2024.2400927] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2024] [Accepted: 08/31/2024] [Indexed: 09/06/2024]
Abstract
BACKGROUND Chronic atrophic gastritis (CAG) is an important precursor of gastric cancer(GC), and there is currently a lack of reliable non-invasive diagnostic markers. This study aims to find a biomarker for non-invasive screening of CAG in the community. METHODS A total of 540 individuals were enrolled (test set = 385, validation set = 155). ROC curve analysis was used to evaluate the diagnostic significance of serum Trefoil Factor 3 (TFF3) alone or in combination with pepsinogen (PG) for CAG in the test and validation set. Furthermore, the diagnostic value of TFF3 and PG in different Helicobacter pylori (H. pylori) infection states was studied. RESULTS When compared with chronic superficial gastritis (CSG), the expression level of serum TFF3 in the CAG was higher (27 ng/ml vs 19.61, P < 0.001). ROC curve analysis found that the sensitivity, specificity, and area under the curve (AUC) of CAG diagnosis using serum TFF3 alone at the optimal cut-off value of 26.55 ng/ml were 0.529, 0.87, and 0.739, respectively. When TFF3 was combined with The Ratio of PGI to PGII (PGR), the AUC and specificity reached 0.755 and 0.825, respectively. TFF3 individual or combined with PGR had good predictive value, especially in the H. Pylori negative patients. CONCLUSION TFF3 combined with PGR can effectively predict CAG, especially in patients with H. pylori negative.
Collapse
Affiliation(s)
- Jiamin Zhao
- Department of Gastroenterology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, China
| | - Wenying Tian
- Department of Gastroenterology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, China
| | - Xiaoxue Zhang
- Department of Gastroenterology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, China
| | - Shengrong Dong
- Department of Gastroenterology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, China
| | - Yao Shen
- Department of Gastroenterology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, China
| | - Xiaojuan Gao
- Department of Gastroenterology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, China
| | - Mei Yang
- Department of Gastroenterology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, China
| | - Jiale Lv
- Department of Gastroenterology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, China
| | - Feifan Hu
- Department of Gastroenterology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, China
| | - Jinglue Han
- Department of Gastroenterology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, China
| | - Qiang Zhan
- Department of Gastroenterology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, China
| | - Fangmei An
- Department of Gastroenterology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, China
| |
Collapse
|
45
|
Xu W, McGuinness MJ, Wells C, Varghese C, Elliott B, Paterson L, Collins R, Lill M, Windsor J, Koea J, Panoho J, Walmsley R, Wright D, Parry S, Harmston C. Protocol for a national, multicentre study of post-endoscopy colorectal and upper gastrointestinal cancers: The POET study. Colorectal Dis 2024; 26:1720-1731. [PMID: 38978156 DOI: 10.1111/codi.17057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Accepted: 05/18/2024] [Indexed: 07/10/2024]
Abstract
AIM The primary aim of the study is to define the post-colonoscopy colorectal cancer (PCCRC) three-year rate and the post-endoscopy upper gastrointestinal cancer (PEUGIC) three-year rate across public hospitals in Aotearoa New Zealand. METHOD This retrospective cohort study will be conducted via the trainee-led STRATA Collaborative network. All public hospitals in Aotearoa New Zealand will be eligible to participate. Data will be collected on all adult patients who are diagnosed with colorectal adenocarcinoma within 6 to 48 months of a colonoscopy and all adult patients diagnosed with gastroesophageal cancer within 6 to 48 months of an upper gastrointestinal endoscopy. The study period will be from 2010 to 2022. The primary outcome is the PCCRC 3-year rate and the PEUGIC 3-year rate. Secondary aims are to define and characterize survival after PCCRC or PEUGIC, the cause of PCCRC as based on the World Endoscopy Organization System of Analysis definitions, trends over time, and centre level variation. CONCLUSION This protocol describes the methodology for a nationwide retrospective cohort study on PCCRC and PEUGIC in Aotearoa New Zealand. These data will lay the foundation for future studies and quality improvement initiatives.
