1
|
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
|
2
|
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
|
3
|
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
|
4
|
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
|
5
|
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
|
6
|
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
|
7
|
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
|
8
|
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
|
9
|
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
|
10
|
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
|
11
|
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
|
12
|
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
|
13
|
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 DOI: 10.1016/j.cgh.2024.09.003] [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: 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
|
14
|
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
|
15
|
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
|
16
|
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
|
17
|
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
|
18
|
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
|
19
|
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
|
20
|
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 2024:10.1007/s11427-023-2558-x. [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] [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
|
21
|
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
|
22
|
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
|
23
|
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
|
24
|
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
|
25
|
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
|
26
|
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
|
27
|
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: 0] [Impact Index Per Article: 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
|
28
|
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
|
29
|
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
|
30
|
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
|
31
|
Yoon JY, Katcher E, Cohen E, Ward SC, Rouphael C, Itzkowitz SH, Wang CP, Kim MK, Shah SC. Endoscopic Surveillance of Gastric Intestinal Metaplasia: A Retrospective Cohort Study. J Clin Gastroenterol 2024:00004836-990000000-00327. [PMID: 39042489 DOI: 10.1097/mcg.0000000000002039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Accepted: 05/28/2024] [Indexed: 07/25/2024]
Abstract
BACKGROUND Gastric intestinal metaplasia (GIM) is a precancerous condition. Limited data exist on real-world clinical practice relative to guidelines. AIM The aim of this study was to evaluate adherence to GIM risk stratification and identify factors associated with follow-up endoscopy. MATERIALS AND METHODS We conducted manual chart review of patients with histologically confirmed GIM at an urban, tertiary medical center were identified retrospectively and details of their demographics, Helicobacter pylori, biopsy protocol, endoscopic/histologic findings, and postendoscopy follow-up were recorded. Multivariable logistic regression was used to identify factors independently associated with follow-up endoscopy. RESULTS Among 253 patients, 59% were female, 37% non-Hispanic White (NHW), 26% Hispanic, 16% non-Hispanic Black (NHB). The median age at index endoscopy was 63.4 years (IQR: 55.9 to 70.0), with median follow-up of 65.1 months (IQR: 44.0 to 72.3). H. pylori was detected in 21.6% patients at index EGD. GIM extent and subtype data were frequently missing (22.9% and 32.8%, respectively). Based on available data, 26% had corpus-extended GIM and 28% had incomplete/mixed-type GIM. Compared with NHW, Hispanic patients had higher odds of follow-up EGD (OR=2.48, 95% CI: 1.23-5.01), while NHB patients had 59% lower odds of follow-up EGD (OR=0.41, 95% CI: 0.18-0.96). Corpus-extended GIM versus limited GIM (OR=2.27, 95% CI: 1.13-4.59) was associated with follow-up EGD, but GIM subtype and family history of gastric cancer were not. CONCLUSIONS We observed suboptimal risk stratification among patients with GIM and notable race and ethnic disparities with respect to endoscopic surveillance. Targeted interventions are needed to improve practice patterns and mitigate observed disparities.
Collapse
Affiliation(s)
| | - Erik Katcher
- Sackler School of Medicine at Tel Aviv University, Tel Aviv, Israel
| | | | - Stephen C Ward
- Pathology, Molecular, and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Carol Rouphael
- Department of Gastroenterology, Hepatology, and Nutrition, Cleveland Clinic Foundation, Cleveland, OH
| | | | | | - Michelle K Kim
- Department of Gastroenterology, Hepatology, and Nutrition, Cleveland Clinic Foundation, Cleveland, OH
| | - Shailja C Shah
- Division of Gastroenterology, University of California San Diego
- Gastroenterology Section, Veterans Affairs San Diego Healthcare System, San Diego, CA
| |
Collapse
|
32
|
Almadi MA, Lu Y, Alali AA, Barkun AN. Peptic ulcer disease. Lancet 2024; 404:68-81. [PMID: 38885678 DOI: 10.1016/s0140-6736(24)00155-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 09/22/2023] [Accepted: 01/24/2024] [Indexed: 06/20/2024]
Abstract
Annual prevalence estimates of peptic ulcer disease range between 0·12% and 1·5%. Peptic ulcer disease is usually attributable to Helicobacter pylori infection, intake of some medications (such as aspirin and non-steroidal anti-inflammatory medications), or being critically ill (stress-related), or it can be idiopathic. The clinical presentation is usually uncomplicated, with peptic ulcer disease management based on eradicating H pylori if present, the use of acid-suppressing medications-most often proton pump inhibitors (PPIs)-or addressing complications, such as with early endoscopy and high-dose PPIs for peptic ulcer bleeding. Special considerations apply to patients on antiplatelet and antithrombotic agents. H pylori treatment has evolved, with the choice of regimen dictated by local antibiotic resistance patterns. Indications for primary and secondary prophylaxis vary across societies; most suggest PPIs for patients at highest risk of developing a peptic ulcer, its complications, or its recurrence. Additional research areas include the use of potassium-competitive acid blockers and H pylori vaccination; the optimal approach for patients at risk of stress ulcer bleeding requires more robust determinations of optimal patient selection and treatment selection, if any. Appropriate continuation of PPI use outweighs most possible side-effects if given for approved indications, while de-prescribing should be trialled when a definitive indication is no longer present.
Collapse
Affiliation(s)
- Majid A Almadi
- Division of Gastroenterology, King Khalid University Hospital, King Saud University, Riyadh, Saudi Arabia; Division of Gastroenterology, The McGill University Health Center, Montréal General Hospital, McGill University, Montréal, QC, Canada
| | - Yidan Lu
- Division of Gastroenterology, The McGill University Health Center, Montréal General Hospital, McGill University, Montréal, QC, Canada
| | - Ali A Alali
- Department of Medicine, Faculty of Medicine, Kuwait University, Jabriyah, Kuwait
| | - Alan N Barkun
- Division of Gastroenterology, The McGill University Health Center, Montréal General Hospital, McGill University, Montréal, QC, Canada; Division of Clinical Epidemiology, The McGill University Health Center, Montréal General Hospital, McGill University, Montréal, QC, Canada.
| |
Collapse
|
33
|
Zeng X, Yang L, Dong Z, Gong D, Li Y, Deng Y, Du H, Li X, Xu Y, Luo C, Wang J, Tao X, Zhang C, Zhu Y, Jiang R, Yao L, Wu L, Jin P, Yu H. The effect of incorporating domain knowledge with deep learning in identifying benign and malignant gastric whitish lesions: A retrospective study. J Gastroenterol Hepatol 2024; 39:1343-1351. [PMID: 38414305 DOI: 10.1111/jgh.16525] [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: 11/28/2023] [Revised: 01/22/2024] [Accepted: 02/05/2024] [Indexed: 02/29/2024]
Abstract
BACKGROUND AND AIM Early whitish gastric neoplasms can be easily misdiagnosed; differential diagnosis of gastric whitish lesions remains a challenge. We aim to build a deep learning (DL) model to diagnose whitish gastric neoplasms and explore the effect of adding domain knowledge in model construction. METHODS We collected 4558 images from two institutions to train and test models. We first developed two sole DL models (1 and 2) using supervised and semi-supervised algorithms. Then we selected diagnosis-related features through literature research and developed feature-extraction models to determine features including boundary, surface, roundness, depression, and location. Then predictions of the five feature-extraction models and sole DL model were combined and inputted into seven machine-learning (ML) based fitting-diagnosis models. The optimal model was selected as ENDOANGEL-WD (whitish-diagnosis) and compared with endoscopists. RESULTS Sole DL 2 had higher sensitivity (83.12% vs 68.67%, Bonferroni adjusted P = 0.024) than sole DL 1. Adding domain knowledge, the decision tree performed best among the seven ML models, achieving higher specificity than DL 1 (84.38% vs 72.27%, Bonferroni adjusted P < 0.05) and higher accuracy than DL 2 (80.47%, Bonferroni adjusted P < 0.001) and was selected as ENDOANGEL-WD. ENDOANGEL-WD showed better accuracy compared with 10 endoscopists (75.70%, P < 0.001). CONCLUSIONS We developed a novel system ENDOANGEL-WD combining domain knowledge and traditional DL to detect gastric whitish neoplasms. Adding domain knowledge improved the performance of traditional DL, which provided a novel solution for establishing diagnostic models for other rare diseases potentially.
