1
|
Karra S, Sinduja R, Gurushankari B, Elamurugan TP, Mahalakshmy T, Kate V, Nanda N, Rajesh NG, Rajeswari M, Raj R, Shankar G. Development of Panel of Three-Dimensional Biomarkers to Identify Gastric Carcinoma and Precancerous Lesions of the Stomach - An Analytical Cross-Sectional Study. Indian J Clin Biochem 2024. [DOI: 10.1007/s12291-024-01257-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Accepted: 07/31/2024] [Indexed: 01/04/2025]
|
2
|
Guzmán J, Castillo D, González-Siccha AD, Bussalleu A, Trespalacios-Rangel AA, Lescano AG, Sauvain M. Helicobacter pylori cagA, vacA, iceA and babA Genotypes from Peruvian Patients with Gastric Intestinal Metaplasia. Cancers (Basel) 2024; 16:1476. [PMID: 38672558 PMCID: PMC11047899 DOI: 10.3390/cancers16081476] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2024] [Revised: 03/10/2024] [Accepted: 03/13/2024] [Indexed: 04/28/2024] Open
Abstract
We explored the clinical-stage association of gastric intestinal metaplasia (IM) compared to cases of chronic non-atrophic gastritis (CNAG) and its relationship with virulence genotypes of Helicobacter pylori (H. pylori) clinical isolates from patients with dyspepsia in Peru. This study was cross-sectional and included 158 H. pylori clinical isolates; each isolate corresponded to a different Peruvian patient, genotyped by polymerase chain reaction to detect cagA gene and EPIYA motifs, the vacA gene (alleles s1, s2, i1, i2, d1, d2, m1, m2 and subtypes s1a, s1b and s1c), the iceA gene (alleles 1 and 2), and the babA gene (allele 2). We observed that 38.6% presented with IM and that all clinical isolates were CagA positive. The EPIYA-ABC motif was predominant (68.4%), and we observed a high frequency for the vacA gene alleles s1 (94.9%), m1 (81.7%), i1 (63.9%), and d1 (70.9%). Strains with both iceA alleles were also detected (69.6%) and 52.2% were babA2 positive. In addition, it was observed that the cagA+/vacAs1m1 (PR: 2.42, 1.14 to 5.13, p < 0.05) and cagA+/vacAs1am1 (PR: 1.67, 1.13 to 2.45, p < 0.01) genotypes were associated with IM. Our findings revealed the cagA and vacA risk genotypes predominance, and we provided clinically relevant associations between Peruvian patients with H. pylori infection and IM clinical stage.
Collapse
Affiliation(s)
- Jesús Guzmán
- Laboratorio Centinela de Helicobacter pylori, Instituto de Medicina Tropical Alexander von Humboldt, Universidad Peruana Cayetano Heredia, Lima 15024, Peru; (D.C.); (A.B.); (M.S.)
- Facultad de Salud Pública y Administración, Universidad Peruana Cayetano Heredia, Lima 15102, Peru;
| | - Denis Castillo
- Laboratorio Centinela de Helicobacter pylori, Instituto de Medicina Tropical Alexander von Humboldt, Universidad Peruana Cayetano Heredia, Lima 15024, Peru; (D.C.); (A.B.); (M.S.)
| | - Anabel D. González-Siccha
- Departamento de Bioquímica, Facultad de Farmacia y Bioquímica, Universidad Nacional de Trujillo, Trujillo 13011, Peru;
| | - Alejandro Bussalleu
- Laboratorio Centinela de Helicobacter pylori, Instituto de Medicina Tropical Alexander von Humboldt, Universidad Peruana Cayetano Heredia, Lima 15024, Peru; (D.C.); (A.B.); (M.S.)
| | - Alba A. Trespalacios-Rangel
- Grupo de Investigación en Enfermedades Infecciosas, Departamento de Microbiología, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogotá 110231, Colombia;
| | - Andres G. Lescano
- Facultad de Salud Pública y Administración, Universidad Peruana Cayetano Heredia, Lima 15102, Peru;
| | - Michel Sauvain
- Laboratorio Centinela de Helicobacter pylori, Instituto de Medicina Tropical Alexander von Humboldt, Universidad Peruana Cayetano Heredia, Lima 15024, Peru; (D.C.); (A.B.); (M.S.)
- UMR 152 Pharmacochimie et Biologie pour le Développement (PHARMA-DEV), Institut de Recherche pour le Développement (IRD), Université de Toulouse, CEDEX 9, 31062 Toulouse, France
| |
Collapse
|
3
|
Zhan J, Zhou L, Zhang H, Zhou J, He Y, Hu T, Le Y, Lin Y, Wang J, Yu H, Liu Y, Xiang X. A comprehensive analysis of the expression, immune infiltration, prognosis and partial experimental validation of CHST family genes in gastric cancer. Transl Oncol 2024; 40:101843. [PMID: 38101175 PMCID: PMC10727950 DOI: 10.1016/j.tranon.2023.101843] [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: 09/12/2023] [Revised: 11/14/2023] [Accepted: 11/18/2023] [Indexed: 12/17/2023] Open
Abstract
Previous studies have demonstrated that carbohydrate sulfotransferase family proteins (CHSTs) play a crucial role in the extracellular matrix structural constituent and cancer progression, however, the effect of CHSTs on gastric cancer is still superficial. To investigate these, our study seeks to provide a comprehensive understanding of CHSTs' expression, immune infiltration, and prognostic implications in gastric cancer, utilizing data from the TCGA, GEO and GTEx databases. Furthermore, we conducted experimental validation to elucidate the role of CHST14 specifically in gastric cancer. Our findings suggest that most CHSTs were highly expressed in gastric cancer. Gene copy number variations further indicated prevalent CHSTs amplification in gastric cancer, pointing to its potential relevance in disease progression. Intriguingly, we noted strong positive correlations between most CHSTs and immune cell infiltration. Importantly, most members of CHSTs were related to OS and PFI with gastric cancer, with particular emphasis on CHST14 and CHST9. Multifactorial regression analysis indicates that CHST14 is an independent prognostic factor influencing the overall survival of gastric cancer patients. In further experimental validation, our results demonstrate elevated expression of CHST14 in gastric cancer, and knocking down CHST14 inhibits gastric cancer cell proliferation, invasion, migration and EMT. Additionally, CHST14 may exert its function through the regulation of the Wnt pathway. In summary, our study comprehensively analyzes the hitherto undescribed role of CHSTs in gastric cancer through the analysis of multi-omics data. Importantly, we identify CHST14 as a pivotal promoter in the malignant progression of gastric cancer, offering potential targets for gastric cancer therapy.