Collapse
Affiliation(s)
- William Xu
- Department of Surgery, Whangārei Hospital, Te Whatu Ora, Whangārei, New Zealand
- Department of Surgery, University of Auckland, Auckland, New Zealand
| | - Matthew James McGuinness
- Department of Surgery, University of Auckland, Auckland, New Zealand
- Department of Surgery, North Shore Hospital, Te Whatu Ora, Auckland, New Zealand
| | - Cameron Wells
- Department of Surgery, University of Auckland, Auckland, New Zealand
- Department of Surgery, Hawke's Bay Hospital, Te Whatu Ora, Hastings, New Zealand
| | - Chris Varghese
- Department of Surgery, University of Auckland, Auckland, New Zealand
- Department of Surgery, Middlemore Hospital, Te Whatu Ora, Auckland, New Zealand
| | - Brodie Elliott
- Department of Surgery, Whangārei Hospital, Te Whatu Ora, Whangārei, New Zealand
- Department of Surgery, University of Auckland, Auckland, New Zealand
| | - Luke Paterson
- Department of Surgery, Whangārei Hospital, Te Whatu Ora, Whangārei, New Zealand
| | - Ray Collins
- Department of Surgery, Middlemore Hospital, Te Whatu Ora, Auckland, New Zealand
| | - Marianne Lill
- New Zealand Association of General Surgeons, New Zealand
- Department of Surgery, Whanganui Hospital, Te Whatu Ora, Whanganui, New Zealand
| | - John Windsor
- Department of Surgery, University of Auckland, Auckland, New Zealand
| | - Jonathan Koea
- Department of Surgery, University of Auckland, Auckland, New Zealand
- Department of Surgery, North Shore Hospital, Te Whatu Ora, Auckland, New Zealand
| | - Joy Panoho
- Te Poutokomanawa, Te Whatu Ora, New Zealand
| | - Russell Walmsley
- Department of Surgery, North Shore Hospital, Te Whatu Ora, Auckland, New Zealand
- Department of Gastroenterology, North Shore Hospital, Te Whatu Ora, Auckland, New Zealand
| | - Deborah Wright
- Department of Surgery, Dunedin Hospital, Te Whatu Ora, Dunedin, New Zealand
- Department of Surgery, University of Otago, Dunedin, New Zealand
| | - Susan Parry
- Department of Gastroenterology, Auckland City Hospital, Te Whatu Ora, Auckland, New Zealand
- Department of Medicine, University of Auckland, Auckland, New Zealand
| | - Christopher Harmston
- Department of Surgery, Whangārei Hospital, Te Whatu Ora, Whangārei, New Zealand
- Department of Surgery, University of Auckland, Auckland, New Zealand
| |
Collapse
|
46
|
Kawamura M, Uedo N, Yao K, Koike T, Kanesaka T, Hatta W, Ogata Y, Iwai W, Yokosawa S, Honda J, Asonuma S, Okata H, Ohyauchi M, Ito H, Abe Y, Ara N, Kayaba S, Shinkai H, Kanemitsu T. Endoscopic and histological risk stratification for gastric cancer using gastric intestinal metaplasia. J Gastroenterol Hepatol 2024; 39:1910-1916. [PMID: 38740510 DOI: 10.1111/jgh.16617] [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: 01/03/2024] [Revised: 04/19/2024] [Accepted: 05/02/2024] [Indexed: 05/16/2024]
Abstract
BACKGROUND AND AIM Intestinal metaplasia (IM) of the gastric mucosa is strongly associated with the risk of gastric cancer (GC). This study was performed to investigate the usefulness of endoscopic and histological risk stratification for GC using IM. METHODS This was a post-hoc analysis of a multicenter prospective study involving 10 Japanese facilities (UMINCTR000027023). The ridge/tubulovillous pattern, light blue crest (LBC), white opaque substance (WOS), endoscopic grading of gastric IM (EGGIM) score using non-magnifying image-enhanced endoscopy, and operative link on gastric IM assessment (OLGIM) were evaluated for their associations with GC risk in all patients. RESULTS In total, 380 patients (115 with GC and 265 without GC) were analyzed. The presence of an LBC (limited to antrum: odds ratio [OR] 2.4 [95% confidence interval 1.1-5.0], extended to corpus: OR 3.6 [2.1-6.3]), the presence of WOS (limited to antrum: OR 3.0 [1.7-5.3], extended to corpus: OR 4.2 [2.1-8.2]), and histological IM (limited to antrum: OR 3.2 [1.4-7.4], extended to corpus: OR 8.5 [4.5-16.0]) were significantly associated with GC risk. Additionally, the EGGIM score (5-8 points: OR 8.8 [4.4-16.0]) and OLGIM (stage III/IV: OR 12.5 [6.1-25.8]) were useful for stratification of GC risk. The area under the receiver operating characteristic curve value for GC risk was 0.740 for OLGIM and 0.706 for EGGIM. CONCLUSIONS The LBC, WOS, EGGIM, and OLGIM were strongly associated with GC risk in Japanese patients. This finding can be useful for GC risk assessment in daily clinical practice.