Collapse
Affiliation(s)
- Xiaoquan Zeng
- Renmin Hospital of Wuhan University, Wuhan, China
- Key Laboratory of Hubei Province for Digestive System Disease, Renmin Hospital of Wuhan University, Wuhan, China
- Hubei Provincial Clinical Research Center for Digestive Disease Minimally Invasive Incision, Renmin Hospital of Wuhan University, Wuhan, China
- Engineering Research Center for Artificial lntelligence Endoscopy Interventional Treatment of Hubei Province, Wuhan, China
| | - Lang Yang
- Department of Gastroenterology, The Seventh Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Zehua Dong
- Renmin Hospital of Wuhan University, Wuhan, China
- Key Laboratory of Hubei Province for Digestive System Disease, Renmin Hospital of Wuhan University, Wuhan, China
- Hubei Provincial Clinical Research Center for Digestive Disease Minimally Invasive Incision, Renmin Hospital of Wuhan University, Wuhan, China
- Engineering Research Center for Artificial lntelligence Endoscopy Interventional Treatment of Hubei Province, Wuhan, China
| | - Dexin Gong
- Renmin Hospital of Wuhan University, Wuhan, China
- Key Laboratory of Hubei Province for Digestive System Disease, Renmin Hospital of Wuhan University, Wuhan, China
- Hubei Provincial Clinical Research Center for Digestive Disease Minimally Invasive Incision, Renmin Hospital of Wuhan University, Wuhan, China
- Engineering Research Center for Artificial lntelligence Endoscopy Interventional Treatment of Hubei Province, Wuhan, China
| | - Yanxia Li
- Renmin Hospital of Wuhan University, Wuhan, China
- Key Laboratory of Hubei Province for Digestive System Disease, Renmin Hospital of Wuhan University, Wuhan, China
- Hubei Provincial Clinical Research Center for Digestive Disease Minimally Invasive Incision, Renmin Hospital of Wuhan University, Wuhan, China
- Engineering Research Center for Artificial lntelligence Endoscopy Interventional Treatment of Hubei Province, Wuhan, China
| | - Yunchao Deng
- Renmin Hospital of Wuhan University, Wuhan, China
- Key Laboratory of Hubei Province for Digestive System Disease, Renmin Hospital of Wuhan University, Wuhan, China
- Hubei Provincial Clinical Research Center for Digestive Disease Minimally Invasive Incision, Renmin Hospital of Wuhan University, Wuhan, China
- Engineering Research Center for Artificial lntelligence Endoscopy Interventional Treatment of Hubei Province, Wuhan, China
| | - Hongliu Du
- Renmin Hospital of Wuhan University, Wuhan, China
- Key Laboratory of Hubei Province for Digestive System Disease, Renmin Hospital of Wuhan University, Wuhan, China
- Hubei Provincial Clinical Research Center for Digestive Disease Minimally Invasive Incision, Renmin Hospital of Wuhan University, Wuhan, China
- Engineering Research Center for Artificial lntelligence Endoscopy Interventional Treatment of Hubei Province, Wuhan, China
| | - Xun Li
- Renmin Hospital of Wuhan University, Wuhan, China
- Key Laboratory of Hubei Province for Digestive System Disease, Renmin Hospital of Wuhan University, Wuhan, China
- Hubei Provincial Clinical Research Center for Digestive Disease Minimally Invasive Incision, Renmin Hospital of Wuhan University, Wuhan, China
- Engineering Research Center for Artificial lntelligence Endoscopy Interventional Treatment of Hubei Province, Wuhan, China
| | - Youming Xu
- Renmin Hospital of Wuhan University, Wuhan, China
- Key Laboratory of Hubei Province for Digestive System Disease, Renmin Hospital of Wuhan University, Wuhan, China
- Hubei Provincial Clinical Research Center for Digestive Disease Minimally Invasive Incision, Renmin Hospital of Wuhan University, Wuhan, China
- Engineering Research Center for Artificial lntelligence Endoscopy Interventional Treatment of Hubei Province, Wuhan, China
| | - Chaijie Luo
- Renmin Hospital of Wuhan University, Wuhan, China
- Key Laboratory of Hubei Province for Digestive System Disease, Renmin Hospital of Wuhan University, Wuhan, China
- Hubei Provincial Clinical Research Center for Digestive Disease Minimally Invasive Incision, Renmin Hospital of Wuhan University, Wuhan, China
- Engineering Research Center for Artificial lntelligence Endoscopy Interventional Treatment of Hubei Province, Wuhan, China
| | - Junxiao Wang
- Renmin Hospital of Wuhan University, Wuhan, China
- Key Laboratory of Hubei Province for Digestive System Disease, Renmin Hospital of Wuhan University, Wuhan, China
- Hubei Provincial Clinical Research Center for Digestive Disease Minimally Invasive Incision, Renmin Hospital of Wuhan University, Wuhan, China
- Engineering Research Center for Artificial lntelligence Endoscopy Interventional Treatment of Hubei Province, Wuhan, China
| | - Xiao Tao
- Renmin Hospital of Wuhan University, Wuhan, China
- Key Laboratory of Hubei Province for Digestive System Disease, Renmin Hospital of Wuhan University, Wuhan, China
- Hubei Provincial Clinical Research Center for Digestive Disease Minimally Invasive Incision, Renmin Hospital of Wuhan University, Wuhan, China
- Engineering Research Center for Artificial lntelligence Endoscopy Interventional Treatment of Hubei Province, Wuhan, China
| | - Chenxia Zhang
- Renmin Hospital of Wuhan University, Wuhan, China
- Key Laboratory of Hubei Province for Digestive System Disease, Renmin Hospital of Wuhan University, Wuhan, China
- Hubei Provincial Clinical Research Center for Digestive Disease Minimally Invasive Incision, Renmin Hospital of Wuhan University, Wuhan, China
- Engineering Research Center for Artificial lntelligence Endoscopy Interventional Treatment of Hubei Province, Wuhan, China
| | - Yijie Zhu
- Renmin Hospital of Wuhan University, Wuhan, China
- Key Laboratory of Hubei Province for Digestive System Disease, Renmin Hospital of Wuhan University, Wuhan, China
- Hubei Provincial Clinical Research Center for Digestive Disease Minimally Invasive Incision, Renmin Hospital of Wuhan University, Wuhan, China
- Engineering Research Center for Artificial lntelligence Endoscopy Interventional Treatment of Hubei Province, Wuhan, China
| | - Ruiqing Jiang
- Renmin Hospital of Wuhan University, Wuhan, China
- Key Laboratory of Hubei Province for Digestive System Disease, Renmin Hospital of Wuhan University, Wuhan, China
- Hubei Provincial Clinical Research Center for Digestive Disease Minimally Invasive Incision, Renmin Hospital of Wuhan University, Wuhan, China
- Engineering Research Center for Artificial lntelligence Endoscopy Interventional Treatment of Hubei Province, Wuhan, China
| | - Liwen Yao
- Renmin Hospital of Wuhan University, Wuhan, China
- Key Laboratory of Hubei Province for Digestive System Disease, Renmin Hospital of Wuhan University, Wuhan, China
- Hubei Provincial Clinical Research Center for Digestive Disease Minimally Invasive Incision, Renmin Hospital of Wuhan University, Wuhan, China
- Engineering Research Center for Artificial lntelligence Endoscopy Interventional Treatment of Hubei Province, Wuhan, China
| | - Lianlian Wu
- Renmin Hospital of Wuhan University, Wuhan, China
- Key Laboratory of Hubei Province for Digestive System Disease, Renmin Hospital of Wuhan University, Wuhan, China
- Hubei Provincial Clinical Research Center for Digestive Disease Minimally Invasive Incision, Renmin Hospital of Wuhan University, Wuhan, China
- Engineering Research Center for Artificial lntelligence Endoscopy Interventional Treatment of Hubei Province, Wuhan, China
| | - Peng Jin
- Department of Gastroenterology, The Seventh Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Honggang Yu
- Renmin Hospital of Wuhan University, Wuhan, China
- Key Laboratory of Hubei Province for Digestive System Disease, Renmin Hospital of Wuhan University, Wuhan, China
- Hubei Provincial Clinical Research Center for Digestive Disease Minimally Invasive Incision, Renmin Hospital of Wuhan University, Wuhan, China
- Engineering Research Center for Artificial lntelligence Endoscopy Interventional Treatment of Hubei Province, Wuhan, China
| |
Collapse
|
34
|
Kang S, Hu L, Zhou HM, Zhang L. Application of conscious sedation with afentanil combined with titrated infusion of remazolam in gastroscopy for obese patients. Asian J Surg 2024; 47:3102-3103. [PMID: 38431478 DOI: 10.1016/j.asjsur.2024.02.134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Accepted: 02/22/2024] [Indexed: 03/05/2024] Open
Affiliation(s)
- Shuai Kang
- Department of Anesthesiology, Second Affiliated Hospital of Jiaxing University, Jiaxing, 314000, Zhejiang Province, China.