Collapse
Affiliation(s)
- Jinbo Zhan
- Department of Oncology, The First Affiliated Hospital of Nanchang University, 1519 Dongyue Avenue, Nanchang, Jiangxi, PR China; Department of Jiangxi Key Laboratory for Individualized Cancer Therapy, 17 Yongwai Street, Nanchang, Jiangxi 330006, PR China
| | - Ling Zhou
- Department of Oncology, The First Affiliated Hospital of Nanchang University, 1519 Dongyue Avenue, Nanchang, Jiangxi, PR China; Department of Jiangxi Key Laboratory for Individualized Cancer Therapy, 17 Yongwai Street, Nanchang, Jiangxi 330006, PR China
| | - Hongjiao Zhang
- Department of Oncology, The First Affiliated Hospital of Nanchang University, 1519 Dongyue Avenue, Nanchang, Jiangxi, PR China; Department of Jiangxi Key Laboratory for Individualized Cancer Therapy, 17 Yongwai Street, Nanchang, Jiangxi 330006, PR China
| | - Juanjuan Zhou
- Department of Oncology, The First Affiliated Hospital of Nanchang University, 1519 Dongyue Avenue, Nanchang, Jiangxi, PR China; Department of Jiangxi Key Laboratory for Individualized Cancer Therapy, 17 Yongwai Street, Nanchang, Jiangxi 330006, PR China
| | - Yan He
- Department of Oncology, The First Affiliated Hospital of Nanchang University, 1519 Dongyue Avenue, Nanchang, Jiangxi, PR China; Department of Jiangxi Key Laboratory for Individualized Cancer Therapy, 17 Yongwai Street, Nanchang, Jiangxi 330006, PR China
| | - Tingting Hu
- Department of Oncology, The First Affiliated Hospital of Nanchang University, 1519 Dongyue Avenue, Nanchang, Jiangxi, PR China; Department of Jiangxi Key Laboratory for Individualized Cancer Therapy, 17 Yongwai Street, Nanchang, Jiangxi 330006, PR China
| | - Yi Le
- Department of Oncology, The First Affiliated Hospital of Nanchang University, 1519 Dongyue Avenue, Nanchang, Jiangxi, PR China; Department of Jiangxi Key Laboratory for Individualized Cancer Therapy, 17 Yongwai Street, Nanchang, Jiangxi 330006, PR China
| | - Yun Lin
- Department of Oncology, The First Affiliated Hospital of Nanchang University, 1519 Dongyue Avenue, Nanchang, Jiangxi, PR China; Department of Jiangxi Key Laboratory for Individualized Cancer Therapy, 17 Yongwai Street, Nanchang, Jiangxi 330006, PR China
| | - Jingru Wang
- Department of Oncology, The First Affiliated Hospital of Nanchang University, 1519 Dongyue Avenue, Nanchang, Jiangxi, PR China; Department of Jiangxi Key Laboratory for Individualized Cancer Therapy, 17 Yongwai Street, Nanchang, Jiangxi 330006, PR China
| | - Haiming Yu
- Department of Oncology, The First Affiliated Hospital of Nanchang University, 1519 Dongyue Avenue, Nanchang, Jiangxi, PR China; Department of Jiangxi Key Laboratory for Individualized Cancer Therapy, 17 Yongwai Street, Nanchang, Jiangxi 330006, PR China
| | - Yawen Liu
- Department of Oncology, Jiangxi Maternal and Child Health Hospital, 318 Bayi Road, Nanchang 330006, PR China.
| | - Xiaojun Xiang
- Department of Oncology, The First Affiliated Hospital of Nanchang University, 1519 Dongyue Avenue, Nanchang, Jiangxi, PR China; Department of Jiangxi Key Laboratory for Individualized Cancer Therapy, 17 Yongwai Street, Nanchang, Jiangxi 330006, PR China.
| |
Collapse
|
4
|
Li Y, Jiang F, Wu CY, Leung WK. Prevalence and temporal trend of gastric preneoplastic lesions in Asia: A systematic review with meta-analysis. United European Gastroenterol J 2024; 12:139-151. [PMID: 38084663 PMCID: PMC10859711 DOI: 10.1002/ueg2.12507] [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: 05/25/2023] [Accepted: 10/20/2023] [Indexed: 02/13/2024] Open
Abstract
BACKGROUND Gastric cancer is the fifth most common cancer globally, with about 75% of cases occurring in Asia. While chronic atrophic gastritis (CAG) and intestinal metaplasia (IM) are well-recognized preneoplastic gastric lesions, we determined the prevalence and temporal trend of CAG and IM in Asia over the past 50 years. METHODS In this systematic review and meta-analysis, we searched PubMed, Embase, MEDLINE, Scopus, and Web of Science for studies reporting the prevalence of CAG and IM in Asia (according to the United Nations geoscheme) published between 1970 and 2022. Heterogeneity was assessed by the I2 index and Cochran Q test. We adopted the random effects model to estimate the pooled prevalence and 95% confidence interval (CI). The slope of prevalence was estimated as a function of time in simple linear regression and weighted meta-regression models to demonstrate the temporal trend. Studies that reported the odds ratio (OR) of Helicobacter pylori infection and CAG/IM were analyzed separately to compile a pooled OR with a 95% CI. This study was registered in INPLASY2022120028. RESULTS Of the 81 studies from 19 Asian countries identified, the pooled prevalence for CAG and IM in Asia was 26.1% (95%CI: 22.7-30.0) and 22.9% (95%CI: 19.7-26.6), respectively. Over the past 5 decades, there was a significant decline in the prevalence of IM (slope in adjusted meta-regression models: -0.79 [95%CI: -1.28 to -0.26], P = 0.003), but there was no significant change in the pooled prevalence of CAG. Within Asia, the prevalence varied significantly among different regions. Southern Asia reported the highest pooled prevalence of CAG (42.9%, 95%CI: 27.5%-67.1%), while Western Asia reported the lowest level (12.7%, 95%CI: 5.0%-32.3%). For IM, Eastern Asia reported the highest prevalence (27.1%, 95%CI: 21.1-34.9), with the lowest prevalence reported in Western Asia (3.1%; 95% CI 1.2%-8.0%). H. pylori infection was linked to CAG and IM with OR of 2.16 (95%CI: 2.09-2.22) and 1.64 (95%CI: 1.57-1.72), respectively. CONCLUSION This updated meta-analysis showed that up to 26% of study individuals in Asia harbored preneoplastic gastric lesions. There was a declining temporal trend in the prevalence of IM, but not for CAG, in Asia.