Collapse
Affiliation(s)
- Masashi Kawamura
- Department of Gastroenterology, Sendai City Hospital, Sendai, Japan
| | - Noriya Uedo
- Department of Gastrointestinal Oncology, Osaka International Cancer Institute, Osaka, Japan
| | - Kenshi Yao
- Department of Endoscopy, Fukuoka University Chikushi Hospital, Fukuoka, Japan
| | - Tomoyuki Koike
- Division of Gastroenterology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Takashi Kanesaka
- Department of Gastrointestinal Oncology, Osaka International Cancer Institute, Osaka, Japan
| | - Waku Hatta
- Division of Gastroenterology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Yohei Ogata
- Division of Gastroenterology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Wataru Iwai
- Department of Gastroenterology, Miyagi Cancer Center, Natori, Japan
| | - Satoshi Yokosawa
- Department of Gastroenterology, Iwate Prefectural Iwai Hospital, Iwate, Japan
| | - Junya Honda
- Department of Gastroenterology, Iwate Prefectural Iwai Hospital, Iwate, Japan
| | - Sho Asonuma
- Department of Gastroenterology, South Miyagi Medical Center, Ogawara, Japan
| | - Hideki Okata
- Department of Gastroenterology, South Miyagi Medical Center, Ogawara, Japan
| | - Motoki Ohyauchi
- Department of Gastroenterology, Osaki Citizen Hospital, Ōsaki, Japan
| | - Hirotaka Ito
- Department of Gastroenterology, Osaki Citizen Hospital, Ōsaki, Japan
| | - Yasuhiko Abe
- Division of Endoscopy, Yamagata University Hospital, Yamagata, Japan
| | - Nobuyuki Ara
- Department of Gastroenterology, National Hospital Organization Sendai Medical Center, Sendai, Japan
| | - Shoichi Kayaba
- Department of Gastroenterology, Iwate Prefectural Isawa Hospital, Iwate, Japan
| | - Hirohiko Shinkai
- Department of Gastroenterology, Iwate Prefectural Isawa Hospital, Iwate, Japan
| | - Takao Kanemitsu
- Department of Gastroenterology, Fukuoka University Chikushi Hospital, Fukuoka, Japan
| |
Collapse
|
47
|
Costa D, Ramai D, Tringali A. Novel classification of gastric polyps: The good, the bad and the ugly. World J Gastroenterol 2024; 30:3640-3653. [PMID: 39192997 PMCID: PMC11346164 DOI: 10.3748/wjg.v30.i31.3640] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 07/19/2024] [Accepted: 08/02/2024] [Indexed: 08/13/2024] Open
Abstract
Gastric polyps (GPs) are increasingly common. On upper endoscopy, they should be examined with white light and occasionally chromoendoscopy, and their morphology classified according to the Paris classification. Most GPs have a typical endoscopic appearance and can be associated with diseases like Helicobacter pylori infection. Histological examination is necessary for an accurate diagnosis. While most polyps are non-neoplastic and do not require treatment, some carry a risk of malignancy or are already malignant. Therefore, understanding the diagnosis, classification, and management of GPs is crucial for patient prognostication. Our new classification categorizes GPs into "good", "bad", and "ugly" based on their likelihood of becoming malignant. We aim to provide descriptions of the endoscopic appearance, pathology, treatment, and follow-up for different GPs, as well as clinical management flowcharts.