| | - Li Hu
- Department of Anesthesiology, Second Affiliated Hospital of Jiaxing University, Jiaxing, 314000, Zhejiang Province, China.
| | - Hong-Mei Zhou
- Department of Anesthesiology, Second Affiliated Hospital of Jiaxing University, Jiaxing, 314000, Zhejiang Province, China.
| | - Liang Zhang
- Department of Anesthesiology, Second Affiliated Hospital of Jiaxing University, Jiaxing, 314000, Zhejiang Province, China.
| |
Collapse
|
35
|
Ksahiwagi K, Yoshida T, Fukuhara K, Bessho R, Ichikawa H, Inoue N, Takaishi H, Iwao Y, Kanai T. Optimal number of images and 2-year interval affect cancer detection in screening esophagogastroduodenoscopy: An observational study. Medicine (Baltimore) 2024; 103:e38774. [PMID: 38941380 PMCID: PMC11466123 DOI: 10.1097/md.0000000000038774] [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: 01/05/2024] [Accepted: 06/10/2024] [Indexed: 06/30/2024] Open
Abstract
We aimed to identify quality indicator for esophagogastroduodenoscopy for detecting upper gastrointestinal (UGI) cancer. Data from 43,526 consecutive health checkups from August 2012 to January 2022 were retrospectively collected. The study ultimately analyzed 42,387 examinations by 12 endoscopists who performed more than 1000 examinations, including all cancers detected. These endoscopists were classified either into fast/slow group based on their mean examination time for a normal finding of esophagogastroduodenoscopy during their first year of the examination, or small/large group based on number of endoscopic images, respectively. The association between UGI cancer detection rate and examination time or the number of images was analyzed, using 5 minutes or 50 images as cutoff values. The detection rate of overall (8 pharyngeal, 39 esophageal, 69 gastric) cancers in the fast, slow, small, and large groups were 0.17%, 0.32%, 0.21%, and 0.31%, respectively. On multivariable analysis, endoscopists in the fast group or the small group were less likely to detect overall UGI cancer (OR: 0.596, 95% CI: 0.373-0.952, P = .030; OR: 0.652, 95% CI: 0.434-0.979, P = .039). Additionally, repeated endoscopy within 2 years had a higher overall cancer detection rate, compared with repeated screening after 2 years. In a sub-analysis, a significant negative relationship was found between the detection rate of gastric cancer and the number of gastric images < 35 (OR: 0.305, 95% CI: 0.189-0.492, P = .000). There was also a negative correlation trend between the detection rate of pharyngeal and esophageal cancers and the number of esophageal images < 11 (OR: 0.395, 95% CI: 0.156-1.001, P = .050). The optimal number of images and screening 2-year interval are considered useful quality indicators for detecting UGI cancer. This study also suggests that a total of 50 images, or 35 images of the stomach are suitable for detecting UGI cancer, or gastric cancer, during screening endoscopy.
Collapse
Affiliation(s)
- Kazuhiro Ksahiwagi
- Center for Preventive Medicine, Keio University, Tokyo, Japan
- Hills Joint Research Laboratory for Future Preventive Medicine and Wellness, School of Medicine, Keio University, Tokyo, Japan
| | | | - Kayoko Fukuhara
- Center for Preventive Medicine, Keio University, Tokyo, Japan
| | - Rieko Bessho
- Center for Preventive Medicine, Keio University, Tokyo, Japan
| | | | - Nagamu Inoue
- Center for Preventive Medicine, Keio University, Tokyo, Japan
| | | | - Yasushi Iwao
- Center for Preventive Medicine, Keio University, Tokyo, Japan
| | - Takanori Kanai
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, School of Medicine, Keio University, Tokyo, Japan
| |
Collapse
|
36
|
Lewis D, Jimenez L, Mansour MH, Horton S, Wong WWL. A Systematic Review of Cost-Effectiveness Studies on Gastric Cancer Screening. Cancers (Basel) 2024; 16:2353. [PMID: 39001415 PMCID: PMC11240801 DOI: 10.3390/cancers16132353] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 06/14/2024] [Accepted: 06/21/2024] [Indexed: 07/16/2024] Open
Abstract
Gastric cancer (GC) poses notable economic and health burdens in settings where the incidence of disease is prevalent. Some countries have established early screening and treatment programs to address these challenges. The objectives of this systematic review were to summarize the cost-effectiveness of gastric cancer screening presented in the literature and to identify the critical factors that influence the cost-effectiveness of screening. This systematic review followed the Preferred Reporting Items for Systematic Reviews and Meta-analyses (PRISMA) guidelines. Economic evaluation studies of gastric cancer screening were reviewed from SCOPUS and PubMed. The Consolidated Health Economic Evaluation Reporting Standards 2022 (CHEERS 2022) was used to assess the quality of reporting presented in the selected articles. Only primary economic evaluation studies addressing the cost-effectiveness, cost-utility, and cost-benefit of gastric cancer screening were selected. Two reviewers scrutinized the selected articles (title, abstract, and full text) to determine suitability for the systematic review based on inclusion and exclusion criteria. Authors' consensus was relied on where disagreements arose. The main outcome measures of concern in the systematic review were cost, effectiveness (as measured by either quality-adjusted life years (QALY) or life-years saved (LYS)), and incremental cost-effectiveness ratio (ICER) of screening versus either no screening or an alternative screening method. Thirty-one studies were selected for the final review. These studies investigated the cost-effectiveness of GC screening based on either primary, secondary, or a combination of primary and secondary interventions. The main primary intervention was Helicobacter pylori (Hp) screening with eradication, while the main secondary intervention was endoscopic screening. Cost-effectiveness was evaluated against no screening or screening using an alternative method in both observational and model-based studies. Screening was mainly cost-effective in Asian countries or their diasporas where the prevalence of GC was high. GC screening was generally not cost-effective among Western countries. GC screening can be cost-effective, but cost-effectiveness is dependent on context-specific factors, including geographical location, the prevalence of GC in the local population, and the screening tool adopted. However, there is benefit in targeting high-risk population groups in Asian countries and their diaspora for GC screening.