Collapse
Affiliation(s)
- Yunhao Li
- Department of MedicineSchool of Clinical MedicineLi Ka Shing Faculty of MedicineUniversity of Hong KongHong KongChina
| | - Fang Jiang
- Department of MedicineSchool of Clinical MedicineLi Ka Shing Faculty of MedicineUniversity of Hong KongHong KongChina
| | - Chun Ying Wu
- Institute of Biomedical InformaticsNational Yang Ming Chiao Tung UniversityTaipeiTaiwan
- Health Innovation CenterNational Yang Ming Chiao Tung UniversityTaipeiTaiwan
- Microbiota Research CenterNational Yang Ming Chiao Tung UniversityTaipeiTaiwan
- Division of Translational ResearchTaipei Veterans General HospitalTaipeiTaiwan
- Institute of Public HealthCollege of MedicineNational Yang Ming Chiao Tung UniversityTaipeiTaiwan
- Faculty of MedicineCollege of MedicineNational Yang Ming Chiao Tung UniversityTaipeiTaiwan
- Department of Public HealthChina Medical UniversityTaichungTaiwan
| | - Wai K. Leung
- Department of MedicineSchool of Clinical MedicineLi Ka Shing Faculty of MedicineUniversity of Hong KongHong KongChina
| |
Collapse
|
5
|
Kaibysheva V, Tykhonov S, Kashin S, Kuvaev R, Kraynova E, Baculina N, Fedorov E, Drapkina O. Algorithm of autoimmune gastritis diagnosis and treatment. RUSSIAN JOURNAL OF PREVENTIVE MEDICINE 2024; 27:101. [DOI: 10.17116/profmed202427091101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
6
|
Zhang L, Yao L, Lu Z, Yu H. Current status of quality control in screening esophagogastroduodenoscopy and the emerging role of artificial intelligence. Dig Endosc 2024; 36:5-15. [PMID: 37522555 DOI: 10.1111/den.14649] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Accepted: 07/27/2023] [Indexed: 08/01/2023]
Abstract
Esophagogastroduodenoscopy (EGD) screening is being implemented in countries with a high incidence of upper gastrointestinal (UGI) cancer. High-quality EGD screening ensures the yield of early diagnosis and prevents suffering from advanced UGI cancer and minimal operational-related discomfort. However, performance varied dramatically among endoscopists, and quality control for EGD screening remains suboptimal. Guidelines have recommended potential measures for endoscopy quality improvement and research has been conducted for evidence. Moreover, artificial intelligence offers a promising solution for computer-aided diagnosis and quality control during EGD examinations. In this review, we summarized the key points for quality assurance in EGD screening based on current guidelines and evidence. We also outline the latest evidence, limitations, and future prospects of the emerging role of artificial intelligence in EGD quality control, aiming to provide a foundation for improving the quality of EGD screening.
Collapse
Affiliation(s)
- Lihui Zhang
- Department of Gastroenterology, 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
| | - Liwen Yao
- Department of Gastroenterology, 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
| | - Zihua Lu
- Department of Gastroenterology, 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
| | - Honggang Yu
- Department of Gastroenterology, 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
| |
Collapse
|
7
|
Bogdanova I, Polaka I, Aleksandraviča I, Dzērve Z, Anarkulova L, Novika V, Tolmanis I, Leja M. Role of pre-existing incomplete intestinal metaplasia in gastric adenocarcinoma: A retrospective case series analysis. World J Clin Cases 2023; 11:2708-2715. [PMID: 37214563 PMCID: PMC10198109 DOI: 10.12998/wjcc.v11.i12.2708] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Revised: 01/19/2023] [Accepted: 03/29/2023] [Indexed: 04/25/2023] Open
Abstract
BACKGROUND Risk stratification for patients with gastric precancerous lesions for endoscopic surveillance remains controversial. AIM To analysis of patients having developed gastric adenocarcinoma during the period of follow-up. METHODS We conducted a retrospective study on patients having undergone upper endoscopy prior to the development of gastric adenocarcinoma. The presence and stage of precancerous lesions as well as subtype of intestinal metaplasia at the baseline endoscopy got evaluated. Literature mini-review was performed. RESULTS Out of 1681 subjects in the Biobank, gastric adenocarcinoma was detected in five cases in whom previous endoscopy data with biopsies either from the corpus or antral part were available. All of the patients had incomplete intestinal metaplasia during the baseline endoscopy; all three subjects in whom intestinal metaplasia subtyping was performed according to Filipe et al, had Type III intestinal metaplasia. Two of the five cases had low Operative Link on Gastritis Assessment (OLGA) and Operative Link on Gastritis Intestinal Metaplasia Assessment (OLGIM) stages (I-II) at the baseline. CONCLUSION The presence of incomplete intestinal metaplasia, in particular, that of Type III is a better predictor for gastric adenocarcinoma development than OLGA/OLGIM staging system. Subtyping of intestinal metaplasia have an important role in the risk stratification for surveillance decisions.
Collapse
Affiliation(s)
- Inga Bogdanova
- Department of Pathology, Academic Histology Laboratory, Riga LV1073, Latvia
- Institute of Clinical and Preventive Medicine, University of Latvia, Riga LV1079, Latvia
| | - Inese Polaka
- Institute of Clinical and Preventive Medicine, University of Latvia, Riga LV1079, Latvia
| | - Ilona Aleksandraviča
- Institute of Clinical and Preventive Medicine, University of Latvia, Riga LV1079, Latvia
- Department of Research, Riga East University Hospital, Riga LV1079, Latvia
| | - Zane Dzērve
- Institute of Clinical and Preventive Medicine, University of Latvia, Riga LV1079, Latvia
- Department of Endoscopy, Digestive Diseases Centre GASTRO, Riga LV1079, Latvia
| | - Linda Anarkulova
- Institute of Clinical and Preventive Medicine, University of Latvia, Riga LV1079, Latvia
| | - Vita Novika
- Department of Endoscopy, Digestive Diseases Centre GASTRO, Riga LV1079, Latvia
| | - Ivars Tolmanis
- Department of Endoscopy, Digestive Diseases Centre GASTRO, Riga LV1079, Latvia
| | - Marcis Leja
- Institute of Clinical and Preventive Medicine, University of Latvia, Riga LV1079, Latvia
- Department of Research, Riga East University Hospital, Riga LV1079, Latvia
- Department of Gastroenterology, Digestive Diseases Centre GASTRO, Riga LV1079, Latvia
| |
Collapse
|
8
|
Noh CK, Lee E, Park B, Lim SG, Shin SJ, Lee KM, Lee GH. Effect of Helicobacter pylori Eradication Treatment on Metachronous Gastric Neoplasm Prevention Following Endoscopic Submucosal Dissection for Gastric Adenoma. J Clin Med 2023; 12:1512. [PMID: 36836045 PMCID: PMC9962017 DOI: 10.3390/jcm12041512] [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: 01/07/2023] [Revised: 02/11/2023] [Accepted: 02/13/2023] [Indexed: 02/17/2023] Open
Abstract
The long-term effect of Helicobacter pylori eradication on metachronous gastric neoplasm prevention after endoscopic submucosal dissection (ESD) of gastric adenoma is unclear. This study included patients with confirmed H. pylori infection after ESD with curative resection for gastric adenoma. Patients were divided based on the success of H. pylori eradication treatment into two groups: eradication and non-eradication. Patients with any newly detected lesion within 1 year after ESD and recurrence at the ESD site were excluded from the analysis. Further, 1:1 propensity score matching was also performed to eliminate baseline differences between the two groups. H. pylori eradication treatment was administered to 673 patients after ESD (163 in the successful eradication group and 510 in the non-eradication group). During the median follow-up periods of 25 and 39 months in the eradication and non-eradication groups, metachronous gastric neoplasm was identified in 6 (3.7%) and 22 patients (4.3%), respectively. Adjusted Cox analysis revealed that H. pylori eradication was not associated with increased risk of metachronous gastric neoplasm after ESD. Kaplan-Meier analysis in the matched population yielded similar findings (p = 0.546). H. pylori eradication treatment was not associated with metachronous gastric neoplasm after ESD with curative resection for gastric adenoma.