Collapse
Affiliation(s)
- Deborah Costa
- Department of Digestive Endoscopy and Gastroenterology, AULSS2, Conegliano Hospital, Conegliano 31015, Italy
| | - Daryl Ramai
- Division of Gastroenterology, Hepatology, and Endoscopy, Brigham and Women's Hospital, Boston, MA 02115, United States
| | - Alberto Tringali
- Department of Digestive Endoscopy and Gastroenterology, AULSS2, Conegliano Hospital, Conegliano 31015, Italy
| |
Collapse
|
48
|
Ren Y, Fang G, Wang K, Yan B, Wang C. The diagnostic value of image-enhanced endoscopy system in sinonasal inverted papilloma. Eur Arch Otorhinolaryngol 2024; 281:4221-4230. [PMID: 38713292 DOI: 10.1007/s00405-024-08707-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Accepted: 04/24/2024] [Indexed: 05/08/2024]
Abstract
PURPOSE This study aimed to evaluate the diagnostic value of image-enhanced endoscopy (IEE) in detecting sinonasal inverted papilloma (SNIP). METHODS Overall, 86 patients with unilateral nasal papillary or lobulated neoplasms were included between July 2018 and June 2019. All patients underwent IEE examinations, and the diagnosis of all neoplasms was confirmed through postoperative pathology. Logistic regression analysis was conducted to screen for independent predictors of various types of vascular patterns of SNIP. Furthermore, a prognostic nomogram was constructed using the independent predictors screened by logistic regression analysis to evaluate its usefulness in distinguishing SNIP from nasal polyp (NP) and papillary mucosa folds (PMF). RESULTS In total, 86 consecutive cases were observed, including 37 with SNIP, 40 with NP, and 9 with PMF. Logistic regression analysis showed that spot, corkscrew, and multilayered vascular patterns were independent predictors of SNIP diagnosis. Furthermore, a nomogram comprising the three independent risk factors was constructed with scores of 5, 2, and 3. The area under the receiver operating characteristic curve for predicting SNIP was 0.954, 0.66, 0.71, and 0.76 for the nomogram model, spot vascular pattern, corkscrew vascular pattern, and multilayered vascular pattern, respectively. CONCLUSION The nomogram model based on spot, corkscrew, and multilayered vascular patterns in SNIP observed using IEE can be a useful diagnostic tool for predicting and distinguishing between NP and PMF.
Collapse
Affiliation(s)
- Yimin Ren
- Department of Otolaryngology, Head and Neck Surgery, Beijing TongRen Hospital, Capital Medical University, No. 1, Dongjiaominxiang, Dongcheng District, Beijing, 100730, China
- Beijing Laboratory of Allergic Diseases, Key Laboratory of Otolaryngology Head and Neck Surgery, Ministry of Education, Beijing Institute of Otolaryngology, Beijing Key Laboratory of Nasal Diseases, Capital Medical University, Beijing, 100005, China
- Research Unit of Diagnosis and Treatment of Chronic Nasal Diseases, Chinese Academy of Medical Sciences, Beijing, 100005, China
| | - Gaoli Fang
- Department of Otolaryngology Head and Neck Surgery, Beijing DiTan Hospital, Capital Medical University, Beijing, China
| | - Kuiji Wang
- Department of Otolaryngology, Head and Neck Surgery, Beijing TongRen Hospital, Capital Medical University, No. 1, Dongjiaominxiang, Dongcheng District, Beijing, 100730, China
- Research Unit of Diagnosis and Treatment of Chronic Nasal Diseases, Chinese Academy of Medical Sciences, Beijing, 100005, China
| | - Bing Yan
- Department of Otolaryngology, Head and Neck Surgery, Beijing TongRen Hospital, Capital Medical University, No. 1, Dongjiaominxiang, Dongcheng District, Beijing, 100730, China.
- Beijing Laboratory of Allergic Diseases, Key Laboratory of Otolaryngology Head and Neck Surgery, Ministry of Education, Beijing Institute of Otolaryngology, Beijing Key Laboratory of Nasal Diseases, Capital Medical University, Beijing, 100005, China.
- Research Unit of Diagnosis and Treatment of Chronic Nasal Diseases, Chinese Academy of Medical Sciences, Beijing, 100005, China.
| | - Chengshuo Wang
- Department of Otolaryngology, Head and Neck Surgery, Beijing TongRen Hospital, Capital Medical University, No. 1, Dongjiaominxiang, Dongcheng District, Beijing, 100730, China.
- Beijing Laboratory of Allergic Diseases, Key Laboratory of Otolaryngology Head and Neck Surgery, Ministry of Education, Beijing Institute of Otolaryngology, Beijing Key Laboratory of Nasal Diseases, Capital Medical University, Beijing, 100005, China.