Collapse
Affiliation(s)
- Diedron Lewis
- School of Pharmacy, University of Waterloo, Waterloo, ON N2G 1C5, Canada
| | - Laura Jimenez
- Department of Community Health and Epidemiology, Dalhousie University, Halifax, NS B3H 4R2, Canada
| | - Manel Haj Mansour
- Department of Haematology and Oncology, Aga Khan University Hospital, Nairobi P.O. Box 30270-00100, Kenya
| | - Susan Horton
- School of Public Health Sciences, University of Waterloo, Waterloo, ON N2L 3G5, Canada
| | - William W L Wong
- School of Pharmacy, University of Waterloo, Waterloo, ON N2G 1C5, Canada
| |
Collapse
|
37
|
Xie XZ, Zuo L, Huang W, Fan QM, Weng YY, Yao WD, Jiang JL, Jin JQ. FDX1 as a novel biomarker and treatment target for stomach adenocarcinoma. World J Gastrointest Surg 2024; 16:1803-1824. [PMID: 38983344 PMCID: PMC11230022 DOI: 10.4240/wjgs.v16.i6.1803] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 04/25/2024] [Accepted: 04/28/2024] [Indexed: 06/27/2024] Open
Abstract
BACKGROUND Stomach adenocarcinoma (STAD) is one of the main reasons for cancer-related deaths worldwide. This investigation aimed to define the connection between STAD and Cuproptosis-related genes (CRGs). Cuproptosis is a newly identified form of mitochondrial cell death triggered by copper. AIM To explore the identification of potential biomarkers for STAD disease based on cuproptosis. METHODS A predictive model using Gene Ontology (GO), Least Absolute Shrinkage and Selection Operator (LASSO), Kyoto Encyclopedia of Genes and Genomes (KEGG), Gene Set Variation Analysis (GSVA), and Gene Set Enrichment Analysis analyzed gene interconnections, focusing on 3 copper-related genes and their expression in The Cancer Genome Atlas-STAD. Networks for mRNA-miRNA and mRNA-transcription factor interactions were constructed. The prognostic significance of CRG scores was evaluated using time-receiver operating characteristic, Kaplan-Meier curves, and COX regression analysis. Validation was conducted with datasets GSE26942, GSE54129, and GSE66229. Expression of copper-related differentially expressed genes was also analyzed in various human tissues and gastric cancer subpopulations using the human protein atlas. RESULTS Three significant genes (FDX1, LIAS, MTF1) were identified and selected via LASSO analysis to predict and classify individuals with STAD into high and low CRG score subgroups. These genes were down-regulated in both risk categories. GO and KEGG analyses highlighted their involvement mainly in the electron transport chain. After validating their differential expression, FDX1 emerged as the most accurate diagnostic marker for gastric cancer. Additionally, the RCircos package localized FDX1 on chromosome 11. CONCLUSION Our study revealed that FDX1 could be a potential biomarker and treatment target for gastric malignancy, providing new ideas for further scientific research.
Collapse
Affiliation(s)
- Xian-Ze Xie
- Department of Pharmacy, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou 310018, Zhejiang Province, China
| | - Lei Zuo
- Anhui Province Huainan City Shou County Agricultural Machinery Affairs Management Center, Huainan 232200, Anhui Province, China
| | - Wei Huang
- Department of Pharmacy, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou 310018, Zhejiang Province, China
| | - Qiao-Mei Fan
- Department of Pharmacy, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou 310018, Zhejiang Province, China
| | - Ya-Yun Weng
- Department of Pharmacy, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou 310018, Zhejiang Province, China
| | - Wen-Dong Yao
- Department of Pharmacy, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou 310018, Zhejiang Province, China
| | - Jia-Li Jiang
- Department of Pharmacy, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou 310018, Zhejiang Province, China
| | - Jia-Qi Jin
- Department of Pharmacy, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou 310018, Zhejiang Province, China
| |
Collapse
|
38
|
Cioates Negut C, Ilie-Mihai RM, Stefan-van Staden RI. Determination of Matrix Metalloproteinase 2 in Biological Samples Using a 3D Stochastic Microsensor Based on Graphene Oxide/AuNanoparticles/(Z)-N-(pyridin-4-yl-methyl) Octadec-9-enamide. Int J Mol Sci 2024; 25:6720. [PMID: 38928425 PMCID: PMC11203526 DOI: 10.3390/ijms25126720] [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: 05/22/2024] [Revised: 06/11/2024] [Accepted: 06/17/2024] [Indexed: 06/28/2024] Open
Abstract
The levels of the MMPs in the biological samples of confirmed patients with gastric cancer are significantly elevated compared to those found in healthy people. Therefore, a novel 3D stochastic microsensor based on graphene oxide, modified with gold nanoparticles and (Z)-N-(pyridin-4-yl-methyl) octadec-9-enamide (namely N2-AuNP/GO), was designed for the determination of MMP-2 in biological samples, and validated for the screening tests of biological samples in order to be used for the early diagnosis of gastric cancer. The proposed sensor presents a low limit of quantification (1.00 × 10-22 g mL-1), high sensitivity (1.84 × 107 s-1 g-1 mL), and a wide working concentration range (1.00 × 10-22-1.00 × 10-7 g mL-1). Recovery values higher than 99.15% were recorded for the assay of MMP-2 in whole blood, gastric tissue tumors, saliva, and urine samples.
Collapse
Affiliation(s)
| | - Ruxandra-Maria Ilie-Mihai
- Laboratory of Electrochemistry and PATLAB, National Institute for Research and Development in Electrochemistry and Condensed Matter, 202 Splaiul Independentei Str., 060021 Bucharest, Romania;
| | - Raluca-Ioana Stefan-van Staden
- Laboratory of Electrochemistry and PATLAB, National Institute for Research and Development in Electrochemistry and Condensed Matter, 202 Splaiul Independentei Str., 060021 Bucharest, Romania;
| |
Collapse
|
39
|
Yu ZH, Zhang LM, Dai ZQ, Zhang MN, Zheng SM. Epidemiology and prognostic nomogram for locally advanced gastric signet ring cell carcinoma: A population-based study. World J Gastrointest Oncol 2024; 16:2610-2630. [PMID: 38994168 PMCID: PMC11236255 DOI: 10.4251/wjgo.v16.i6.2610] [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: 01/05/2024] [Revised: 04/06/2024] [Accepted: 04/12/2024] [Indexed: 06/14/2024] Open
Abstract
BACKGROUND Gastric signet ring cell carcinoma (GSRC) represents a specific subtype of gastric cancer renowned for its contentious epidemiological features, treatment principles, and prognostic factors. AIM To investigate the epidemiology of GSRC and establish an improved model for predicting the prognosis of patients with locally advanced GSRC (LAGSRC) after surgery. METHODS The annual rates of GSRC incidence and mortality, covering the years 1975 to 2019, were extracted from the Surveillance, Epidemiology, and End Results (SEER) database to explore the temporal trends in both disease incidence and mortality rates using Joinpoint software. The clinical data of 3793 postoperative LAGSRC patients were collected from the SEER database for the analysis of survival rates. The Cox regression model was used to explore the independent prognostic factors for overall survival (OS). The risk factors extracted were used to establish a prognostic nomogram. RESULTS The overall incidence of GSRC increased dramatically between 1975 and 1998, followed by a significant downward trend in incidence after 1998. In recent years, there has been a similarly optimistic trend in GSRC mortality rates. The trend in GSRC showed discrepancies based on age and sex. Receiver operating characteristic curves, calibration curves, and decision curve analysis for 1-year, 3-year, and 5-year OS demonstrated the high discriminative ability and clinical utility of this nomogram. The area under the curve indicated that the performance of the new model outperformed that of the pathological staging system. CONCLUSION The model we established can aid clinicians in the early prognostication of LAGSRC patients, resulting in improved clinical outcomes by modifying management strategies and patient health care.