Collapse
Affiliation(s)
- Choong-Kyun Noh
- Department of Gastroenterology, Ajou University School of Medicine, Suwon 16499, Republic of Korea
| | - Eunyoung Lee
- Department of Biomedical Informatics, Ajou University School of Medicine, Suwon 16499, Republic of Korea
- Office of Biostatistics, Ajou Research Institute for Innovative Medicine, Ajou University Medical Center, Suwon 16499, Republic of Korea
| | - Bumhee Park
- Department of Biomedical Informatics, Ajou University School of Medicine, Suwon 16499, Republic of Korea
- Office of Biostatistics, Ajou Research Institute for Innovative Medicine, Ajou University Medical Center, Suwon 16499, Republic of Korea
| | - Sun Gyo Lim
- Department of Gastroenterology, Ajou University School of Medicine, Suwon 16499, Republic of Korea
| | - Sung Jae Shin
- Department of Gastroenterology, Ajou University School of Medicine, Suwon 16499, Republic of Korea
| | - Kee Myung Lee
- Department of Gastroenterology, Ajou University School of Medicine, Suwon 16499, Republic of Korea
| | - Gil Ho Lee
- Department of Gastroenterology, Ajou University School of Medicine, Suwon 16499, Republic of Korea
| |
Collapse
|
9
|
Gastric bacteria as potential biomarkers for the diagnosis of atrophic gastritis. Mol Biol Rep 2023; 50:655-664. [PMID: 36371556 DOI: 10.1007/s11033-022-08001-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2022] [Accepted: 10/03/2022] [Indexed: 11/15/2022]
Abstract
BACKGROUND Identification of the risk factors for atrophic gastritis (AG) and prevention of further deterioration of the gastritis are effective approaches to reduce the incidence of gastric cancer. Previous studies found that dysbiosis has been implicated in a wide range of diseases, while the role of gastric bacteria as a biomarker for AG has not been explored. METHODS AND RESULTS Gastric juices from cases with non-atrophic gastritis (NAG) and AG were collected for investigation of bacterial composition and function. The β-diversity of microbiota exhibited a significant reduction in AG samples compared with that in NAG samples. Differential abundance analysis revealed that a total of 23 predicted species changed their distributions; meanwhile, all obligate anaerobic bacteria with a relatively high abundance lowered their contents in AG samples. Additionally, the correlation analysis indicated a clear shift in bacterial correlation pattern between the two groups. Functional interrogation of the gastric microbiota showed that bacterial metabolisms associated with enzyme families, digestive system, and endocrine system were downregulated in AG samples. The compositional dissection of "core microbiota" exhibited that oral pathogens, including Porphyromonas gingivalis, Campylobacter gracilis, and Granulicatella elegans, were magnified in AG samples, suggesting that oral diseases may be a trigger factor for early exacerbation of gastritis. Then, the differentially expressed bacteria were used as diagnostic biomarkers for the random forest classifier model for group prediction. CONCLUSIONS The results showed that bacterial biomarkers could distinguish AG patients from NAG cases with an accuracy of 90% at the genus level.
Collapse
|
10
|
Xu J, Chen S, Liang J, Hao T, Wang H, Liu G, Jin X, Li H, Zhang J, Zhang C, He Y. Schlafen family is a prognostic biomarker and corresponds with immune infiltration in gastric cancer. Front Immunol 2022; 13:922138. [PMID: 36090985 PMCID: PMC9452737 DOI: 10.3389/fimmu.2022.922138] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2022] [Accepted: 08/08/2022] [Indexed: 11/13/2022] Open
Abstract
The Schlafen (SLFN) gene family plays an important role in immune cell differentiation and immune regulation. Previous studies have found that the increased SLFN5 expression in patients with intestinal metaplasia correlates with gastric cancer (GC) progression. However, no investigation has been conducted on the SLFN family in GC. Therefore, we systematically explore the expression and prognostic value of SLFN family members in patients with GC, elucidating their possible biological function and its correlation with tumor immune cells infiltration. TCGA database results indicated that the SLFN5, SLFN11, SLFN12, SLFN12L, and SLFN13 expression was significantly higher in GC. The UALCAN and KM plotter databases indicated that enhanced the SLFN family expression was associated with lymph node metastasis, tumor stage, and tumor grade and predicted an adverse prognosis. cBioportal database revealed that the SLFN family had a high frequency of genetic alterations in GC (about 12%), including mutations and amplification. The GeneMANIA and STRING databases identified 20 interacting genes and 16 interacting proteins that act as potential targets of the SLFN family. SLFN5, SLFN11, SLFN12, SLFN12L, and SLFN14 may be implicated in the immunological response, according to Gene Set Enrichment Analysis (GSEA) and Kyoto Encyclopedia of Genes and Genomes (KEGG). Additionally, Timer and TISIDB databases indicate that SLFN5, SLFN11, SLFN12, SLFN12L, and SLFN14 are involved in the immune response. Furthermore, Timer, TCGA, and TISIDB databases suggested that the SLFN5, SLFN11, SLFN12, SLFN12L, and SLFN14 expression in GC is highly linked with immune cell infiltration levels, immune checkpoint, and the many immune cell marker sets expression. We isolated three samples of peripheral blood mononuclear cell (PBMC) and activated T cells; the results showed the expression of SLFN family members decreased significantly when T cell active. In conclusion, the SLFN family of proteins may act as a prognostic indicator of GC and is associated with immune cell infiltration and immune checkpoint expression in GC. Additionally, it may be involved in tumor immune evasion by regulating T cell activation.