- Research Unit of Diagnosis and Treatment of Chronic Nasal Diseases, Chinese Academy of Medical Sciences, Beijing, 100005, China.
| |
Collapse
|
49
|
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
|
50
|
Cui M, Wang X, Qiao H, Wu S, Shang B. ELANE is a promising prognostic biomarker that mediates pyroptosis in gastric cancer. Heliyon 2024; 10:e34360. [PMID: 39130462 PMCID: PMC11315173 DOI: 10.1016/j.heliyon.2024.e34360] [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: 04/28/2024] [Revised: 07/08/2024] [Accepted: 07/08/2024] [Indexed: 08/13/2024] Open
Abstract
Background Gastric cancer (GC) is a typical malignant tumor and the main cause of cancer-related deaths. Its pathogenesis involves multiple steps, including pyroptosis, although these steps are still uncertain. Pyroptosis, also known as gasdermin-mediated programmed necrosis, participates in various pathological processes in tumors, including GC. ELANE, which encodes neutrophil elastase, is closely associated with GC. Additionally, ELANE has been implicated in GC cell pyroptosis, but this has not been confirmed. Therefore, investigating the link between ELANE and pyroptosis in GC is warranted. This research uses bioinformatics and experiments to examine the relationship between ELANE, pyroptosis, and GC prognosis. Methods The GEO and TCGA databases, along with pyroptosis-related genes, were applied to identify pyroptosis-related differentially expressed genes (DEGs). ELANE was selected via primary screening. Using the median expression level of ELANE as the threshold, pyroptosis-related DEGs were divided into low- and high-ELANE groups. Based on the DEGs in these two groups, GO, KEGG and GSEA analyses were conducted to elucidate the mechanisms of ELANE in GC. Furthermore, we plotted ROC and Kaplan-Meier curves to analyze the clinical and pathological features of ELANE expression. The Nomograms tool was applied to calculate the predictive value of ELANE for the clinical outcomes of GC cases. Immunohistochemical analysis was performed to detect the level of ELANE in GC tissues and to validate whether ELANE was involved in pyroptosis in GC cells through cell experiments. Finally, the immune infiltration of ELANE was investigated, and interaction networks (proteins-ELANE, microRNA-ELANE, and small-molecule drug-ELANE) were constructed. Results We aimed to investigate the expression of the ELANE gene in GC and study the relationship among ELANE, pyroptosis, and the prognosis of patients with GC. Differential expression analysis of gene-expression datasets from TCGA-STAD and GSE49051 revealed that the expression of the ELANE gene was significantly up-regulated in GC. Using STRING network analysis, we identified multiple proteins involved in the occurrence and development of GC, including interactions between ELANE and GSDMC, a member of the gasdermin protein family. Survival analysis showed that ELANE expression levels significantly affected overall survival (OS), disease-free survival (DFS), and progression-free survival (PFS) in patients with GC. Additionally, ROC analysis demonstrated that ELANE was effective in distinguishing GC patients from normal controls (AUC = 0.812). Immunohistochemical analysis showed that ELANE was highly expressed in gastric cancer tissues and was closely related to age, tumor grade, and stage. The cell experiments further confirmed that the high expression of ELANE in gastric cancer cells was associated with pyroptosis. Comprehensive analysis indicated that ELANE could be used as a potential prognostic marker for GC and plays an important role in pyroptosis. Conclusion High ELANE expression is related to poor survival and prognosis of patients with GC. It participates in pyroptosis and immune infiltration in GC. Therefore, ELANE is a promising prognostic biomarker for pyroptosis in GC.
Collapse
Affiliation(s)
- Ming Cui
- The Second Hospital of Dalian Medical University, Dalian, Liaoning province, China
| | - Xiaowu Wang
- The Third Affiliated Hospital of Wenzhou Medical University, Ruian, Zhejiang Province, China
| | - Haiyan Qiao
- Laboratory Animal Center, Dalian Medical University, Dalian, Liaoning Province, China
| | - Shixi Wu
- The Second Hospital of Dalian Medical University, Dalian, Liaoning province, China
| | - Bingbing Shang
- The Second Hospital of Dalian Medical University, Dalian, Liaoning province, China
| |
Collapse
|