Collapse
Affiliation(s)
- Ze-Hao Yu
- Health Science Center, The First Affiliated Hospital of Ningbo University, Ningbo 315000, Zhejiang Province, China
- Health Science Center, Ningbo University, Ningbo 315211, Zhejiang Province, China
| | - Lei-Ming Zhang
- Health Science Center, Ningbo University, Ningbo 315211, Zhejiang Province, China
| | - Zhi-Qi Dai
- Health Science Center, The First Affiliated Hospital of Ningbo University, Ningbo 315000, Zhejiang Province, China
- Health Science Center, Ningbo University, Ningbo 315211, Zhejiang Province, China
| | - Meng-Na Zhang
- Health Science Center, The First Affiliated Hospital of Ningbo University, Ningbo 315000, Zhejiang Province, China
- College of Medicine, The First Affiliated Hospital of Zhejiang University, Hangzhou 310058, Zhejiang Province, China
| | - Si-Ming Zheng
- Health Science Center, The First Affiliated Hospital of Ningbo University, Ningbo 315000, Zhejiang Province, China
| |
Collapse
|
40
|
Yu ZH, Zhang LM, Dai ZQ, Zhang MN, Zheng SM. Epidemiology and prognostic nomogram for locally advanced gastric signet ring cell carcinoma: A population-based study. World J Gastrointest Oncol 2024; 16:2598-2618. [DOI: 10.4251/wjgo.v16.i6.2598] [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: 01/05/2024] [Revised: 04/06/2024] [Accepted: 04/12/2024] [Indexed: 06/13/2024] Open
Abstract
BACKGROUND Gastric signet ring cell carcinoma (GSRC) represents a specific subtype of gastric cancer renowned for its contentious epidemiological features, treatment principles, and prognostic factors.
AIM To investigate the epidemiology of GSRC and establish an improved model for predicting the prognosis of patients with locally advanced GSRC (LAGSRC) after surgery.
METHODS The annual rates of GSRC incidence and mortality, covering the years 1975 to 2019, were extracted from the Surveillance, Epidemiology, and End Results (SEER) database to explore the temporal trends in both disease incidence and mortality rates using Joinpoint software. The clinical data of 3793 postoperative LAGSRC patients were collected from the SEER database for the analysis of survival rates. The Cox regression model was used to explore the independent prognostic factors for overall survival (OS). The risk factors extracted were used to establish a prognostic nomogram.
RESULTS The overall incidence of GSRC increased dramatically between 1975 and 1998, followed by a significant downward trend in incidence after 1998. In recent years, there has been a similarly optimistic trend in GSRC mortality rates. The trend in GSRC showed discrepancies based on age and sex. Receiver operating characteristic curves, calibration curves, and decision curve analysis for 1-year, 3-year, and 5-year OS demonstrated the high discriminative ability and clinical utility of this nomogram. The area under the curve indicated that the performance of the new model outperformed that of the pathological staging system.
CONCLUSION The model we established can aid clinicians in the early prognostication of LAGSRC patients, resulting in improved clinical outcomes by modifying management strategies and patient health care.
Collapse
Affiliation(s)
- Ze-Hao Yu
- Health Science Center, The First Affiliated Hospital of Ningbo University, Ningbo 315000, Zhejiang Province, China
- Health Science Center, Ningbo University, Ningbo 315211, Zhejiang Province, China
| | - Lei-Ming Zhang
- Health Science Center, Ningbo University, Ningbo 315211, Zhejiang Province, China
| | - Zhi-Qi Dai
- Health Science Center, The First Affiliated Hospital of Ningbo University, Ningbo 315000, Zhejiang Province, China
- Health Science Center, Ningbo University, Ningbo 315211, Zhejiang Province, China
| | - Meng-Na Zhang
- Health Science Center, The First Affiliated Hospital of Ningbo University, Ningbo 315000, Zhejiang Province, China
- College of Medicine, The First Affiliated Hospital of Zhejiang University, Hangzhou 310058, Zhejiang Province, China
| | - Si-Ming Zheng
- Health Science Center, The First Affiliated Hospital of Ningbo University, Ningbo 315000, Zhejiang Province, China
| |
Collapse
|
41
|
Hartley I, Connoley D, Sane N, Hirsch R, Abeywickrama D, Sim N, Ea V, Azzopardi R, Simpson I, Bell S, Hew S. Gastric intestinal metaplasia: Prevalence in a large Australian center and nationwide survey of endoscopic practice. JGH Open 2024; 8:e13115. [PMID: 38933895 PMCID: PMC11199814 DOI: 10.1002/jgh3.13115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 05/09/2024] [Accepted: 06/12/2024] [Indexed: 06/28/2024]
Abstract
Background and Aim Atrophic gastritis (AG) and gastric intestinal metaplasia (GIM) are early changes in the stepwise progression to gastric adenocarcinoma. There is heterogeneity in international guidelines regarding the endoscopic diagnosis and surveillance of AG and GIM. This study aims to determine the prevalence of GIM in an Australian center and assess the approach of Australian endoscopists for these two conditions. Methods We conducted a single-center retrospective study of adult patients between January 2015 and December 2020 diagnosed with GIM on gastric biopsy following upper gastric endoscopy. A web-based, 25-question, investigator-designed, multiple-choice survey was distributed among all registered endoscopists in Australia. Results The overall prevalence of GIM within a single Australian center was 11.7% over 5 years. Of the 1026 patients identified, only 58.7% underwent mapping biopsies using the modified Sydney protocol. Among the cohort, 1.6% had low-grade dysplasia, 0.9% had high-grade dysplasia, and 1.8% had malignancy on initial gastroscopy. Two hundred and sixty-seven (7.2%) endoscopists completed the survey, 44.2% indicated they would perform mapping for all patients, and 36% only for high-risk patients. Only 1.5% (n = 4) of respondents were able to correctly identify all six endoscopic photos of GIM/AG. Conclusion This study demonstrates that in a large tertiary center, GIM is a prevalent endoscopic finding, but the associated rates of dysplasia and cancer were low. Additionally, among a small proportion of surveyed Australian endoscopists, there is notable variability in the endoscopic approach for AG and GIM and significant knowledge gaps. More training is required to increase the recognition of GIM and compliance with histological mapping.
Collapse
Affiliation(s)
- Imogen Hartley
- Gastroenterology DepartmentMonash HealthMelbourneVictoriaAustralia
| | - Declan Connoley
- Gastroenterology DepartmentMonash HealthMelbourneVictoriaAustralia
| | - Nikhita Sane
- Gastroenterology DepartmentMonash HealthMelbourneVictoriaAustralia
| | - Ryan Hirsch
- Gastroenterology DepartmentMonash HealthMelbourneVictoriaAustralia
| | | | - Nicholle Sim
- Gastroenterology DepartmentMonash HealthMelbourneVictoriaAustralia
| | - Vinny Ea
- Gastroenterology DepartmentMonash HealthMelbourneVictoriaAustralia
| | - Robert Azzopardi
- Gastroenterology DepartmentMonash HealthMelbourneVictoriaAustralia
| | - Ian Simpson
- Gastroenterology DepartmentMonash HealthMelbourneVictoriaAustralia
| | - Sally Bell
- Gastroenterology DepartmentMonash HealthMelbourneVictoriaAustralia
| | - Simon Hew
- Gastroenterology DepartmentMonash HealthMelbourneVictoriaAustralia
| |
Collapse
|
42
|
Yoo JW, Laszkowska M, Mendelsohn RB. The Role of Screening and Early Detection in Upper Gastrointestinal Cancers. Hematol Oncol Clin North Am 2024; 38:693-710. [PMID: 38431494 DOI: 10.1016/j.hoc.2024.01.007] [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] [Indexed: 03/05/2024]
Abstract
Upper gastrointestinal cancers are among the leading causes of cancer deaths worldwide with exceptionally poor prognosis, which is largely attributable to frequently delayed diagnosis. Although effective screening is critical for early detection, the highly variable incidence of upper gastrointestinal cancers presents challenges, rendering universal screening programs suboptimal in most populations globally. Optimal strategies in regions of modest incidence, such as the United States, require a targeted approach, focused on high-risk individuals based on demographic, familial, and clinicopathologic risk factors. Assessment of underlying precancerous lesions has key implications for risk stratification and informing clinical decisions to improve patient outcomes.