Collapse
Affiliation(s)
- Jiannan Xu
- Digestive Diseases Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
- Department of Thoracic Surgery, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Songyao Chen
- Digestive Diseases Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Jianming Liang
- Digestive Diseases Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Tengfei Hao
- Digestive Diseases Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Huabin Wang
- Division of Hematology/Oncology, Department of Pediatrics, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Guangyao Liu
- Digestive Diseases Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Xinghan Jin
- Digestive Diseases Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Huan Li
- Digestive Diseases Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Junchang Zhang
- Digestive Diseases Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Changhua Zhang
- Digestive Diseases Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
- *Correspondence: Changhua Zhang, ; Yulong He,
| | - Yulong He
- Digestive Diseases Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
- Center of Gastrointestinal Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
- *Correspondence: Changhua Zhang, ; Yulong He,
| |
Collapse
|
11
|
Robles C, Rudzite D, Polaka I, Sjomina O, Tzivian L, Kikuste I, Tolmanis I, Vanags A, Isajevs S, Liepniece-Karele I, Razuka-Ebela D, Parshutin S, Murillo R, Herrero R, Young Park J, Leja M. Assessment of Serum Pepsinogens with and without Co-Testing with Gastrin-17 in Gastric Cancer Risk Assessment-Results from the GISTAR Pilot Study. Diagnostics (Basel) 2022; 12:1746. [PMID: 35885649 PMCID: PMC9325279 DOI: 10.3390/diagnostics12071746] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2022] [Revised: 07/06/2022] [Accepted: 07/13/2022] [Indexed: 12/03/2022] Open
Abstract
Introduction−−Serum pepsinogen tests for gastric cancer screening have been debated for decades. We assessed the performance of two pepsinogen assays with or without gastrin-17 for the detection of different precancerous lesions alone or as a composite endpoint in a Latvian cohort. Methods−−Within the intervention arm of the GISTAR population-based study, participants with abnormal pepsinogen values by ELISA or latex-agglutination tests, or abnormal gastrin-17 by ELISA and a subset of subjects with all normal biomarker values were referred for upper endoscopy with biopsies. Performance of biomarkers, corrected by verification bias, to detect five composite outcomes based on atrophy, intestinal metaplasia, dysplasia or cancer was explored. Results−−Data from 1045 subjects were analysed, of those 273 with normal biomarker results. Both pepsinogen assays showed high specificity (>93%) but poor sensitivity (range: 18.4−31.1%) that slightly improved when lesions were restricted to corpus location (40.5%) but decreased when dysplasia and prevalent cancer cases were included (23.8%). Adding gastrin-17 detection, sensitivity reached 33−45% while specificity decreased (range: 61.1−62%) and referral rate for upper endoscopy increased to 38.6%. Conclusions−−Low sensitivity of pepsinogen assays is a limiting factor for their use in population-based primary gastric cancer screening, however their high specificity could be useful for triage.
Collapse
Affiliation(s)
- Claudia Robles
- Early Detection, Prevention and Infections Branch, International Agency for Research on Cancer, 69372 Lyon, France; (R.M.); (R.H.); (J.Y.P.)
- Cancer Epidemiology Research Program, Catalan Institute of Oncology, IDIBELL, 08908 L’Hospitalet de Llobregat, Spain
| | - Dace Rudzite
- Institute of Clinical and Preventive Medicine, Faculty of Medicine, University of Latvia, 1007 Riga, Latvia; (D.R.); (I.P.); (O.S.); (L.T.); (I.K.); (S.I.); (I.L.-K.); (D.R.-E.); (S.P.); (M.L.)
- Department of Research, Riga East University Hospital, 1038 Riga, Latvia
| | - Inese Polaka
- Institute of Clinical and Preventive Medicine, Faculty of Medicine, University of Latvia, 1007 Riga, Latvia; (D.R.); (I.P.); (O.S.); (L.T.); (I.K.); (S.I.); (I.L.-K.); (D.R.-E.); (S.P.); (M.L.)
| | - Olga Sjomina
- Institute of Clinical and Preventive Medicine, Faculty of Medicine, University of Latvia, 1007 Riga, Latvia; (D.R.); (I.P.); (O.S.); (L.T.); (I.K.); (S.I.); (I.L.-K.); (D.R.-E.); (S.P.); (M.L.)
| | - Lilian Tzivian
- Institute of Clinical and Preventive Medicine, Faculty of Medicine, University of Latvia, 1007 Riga, Latvia; (D.R.); (I.P.); (O.S.); (L.T.); (I.K.); (S.I.); (I.L.-K.); (D.R.-E.); (S.P.); (M.L.)
| | - Ilze Kikuste
- Institute of Clinical and Preventive Medicine, Faculty of Medicine, University of Latvia, 1007 Riga, Latvia; (D.R.); (I.P.); (O.S.); (L.T.); (I.K.); (S.I.); (I.L.-K.); (D.R.-E.); (S.P.); (M.L.)
- Digestive Diseases Centre GASTRO, 1586 Riga, Latvia; (I.T.); (A.V.)
| | - Ivars Tolmanis
- Digestive Diseases Centre GASTRO, 1586 Riga, Latvia; (I.T.); (A.V.)
| | - Aigars Vanags
- Digestive Diseases Centre GASTRO, 1586 Riga, Latvia; (I.T.); (A.V.)
| | - Sergejs Isajevs
- Institute of Clinical and Preventive Medicine, Faculty of Medicine, University of Latvia, 1007 Riga, Latvia; (D.R.); (I.P.); (O.S.); (L.T.); (I.K.); (S.I.); (I.L.-K.); (D.R.-E.); (S.P.); (M.L.)
- Department of Research, Riga East University Hospital, 1038 Riga, Latvia
- Academic Histology Laboratory, 1073 Riga, Latvia
| | - Inta Liepniece-Karele
- Institute of Clinical and Preventive Medicine, Faculty of Medicine, University of Latvia, 1007 Riga, Latvia; (D.R.); (I.P.); (O.S.); (L.T.); (I.K.); (S.I.); (I.L.-K.); (D.R.-E.); (S.P.); (M.L.)
- Department of Research, Riga East University Hospital, 1038 Riga, Latvia
- Academic Histology Laboratory, 1073 Riga, Latvia
| | - Danute Razuka-Ebela
- Institute of Clinical and Preventive Medicine, Faculty of Medicine, University of Latvia, 1007 Riga, Latvia; (D.R.); (I.P.); (O.S.); (L.T.); (I.K.); (S.I.); (I.L.-K.); (D.R.-E.); (S.P.); (M.L.)
| | - Sergej Parshutin
- Institute of Clinical and Preventive Medicine, Faculty of Medicine, University of Latvia, 1007 Riga, Latvia; (D.R.); (I.P.); (O.S.); (L.T.); (I.K.); (S.I.); (I.L.-K.); (D.R.-E.); (S.P.); (M.L.)
| | - Raul Murillo
- Early Detection, Prevention and Infections Branch, International Agency for Research on Cancer, 69372 Lyon, France; (R.M.); (R.H.); (J.Y.P.)
- Centro Javeriano de Oncología, Hospital Universitario San Ignacio, Bogotá 11001, Colombia
| | - Rolando Herrero
- Early Detection, Prevention and Infections Branch, International Agency for Research on Cancer, 69372 Lyon, France; (R.M.); (R.H.); (J.Y.P.)