Collapse
Affiliation(s)
- Jin Woo Yoo
- Gastroenterology, Hepatology and Nutrition Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA.
| | - Monika Laszkowska
- Gastroenterology, Hepatology and Nutrition Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA
| | - Robin B Mendelsohn
- Gastroenterology, Hepatology and Nutrition Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA
| |
Collapse
|
43
|
Lykkegaard J, Olsen JK, Wehberg S, Jarbøl DE. The durability of previous examinations for cancer: Danish nationwide cohort study. Scand J Prim Health Care 2024; 42:246-253. [PMID: 38251839 PMCID: PMC11003324 DOI: 10.1080/02813432.2024.2305942] [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: 08/11/2023] [Accepted: 01/10/2024] [Indexed: 01/23/2024] Open
Abstract
OBJECTIVE Patients previously examined for cancer with a negative result may present in general practice with ongoing or new symptoms or signs suggestive of cancer. This paper explores the potential existence of a relatively safe period for cancer occurrence after receiving negative examination results for specific types of cancer, including lung (CT thorax), upper gastrointestinal (gastroscopy), colorectal (colonoscopy), bladder (cystoscopy), and breast (clinical mammography). DESIGN Register-based time-to-event analyses. SETTING Denmark. SUBJECTS All 3.3 million citizens aged 30-85 years who on January first, 2017, had not previously been diagnosed with the specific type of cancer were categorized based on the time since their most recent examination. MAIN OUTCOME MEASURES Using 1-year follow-up, we calculated the age- and sex-adjusted hazard ratios of being diagnosed with the related cancer, with non-examined individuals as reference. Negative examination results were defined as the absence of a cancer diagnosis within 6 months following the examination. RESULTS Previous negative examination results were common, also among those diagnosed with cancer during follow-up. For 10 years after a negative colonoscopy the risk of diagnosing a colorectal cancer was nearly halved. However, already 1 year after a clinical mammography and 2 years after a CT thorax the risk of diagnosing the related cancers was significantly higher among those with a previous negative result compared to non-examined individuals. CONCLUSION This study did not identify a post-examination period in which the cancer risk, compared to non-examined individuals, was sufficiently low to confidently rule out any of the investigated cancers.
Collapse
Affiliation(s)
- Jesper Lykkegaard
- Research Unit for General Practice, Department of Public Health, University of Southern Denmark, Odense, Denmark
| | - Jonas Kanstrup Olsen
- Research Unit for General Practice, Department of Public Health, University of Southern Denmark, Odense, Denmark
| | - Sonja Wehberg
- Research Unit for General Practice, Department of Public Health, University of Southern Denmark, Odense, Denmark
| | - Dorte Ejg Jarbøl
- Research Unit for General Practice, Department of Public Health, University of Southern Denmark, Odense, Denmark
| |
Collapse
|
44
|
Gul MO, Oguz Aslayan S, Corbaci K, Selman A, Akcay EB, Unal Ozdemir Z, Ozdemir H, Akyuz C. Gastric Polyps Detected Incidentally during Gastroscopy and Follow-Up Results. J Clin Med 2024; 13:3117. [PMID: 38892828 PMCID: PMC11172658 DOI: 10.3390/jcm13113117] [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: 04/25/2024] [Revised: 05/20/2024] [Accepted: 05/23/2024] [Indexed: 06/21/2024] Open
Abstract
(1) Background: We aimed to identify the possible relationship between various diseases of the upper digestive system and colon polyps by analyzing patients with gastric polyps and evaluating the cancers and diseases accompanying the polyps. (2) Methods: Each patient's age; gender; polyp type and size; presence of Helicobacter pylori (H. pylori), atrophic gastritis, and intestinal metaplasia; status of whether cancer developed during follow-up; status of whether a colonoscopy was performed or not; and colon pathologies detected during colonoscopy were analyzed retrospectively using hospital records. (3) Results: Between the study dates, 19,214 esophagogastroduodenoscopies were performed in the endoscopy unit of our hospital. Gastric polyps were detected in 178 (0.9%) patients. No significant relationship was found between the gastric polyp size and the occurrence of gastric cancer or gastrointestinal system malignancy (p > 0.05). A colonoscopy was performed in 86 of the 178 patients who underwent gastroscopy. The frequency of polyp detection during colonoscopy was statistically significantly higher in patients with gastric polyps than in patients without gastric polyps (p < 0.001). (4) Conclusions: New prospective studies are needed regarding the relationship between gastric polyps and gastrointestinal system diseases. Going forward, a colonoscopy will be required in gastric polyp patients, especially with FGP.
Collapse
Affiliation(s)
- Mehmet Onur Gul
- Surgical Oncology Clinic, Malatya Training and Research Hospital, Malatya 44330, Turkey
| | - Selda Oguz Aslayan
- General Surgery Department, Üsküdar State Hospital, Istanbul 34662, Turkey;
| | - Kadir Corbaci
- General Surgery Department, Osmaneli Mustafa Selahattin Çetintaş State Hospital, Bilecik 11500, Turkey;
| | - Aytac Selman
- General Surgery Department, Haydarpasa Numune Training and Research Hospital, Istanbul 34668, Turkey; (A.S.); (E.B.A.); (Z.U.O.); (H.O.); (C.A.)
| | - Emre Berat Akcay
- General Surgery Department, Haydarpasa Numune Training and Research Hospital, Istanbul 34668, Turkey; (A.S.); (E.B.A.); (Z.U.O.); (H.O.); (C.A.)
| | - Zehra Unal Ozdemir
- General Surgery Department, Haydarpasa Numune Training and Research Hospital, Istanbul 34668, Turkey; (A.S.); (E.B.A.); (Z.U.O.); (H.O.); (C.A.)
| | - Hakan Ozdemir
- General Surgery Department, Haydarpasa Numune Training and Research Hospital, Istanbul 34668, Turkey; (A.S.); (E.B.A.); (Z.U.O.); (H.O.); (C.A.)
| | - Cebrail Akyuz
- General Surgery Department, Haydarpasa Numune Training and Research Hospital, Istanbul 34668, Turkey; (A.S.); (E.B.A.); (Z.U.O.); (H.O.); (C.A.)
| |
Collapse
|
45
|
Horii T, Ikehara H, Arata S, Domen T, Kusano C. Efficacy of Cricoid Pressure Application during Esophagogastroduodenoscopy in Patients with Poor Gastric Wall Extension. Digestion 2024; 105:291-298. [PMID: 38744247 DOI: 10.1159/000539318] [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: 03/08/2024] [Accepted: 05/07/2024] [Indexed: 05/16/2024]
Abstract
INTRODUCTION Esophagogastroduodenoscopy (EGD) requires adequate air infusion. However, cases of poor gastrointestinal wall extension due to frequent eructation have been reported. Sufficient gastrointestinal wall extension can be achieved by applying cricoid pressure during EGD. Herein, we evaluated the frequency of cases with poor gastrointestinal wall extension and the efficacy and safety of applying cricoid pressure during EGD. METHODS This interventional study included patients who underwent EGD between January 2020 and December 2020 at the JA Akita Koseiren Yuri Kumiai General Hospital. Cases wherein folds of the greater curvature of the upper gastric body were not sufficiently extended during EGD were considered to have poor gastrointestinal wall extension. In such cases, air infusion was performed while applying cricoid pressure. This procedure was considered effective when gastric wall extension was achieved. RESULTS A total of 2,000 patients were enrolled and underwent upper gastrointestinal endoscopy; however, five were excluded because of upper gastrointestinal tract stenosis. Observation of gastric wall extension of the greater curvature in the upper gastric body with normal air insufflation was difficult in 113 (5.7%) cases. Applying cricoid pressure was effective in 93 (82.3%) patients with poor gastric wall extension. Sufficient gastric wall extension was achieved within an average of 12.8 s in cases where cricoid pressure application was effective. No adverse events were associated with cricoid pressure application. CONCLUSIONS Cricoid pressure application for patients with poor gastric wall extension during EGD is useful for ensuring a sufficient field of view during observation of the gastric body.
Collapse
Affiliation(s)
- Toshiki Horii
- Department of Gastroenterology, Kitasato University of Medicine, Sagamihara, Japan,
- Department of Gastroenterology, Yuri Kumiai General Hospital, Akita, Japan,
| | - Hisatomo Ikehara
- Department of Gastroenterology, Kitasato University of Medicine, Sagamihara, Japan
| | - Suguru Arata
- Department of Gastroenterology, Yuri Kumiai General Hospital, Akita, Japan
| | - Takahiro Domen
- Department of Gastroenterology, Yuri Kumiai General Hospital, Akita, Japan
| | - Chika Kusano
- Department of Gastroenterology, Kitasato University of Medicine, Sagamihara, Japan
| |
Collapse
|
46
|
Che B, Yuan S, Zhang H, Zhai J, Zhang Y, Wu C, Tang K. Causal inference between pernicious anemia and cancers: a bidirectional two-sample mendelian randomization analysis. BMC Cancer 2024; 24:586. [PMID: 38741062 DOI: 10.1186/s12885-024-12354-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: 12/20/2023] [Accepted: 05/07/2024] [Indexed: 05/16/2024] Open
Abstract
BACKGROUND Observational study investigated the association between pernicious anemia (PA) and cancers. However, with the exception of gastric cancer, the results are mostly contradictory. The purpose of this study was to investigate the potential causal relationship between PA and cancers through bidirectional two-sample Mendelian randomized (MR) analysis. METHODS The European sample FinnGen project provided the genetic summary data for PA and 20 site-specific cancers. This bidirectional two-sample MR design mainly used the inverse variance weighting (IVW) method to evaluate the causal relationship between PA and cancer risk. Benjamini-Hochberg correction was performed to reduce the bias caused by multiple tests. RESULTS Our study shows that there was a causal relationship between PA and gastric cancer, prostate cancer, testicular cancer and malignant melanoma of skin, and there was a reverse causal relationship between prostate cancer or gastric cancer and PA (P < 0.05). After Benjamini-Hochberg correction test, there was still a causal correlation between PA and gastric or prostate cancer (P' < 0.05), while there was only an implied causal association between PA and testicular cancer and malignant melanoma of skin (P'> 0.05). There was still a reverse causal relationship between gastric cancer and PA (P'< 0.05), while prostate cancer shows an implied reverse causal relationship(P'> 0.05). In addition, MR-Egger and MR-PRESSO tests showed no significant horizontal pleiotropy. CONCLUSIONS PA may be genetically associated with testicular cancer, prostate cancer, gastric cancer, and malignant melanoma of skin.
Collapse
Affiliation(s)
- Bangwei Che
- Department of Urology & Andrology, The First Affiliated of Guizhou University of Traditional Chinese Medicine, Guiyang, Guiyang, 550001, China
| | - Shenglan Yuan
- The First Clinical College, Guizhou University of Traditional Chinese Medicine, Guiyang, 550001, China
| | - Hongyan Zhang
- Physical examination center, The First Affiliated of Guizhou University of Traditional Chinese Medicine, Guiyang, 550001, China
| | - Jiancheng Zhai
- Department of Urology & Andrology, The First Affiliated of Guizhou University of Traditional Chinese Medicine, Guiyang, Guiyang, 550001, China
| | - Yang Zhang
- The First Clinical College, Guizhou University of Traditional Chinese Medicine, Guiyang, 550001, China
| | - Chuanchuan Wu
- The First Clinical College, Guizhou University of Traditional Chinese Medicine, Guiyang, 550001, China
| | - Kaifa Tang
- Department of Urology & Andrology, The First Affiliated of Guizhou University of Traditional Chinese Medicine, Guiyang, Guiyang, 550001, China.
| |
Collapse
|
47
|
Wang S, Zhang W, Wu X, Zhu Z, Chen Y, Liu W, Xu J, Chen L, Zhuang C. Comprehensive analysis of T-cell regulatory factors and tumor immune microenvironment in stomach adenocarcinoma. BMC Cancer 2024; 24:570. [PMID: 38714987 PMCID: PMC11077837 DOI: 10.1186/s12885-024-12302-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Accepted: 04/22/2024] [Indexed: 05/12/2024] Open
Abstract
BACKGROUND Gastric cancer (GC) is one of the most prevalent malignant tumors worldwide and is associated with high morbidity and mortality rates. However, the specific biomarkers used to predict the postoperative prognosis of patients with gastric cancer remain unknown. Recent research has shown that the tumor microenvironment (TME) has an increasingly positive effect on anti-tumor activity. This study aims to build signatures to study the effect of certain genes on gastric cancer. METHODS Expression profiles of 37 T cell-related genes and their TME characteristics were comprehensively analyzed. A risk signature was constructed and validated based on the screened T cell-related genes, and the roles of hub genes in GC were experimentally validated. RESULTS A novel T cell-related gene signature was constructed based on CD5, ABCA8, SERPINE2, ESM1, SERPINA5, and NMU. The high-risk group indicated lower overall survival (OS), poorer immune efficacy, and higher drug resistance, with SERPINE2 promoting GC cell proliferation, according to experiments. SERPINE2 and CXCL12 were significantly correlated, indicating poor OS via the Youjiang cohort. CONCLUSIONS This study identified T cell-related genes in patients with stomach adenocarcinoma (STAD) for prognosis estimation and proposed potential immunotherapeutic targets for STAD.
Collapse
Affiliation(s)
- Shuchang Wang
- Department of Gastrointestinal Surgery, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| | - Weifeng Zhang
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| | - Xinrui Wu
- Department of Urology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| | - Zhu Zhu
- Department of Clinical Medicine, Medical School of Nantong University, Nantong, China
| | - Yuanbiao Chen
- Department of Neurosurgery, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, China
| | - Wangrui Liu
- Department of Interventional Oncology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| | - Junnfei Xu
- Department of General Surgery, Affiliated Hospital of Nantong University, Nantong, 226001, Jiangsu, China.
| | - Li Chen
- Department of Urology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China.