- Agencia Costarricense de Investigaciones Biomedicas, Fundacion INCIENSA, San Jose 2250, Costa Rica
| | - Jin Young Park
- Early Detection, Prevention and Infections Branch, International Agency for Research on Cancer, 69372 Lyon, France; (R.M.); (R.H.); (J.Y.P.)
| | - Marcis Leja
- Institute of Clinical and Preventive Medicine, Faculty of Medicine, University of Latvia, 1007 Riga, Latvia; (D.R.); (I.P.); (O.S.); (L.T.); (I.K.); (S.I.); (I.L.-K.); (D.R.-E.); (S.P.); (M.L.)
- Department of Research, Riga East University Hospital, 1038 Riga, Latvia
- Digestive Diseases Centre GASTRO, 1586 Riga, Latvia; (I.T.); (A.V.)
| |
Collapse
|
12
|
Rugge M. Big Data on Gastric Dysplasia Support Gastric Cancer Prevention. Clin Gastroenterol Hepatol 2022; 20:1226-1228. [PMID: 35123082 DOI: 10.1016/j.cgh.2022.01.038] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 01/19/2022] [Accepted: 01/20/2022] [Indexed: 02/07/2023]
Affiliation(s)
- Massimo Rugge
- Department of Medicine DIMED, Surgical Pathology and Cytopathology Unit, University of Padova, Padova, Italy; Azienda Zero Padova, Veneto Tumor Cancer Registry, Padova, Italy; Baylor College of Medicine, Houston, Texas
| |
Collapse
|
13
|
Razuka-Ebela D, Polaka I, Daugule I, Parshutin S, Santare D, Ebela I, Rudzite D, Vangravs R, Herrero R, Young Park J, Leja M. Factors Associated with False Negative Results in Serum Pepsinogen Testing for Precancerous Gastric Lesions in a European Population in the GISTAR Study. Diagnostics (Basel) 2022; 12:1166. [PMID: 35626319 PMCID: PMC9139962 DOI: 10.3390/diagnostics12051166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 05/02/2022] [Accepted: 05/04/2022] [Indexed: 01/27/2023] Open
Abstract
The accuracy of plasma pepsinogen (Pg) as a marker for precancerous gastric lesions (PGL) has shown variable results. We aimed to identify factors associated with false negative (FN) cases in Pg testing and to adjust cut-off values for these factors in order to improve Pg yield. Plasma Pg was measured and upper endoscopy with biopsy was performed within the "Multicentric randomized study of Helicobacter pylori eradication and pepsinogen testing for prevention of gastric cancer mortality: the GISTAR study". A multivariable logistic model was built for FN and multiple factors. Values of Pg were compared and sensitivity and specificity were calculated using pre-existing Pg cut-offs for factors showing strong associations with FN. New cut-offs were calculated for factors that showed substantially lower sensitivity. Of 1210 participants, 364 (30.1%) had histologically confirmed PGL, of which 160 (44.0%) were FN. Current smokers, men, and H. pylori positives were more likely FN. Smoking in H. pylori negatives was associated with a higher Pg I/II ratio and substantially lower sensitivity of Pg testing than in other groups. Adjusting Pg cut-offs for current smokers by H. pylori presence improved sensitivity for detecting PGL in this group. Our study suggests that adjusting Pg cut-offs for current smokers by H. pylori status could improve Pg test performance.
Collapse
Affiliation(s)
- Danute Razuka-Ebela
- Faculty of Medicine, University of Latvia, Jelgavas iela 3, LV-1004 Riga, Latvia; (I.D.); (D.S.); (I.E.); (M.L.)
- Institute of Clinical and Preventive Medicine, University of Latvia, Jelgavas iela 3, LV-1004 Riga, Latvia; (I.P.); (S.P.); (D.R.); (R.V.)
| | - Inese Polaka
- Institute of Clinical and Preventive Medicine, University of Latvia, Jelgavas iela 3, LV-1004 Riga, Latvia; (I.P.); (S.P.); (D.R.); (R.V.)
| | - Ilva Daugule
- Faculty of Medicine, University of Latvia, Jelgavas iela 3, LV-1004 Riga, Latvia; (I.D.); (D.S.); (I.E.); (M.L.)
- Institute of Clinical and Preventive Medicine, University of Latvia, Jelgavas iela 3, LV-1004 Riga, Latvia; (I.P.); (S.P.); (D.R.); (R.V.)
| | - Sergei Parshutin
- Institute of Clinical and Preventive Medicine, University of Latvia, Jelgavas iela 3, LV-1004 Riga, Latvia; (I.P.); (S.P.); (D.R.); (R.V.)
| | - Daiga Santare
- Faculty of Medicine, University of Latvia, Jelgavas iela 3, LV-1004 Riga, Latvia; (I.D.); (D.S.); (I.E.); (M.L.)
- Institute of Clinical and Preventive Medicine, University of Latvia, Jelgavas iela 3, LV-1004 Riga, Latvia; (I.P.); (S.P.); (D.R.); (R.V.)
| | - Inguna Ebela
- Faculty of Medicine, University of Latvia, Jelgavas iela 3, LV-1004 Riga, Latvia; (I.D.); (D.S.); (I.E.); (M.L.)
| | - Dace Rudzite
- Institute of Clinical and Preventive Medicine, University of Latvia, Jelgavas iela 3, LV-1004 Riga, Latvia; (I.P.); (S.P.); (D.R.); (R.V.)
- Riga East University Hospital, Hipokrāta iela 2, LV-1038 Riga, Latvia
| | - Reinis Vangravs
- Institute of Clinical and Preventive Medicine, University of Latvia, Jelgavas iela 3, LV-1004 Riga, Latvia; (I.P.); (S.P.); (D.R.); (R.V.)
| | - Rolando Herrero
- International Agency for Research on Cancer, 150 Cours Albert Thomas, CEDEX 08, 69372 Lyon, France; (R.H.); (J.Y.P.)
- Agencia Costarricense de Investigaciones Biomedicas, Fundación INCIENSA, Avenida 9a Calles 64-68, San Jose 2250, Costa Rica
| | - Jin Young Park
- International Agency for Research on Cancer, 150 Cours Albert Thomas, CEDEX 08, 69372 Lyon, France; (R.H.); (J.Y.P.)
| | - Marcis Leja
- Faculty of Medicine, University of Latvia, Jelgavas iela 3, LV-1004 Riga, Latvia; (I.D.); (D.S.); (I.E.); (M.L.)
- Institute of Clinical and Preventive Medicine, University of Latvia, Jelgavas iela 3, LV-1004 Riga, Latvia; (I.P.); (S.P.); (D.R.); (R.V.)
| |
Collapse
|
14
|
Yu T, Lin N, Zhong X, Zhang X, Zhang X, Chen Y, Liu J, Hu W, Duan H, Si J. Multi-label recognition of cancer-related lesions with clinical priors on white-light endoscopy. Comput Biol Med 2022; 143:105255. [PMID: 35151153 DOI: 10.1016/j.compbiomed.2022.105255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 01/07/2022] [Accepted: 01/20/2022] [Indexed: 11/28/2022]
Abstract
Deep learning-based computer-aided diagnosis techniques have demonstrated encouraging performance in endoscopic lesion identification and detection, and have reduced the rate of missed and false detections of disease during endoscopy. However, the interpretability of the model-based results has not been adequately addressed by existing methods. This phenomenon is directly manifested by a significant bias in the representation of feature localization. Good recognition models experience severe feature localization errors, particularly for lesions with subtle morphological features, and such unsatisfactory performance hinders the clinical deployment of models. To effectively alleviate this problem, we proposed a solution to optimize the localization bias in feature representations of cancer-related recognition models that is difficult to accurately label and identify in clinical practice. Optimization was performed in the training phase of the model through the proposed data augmentation method and auxiliary loss function based on clinical priors. The data augmentation method, called partial jigsaw, can "break" the spatial structure of lesion-independent image blocks and enrich the data feature space to decouple the interference of background features on the space and focus on fine-grained lesion features. The annotation-based auxiliary loss function used class activation maps for sample distribution correction and led the model to present localization representation converging on the gold standard annotation of visualization maps. The results show that with the improvement of our method, the precision of model recognition reached an average of 92.79%, an F1-score of 92.61%, and accuracy of 95.56% based on a dataset constructed from 23 hospitals. In addition, we quantified the evaluation representation of visualization feature maps. The improved model yielded significant offset correction results for visualized feature maps compared with the baseline model. The average visualization-weighted positive coverage improved from 51.85% to 83.76%. The proposed approach did not change the deployment capability and inference speed of the original model and can be incorporated into any state-of-the-art neural network. It also shows the potential to provide more accurate localization inference results and assist in clinical examinations during endoscopies.
Collapse
Affiliation(s)
- Tao Yu
- Key Laboratory for Biomedical Engineering of Ministry of Education, College of Biomedical Engineering and Instrument Science, Zhejiang University, Hangzhou, China
| | - Ne Lin
- Department of Gastroenterology, Sir Run Run Shaw Hospital, Medical School, Zhejiang University, Hangzhou, China
| | - Xingwei Zhong
- Department of Gastroenterology, Sir Run Run Shaw Hospital, Medical School, Zhejiang University, Hangzhou, China
| | - Xiaoyan Zhang
- Key Laboratory for Biomedical Engineering of Ministry of Education, College of Biomedical Engineering and Instrument Science, Zhejiang University, Hangzhou, China
| | - Xinsen Zhang
- Key Laboratory for Biomedical Engineering of Ministry of Education, College of Biomedical Engineering and Instrument Science, Zhejiang University, Hangzhou, China
| | - Yihe Chen
- Key Laboratory for Biomedical Engineering of Ministry of Education, College of Biomedical Engineering and Instrument Science, Zhejiang University, Hangzhou, China
| | - Jiquan Liu
- Key Laboratory for Biomedical Engineering of Ministry of Education, College of Biomedical Engineering and Instrument Science, Zhejiang University, Hangzhou, China.
| | - Weiling Hu
- Department of Gastroenterology, Sir Run Run Shaw Hospital, Medical School, Zhejiang University, Hangzhou, China; Institute of Gastroenterology, Zhejiang University, Hangzhou, China
| | - Huilong Duan
- Key Laboratory for Biomedical Engineering of Ministry of Education, College of Biomedical Engineering and Instrument Science, Zhejiang University, Hangzhou, China
| | - Jianmin Si
- Department of Gastroenterology, Sir Run Run Shaw Hospital, Medical School, Zhejiang University, Hangzhou, China; Institute of Gastroenterology, Zhejiang University, Hangzhou, China
| |
Collapse
|
15
|
Sobrino-Cossío S, Teramoto-Matsubara O, Emura F, Araya R, Arantes V, Galvis-García ES, Meza-Caballero M, García-Aguilar BS, Reding-Bernal A, Uedo N. Usefulness of optical enhancement endoscopy combined with magnification to improve detection of intestinal metaplasia in the stomach. Endosc Int Open 2022; 10:E441-E447. [PMID: 35433218 PMCID: PMC9010107 DOI: 10.1055/a-1759-2568] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Accepted: 01/11/2022] [Indexed: 11/19/2022] Open
Abstract
Background and study aims The light blue crest observed in narrow band imaging endoscopy has high diagnostic accuracy for diagnosis of gastric intestinal metaplasia (GIM). The objective of this prospective study was to evaluate the diagnostic accuracy of magnifying i-scan optical enhancement (OE) imaging for diagnosing the LBC sign in patients with different levels of risk for gastric cancer in a Mexican clinical practice. Patients and methods Patients with a history of peptic ulcer and symptoms of dyspepsia or gastroesophageal reflux disease were enrolled. Diagnosis of GIM was made at the predetermined anatomical location and white light endoscopy and i-scan OE Mode 1 were captured at the two predetermined biopsy sites (antrum and pyloric regions). Results A total of 328 patients were enrolled in this study. Overall GIM prevalence was 33.8 %. The GIM distribution was 95.4 % in the antrum and 40.5 % in the corpus. According to the Operative Link on Gastritis/Intestinal-Metaplasia Assessment staging system, only two patients (1.9 %) were classified with high-risk stage disease. Sensitivity, specificity, positive and negative predictive values, positive and negative likelihood ratios, and accuracy of both methods (95 % C. I.) were 0.50 (0.41-0.60), 0.55 (0.48-0.62), 0.36 (0.31-0.42), 0.68 (0.63-0.73), 1.12 (0.9-1.4), 0.9 (0.7-1.1), and 0.53 (0.43-0.60) for WLE, and 0.96 (0.90-0.99), 0.91 (0.86-0.94), 0.84 (0.78-0.89), 0.98 (0.94-0.99), 10.4 (6.8-16), 0.05 (0.02-0.12), and 0.93 (0.89-0.95), respectively. The kappa concordance was 0.67 and the reliability coefficient was 0.7407 for interobserver variability. Conclusions Our study demonstrated the high performance of magnifying i-scan OE imaging for endoscopic diagnosis of GIM in Mexican patients.