- Department of Nursing, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China.
| | - Chun Zhuang
- Department of Gastrointestinal Surgery, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China.
| |
Collapse
|
48
|
Safavian N, Toh SKC, Pani M, Lee R. Enhancing endoscopic measurement: validating a quantitative method for polyp size and location estimation in upper gastrointestinal endoscopy. Surg Endosc 2024; 38:2505-2514. [PMID: 38467860 PMCID: PMC11078852 DOI: 10.1007/s00464-024-10758-2] [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: 11/26/2023] [Accepted: 02/16/2024] [Indexed: 03/13/2024]
Abstract
BACKGROUND Accurate measurement of polyps size is crucial in predicting malignancy, planning relevant intervention strategies and surveillance schedules. Endoscopists' visual estimations can lack precision. This study builds on our prior research, with the aim to evaluate a recently developed quantitative method to measure the polyp size and location accurately during a simulated endoscopy session. METHODS The quantitative method merges information about endoscopic positions obtained from an electromagnetic tracking sensor, with corresponding points on the images of the segmented polyp border. This yields real-scale 3D coordinates of the border of the polyp. By utilising the sensor, positions of any anatomical landmarks are attainable, enabling the estimation of a polyp's location relative to them. To verify the method's reliability and accuracy, simulated endoscopies were conducted in pig stomachs, where polyps were artificially created and assessed in a test-retest manner. The polyp measurements were subsequently compared against clipper measurements. RESULTS The average size of the fifteen polyps evaluated was approximately 12 ± 4.3 mm, ranging from 5 to 20 mm. The test-retest reliability, measured by the Intraclass Correlation Coefficient (ICC) for polyp size estimation, demonstrated an absolute agreement of 0.991 (95% CI 0.973-0.997, p < 0.05). Bland & Altman analysis revealed a mean estimation difference of - 0.17 mm (- 2.03%) for polyp size and, a mean difference of - 0.4 mm (- 0.21%) for polyp location. Both differences were statistically non-significant (p > 0.05). When comparing the proposed method with calliper measurements, the Bland & Altman plots showed 95% of size estimation differences between - 1.4 and 1.8 mm (- 13 to 17.4%) which was not significant (p > 0.05). CONCLUSIONS The proposed method of measurements of polyp size and location was found to be highly accurate, offering great potential for clinical implementation to improve polyp assessment. This level of performance represents a notable improvement over visual estimation technique used in clinical practice.
Collapse
Affiliation(s)
| | - Simon K C Toh
- Department of Upper GI Surgery, Queen Alexandra Hospital, Portsmouth Hospital University NHS Trust, Portsmouth, UK
| | - Martino Pani
- Faculty of Technology, University of Portsmouth, Portsmouth, UK
| | - Raymond Lee
- Faculty of Technology, University of Portsmouth, Portsmouth, UK.
| |
Collapse
|
49
|
Jalaeefar A, Mousavi SZ, Shirkhoda M, Mahmoodzadeh H, Mohammadzadeh N, Sharifi A. Is there any difference between Eastern and Western clinical practice guidelines in management of gastric cancer? Cancer Rep (Hoboken) 2024; 7:e2076. [PMID: 38711281 PMCID: PMC11074515 DOI: 10.1002/cnr2.2076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Revised: 03/10/2024] [Accepted: 04/08/2024] [Indexed: 05/08/2024] Open
Abstract
BACKGROUND AND RECENT FINDINGS Gastric cancer (GC) has been known as one of the most common causes of cancer mortality both in Western and Eastern countries. However, there might be differences between how it is managed in different countries. Thus, we aimed to investigate these differences. MATERIALS AND METHODS The most well-known clinical guidelines in field of GC management including Korean GC Association (KGCA), Japanese GC Association (JGCA), National Comprehensive Cancer Network (NCCN), European Society for Medical Oncology (ESMO), British Society of Gastroenterology (BSG), and National Institute for health and Care Excellence (NICE) have been reviewed. RESULTS The contents of these guidelines were categorized under eight headings including (1) genetic predisposition, (2) prevention, (3) management of gastric polyp, atrophy, dysplasia and metaplasia, (4) diagnosis, (5) pathology and molecular biology, (6) treatment, (7) supportive and palliative care, and (8) follow up. Difference in each section was discussed. CONCLUSION Considering KGCA and JGCA as Eastern and NCCN, ESMO, BSG, and NICE as Western guidelines, it is revealed that both sets of guidelines share common practices such as prioritizing comprehensive diagnostic evaluations, personalizing treatment plans, and palliative care. However, main differences can be seen in treatment regimens, the adoption of newer therapies like immunotherapy, and the utilization of emerging techniques such as HIPEC. These differences reflect the diverse clinical landscapes, research focuses, and healthcare systems within these regions.
Collapse
Affiliation(s)
- Amirmohsen Jalaeefar
- Department of Surgery, Subdivision of Surgical Oncology, Cancer InstituteTehran University of Medical SciencesTehranIran
| | - Seyedeh Zahra Mousavi
- Cancer Research Center of Cancer Institute, Tehran University of Medical SciencesTehranIran
| | - Mohammad Shirkhoda
- Department of Surgery, Subdivision of Surgical Oncology, Cancer InstituteTehran University of Medical SciencesTehranIran
| | | | - Narjes Mohammadzadeh
- Department of Surgery, Imam Khomeini Hospital ComplexTehran University of Medical SciencesTehranIran
| | - Amirsina Sharifi
- Sina Trauma and Surgery Research CenterTehran University of Medical SciencesTehranIran
| |
Collapse
|
50
|
Kayamba V, Munshi H, Hankolwe MN, Kaluba Kavimba C, Chongwe G, Knaze V, Park JY, Kelly P. Nationwide survey of Helicobacter pylori seropositivity and gastric atrophy in Zambia. Helicobacter 2024; 29:e13096. [PMID: 38790093 DOI: 10.1111/hel.13096] [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: 03/01/2024] [Revised: 05/09/2024] [Accepted: 05/10/2024] [Indexed: 05/26/2024]
Abstract
BACKGROUND Helicobacter pylori (H. pylori) is a common bacterial infection which predominately drives upper gastrointestinal pathology. We carried out a nationwide serological survey in response to the deficiency of robust African data on H. pylori prevalence, age of acquisition, socio-geographic determinants, and impact on gastric physiology. MATERIALS AND METHODS This was a cross-sectional study of archival plasma samples collected during the Zambia Population-based HIV impact Assessment (ZAMPHIA) 2016 survey. ZAMPHIA used a two-stage door-to-door stratified cluster sample approach to collect samples from adults and children from age 0 to 59 years (n = 24,266). We randomly retrieved one fifth of these samples from each of Zambia's 10 provinces and used ELISA to test for H. pylori IgG antibodies, pepsinogen 1 and 2 and gastrin-17. A pepsinogen 1:2 ratio of <3 was used to define gastric atrophy. RESULTS The analysis of 4050 plasma samples (30% <16 years, 53% females) revealed an overall H. pylori seroprevalence of 79%. By the age of 10 years, more than 75% of the children had H. pylori. Urban residence was associated with increased odds (OR 1.8, 95% CI 1.5-2.2, p < 0.001) and HIV infection was associated with reduced odds (OR 0.7, 95% CI 0.5-0.9, p = 0.02) of H. pylori seropositivity. Gastric atrophy was detected in 6% of H. pylori seropositive adults below 45 years of age and 9% in those between 45 and 59 years. CONCLUSIONS We have confirmed a high prevalence of H. pylori seropositivity in Zambia, predominantly in urban settings. The prevalence of gastric atrophy is broadly consistent with other populations around the globe, but our sample did not include adults over 60 years.
Collapse
Affiliation(s)
- Violet Kayamba
- Tropical Gastroenterology and Nutrition Group, Lusaka, Zambia
- University of Zambia School of Medicine, Lusaka, Zambia
| | - Husna Munshi
- Tropical Gastroenterology and Nutrition Group, Lusaka, Zambia
| | | | | | | | - Viktoria Knaze
- Early Detection, Prevention, and Infections Branch, International Agency for Research on Cancer (IARC/WHO), Lyon, France
| | - Jin Young Park
- Early Detection, Prevention, and Infections Branch, International Agency for Research on Cancer (IARC/WHO), Lyon, France
| | - Paul Kelly
- Tropical Gastroenterology and Nutrition Group, Lusaka, Zambia
- University of Zambia School of Medicine, Lusaka, Zambia
- Blizard Institute, Barts & The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| |
Collapse
|