Collapse
Affiliation(s)
- Sergio Sobrino-Cossío
- Hospital Ángeles del Pedregal, Ciudad de Mexico, Mexico
- Gástrica, Centro Avanzado en Endoscopia y Estudios Funcionales, Mexico City, Mexico
| | - Oscar Teramoto-Matsubara
- Gástrica, Centro Avanzado en Endoscopia y Estudios Funcionales, Mexico City, Mexico
- Hospital ABC Ciudad de Mexico, Mexico
| | - Fabian Emura
- Endoscopia Gastrointestinal Avanzada, EmuraCenter Latinoamérica y Departamento de Gastroenterología de la Universidad de la Sabana, Bogotá, Colombia
- Departamento de Gastroenterología de la Universidad de la Sabana, Chia, Cundinamarca, Colombia
| | - Raúl Araya
- Servicio de Endoscopia y Gastroenterología de la Clínica Universidad de Los Andes y del Hospital Militar de Santiago y Clínica Universidad de los Andes, Santiago, Chile
| | - Vítor Arantes
- Hospital das Clínicas, Faculdade de Medicina, Universidade Federal de Minas Gerais, Hospital Mater Dei Contorno, Belo Horizonte, Brasil
| | - Elymir S. Galvis-García
- Department of Endoscopy. Hospital General de Mexico “Dr. Eduardo Liceaga,” Mexico City, Mexico
| | | | | | - Arturo Reding-Bernal
- Research Division, Hospital General de Mexico “Dr. Eduardo Liceaga,” Mexico City, Mexico
| | - Noriya Uedo
- Osaka International Cancer Institute, Department of Gastrointestinal Oncology, Osaka, Japan
| |
Collapse
|
16
|
Park JM, Han YM, Hahm KB. Rejuvenation of Helicobacter pylori-Associated Atrophic Gastritis Through Concerted Actions of Placenta-Derived Mesenchymal Stem Cells Prevented Gastric Cancer. Front Pharmacol 2021; 12:675443. [PMID: 34483897 PMCID: PMC8416416 DOI: 10.3389/fphar.2021.675443] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Accepted: 06/22/2021] [Indexed: 01/06/2023] Open
Abstract
Chronic Helicobacter pylori infection causes gastric cancer via the progression of precancerous chronic atrophic gastritis (CAG). Therefore, repairing gastric atrophy could be a useful strategy in preventing H. pylori-associated gastric carcinogenesis. Although eradication of the bacterial pathogen offers one solution to this association, this study was designed to evaluate an alternative approach using mesenchymal stem cells to treat CAG and prevent carcinogenesis. Here, we used human placenta-derived mesenchymal stem cells (PD-MSCs) and their conditioned medium (CM) to treat H. pylori-associated CAG in a mice/cell model to explore their therapeutic effects and elucidate their molecular mechanisms. We compared the changes in the fecal microbiomes in response to PD-MSC treatments, and chronic H. pylori-infected mice were given ten treatments with PD-MSCs before being sacrificed for end point assays at around 36 weeks of age. These animals presented with significant reductions in the mean body weights of the control group, which were eradicated following PD-MSC treatment (p < 0.01). Significant changes in various pathological parameters including inflammation, gastric atrophy, erosions/ulcers, and dysplastic changes were noted in the control group (p < 0.01), but these were all significantly reduced in the PD-MSC/CM-treated groups. Lgr5+, Ki-67, H+/K+-ATPase, and Musashi-1 expressions were all significantly increased in the treated animals, while inflammatory mediators, MMP, and apoptotic executors were significantly decreased in the PD-MSC group compared to the control group (p < 0.001). Our model showed that H. pylori-initiated, high-salt diet-promoted gastric atrophic gastritis resulted in significant changes in the fecal microbiome at the phylum/genus level and that PD-MSC/CM interventions facilitated a return to more normal microbial communities. In conclusion, administration of PD-MSCs or their conditioned medium may present a novel rejuvenating agent in preventing the progression of H. pylori-associated premalignant lesions.
Collapse
Affiliation(s)
- Jong Min Park
- College of Oriental Medicine, Daejeon University, Daejeon, Korea
| | - Young Min Han
- Western Seoul Center, Korea Basic Science Institute, Seoul, Korea
| | - Ki Baik Hahm
- Medpacto Research Institute, Medpacto, Seoul, Korea.,CHA Cancer Preventive Research Center, CHA Bio Complex, Seongnam, Korea
| |
Collapse
|
17
|
Helicobacter pylori-Induced Inflammation: Possible Factors Modulating the Risk of Gastric Cancer. Pathogens 2021; 10:pathogens10091099. [PMID: 34578132 PMCID: PMC8467880 DOI: 10.3390/pathogens10091099] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 08/21/2021] [Accepted: 08/25/2021] [Indexed: 02/07/2023] Open
Abstract
Chronic inflammation and long-term tissue injury are related to many malignancies, including gastric cancer (GC). Helicobacter pylori (H. pylori), classified as a class I carcinogen, induces chronic superficial gastritis followed by gastric carcinogenesis. Despite a high prevalence of H. pylori infection, only about 1–3% of people infected with this bacterium develop GC worldwide. Furthermore, the development of chronic gastritis in some, but not all, H. pylori-infected subjects remains unexplained. These conflicting findings indicate that clinical outcomes of aggressive inflammation (atrophic gastritis) to gastric carcinogenesis are influenced by several other factors (in addition to H. pylori infection), such as gut microbiota, co-existence of intestinal helminths, dietary habits, and host genetic factors. This review has five goals: (1) to assess our current understanding of the process of H. pylori-triggered inflammation and gastric precursor lesions; (2) to present a hypothesis on risk modulation by the gut microbiota and infestation with intestinal helminths; (3) to identify the dietary behavior of the people at risk of GC; (4) to check the inflammation-related genetic polymorphisms and role of exosomes together with other factors as initiators of precancerous lesions and gastric carcinoma; and (5) finally, to conclude and suggest a new direction for future research.
Collapse
|
18
|
Wang S, Ye F, Sheng Y, Yu W, Liu Y, Liu D, Zhang K. Development and Validation of Nomograms to Predict Operative Link for Gastritis Assessment Any-Stage and Stages III-IV in the Chinese High-Risk Gastric Cancer Population. Front Med (Lausanne) 2021; 8:724566. [PMID: 34447771 PMCID: PMC8383045 DOI: 10.3389/fmed.2021.724566] [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: 06/13/2021] [Accepted: 07/09/2021] [Indexed: 12/24/2022] Open
Abstract
Purpose: It is very essential to diagnose gastric atrophy in the area with high prevalence of gastric cancer. Operative link for gastritis assessment (OLGA) was developed to detect the severity of gastric atrophy. The aim of this study was to develop and validate nomograms for predicting OLGA any-stage and stages III-IV in the Chinese high-risk gastric cancer population. Methods: We retrospectively analyzed 7,945 participants obtained by a multicenter cross-sectional study. We randomly selected 55% individuals (4,370 participants, training cohort) to analyze and generate the prediction models and validated the models on the remaining individuals (3,575 participants, validation cohort). A multivariate logistic regression model was used to select variables in the training cohort. The corresponding nomograms were developed to predict OLGA any-stage and stages III-IV, respectively. The area under the receiver operating characteristic curves and the GiViTI calibration belts were used to estimate the discrimination and calibration of the prediction models. Results: There were 1,226 (28.05%) participants in the training sample and 970 (27.13%) in the validation sample who were diagnosed with gastric atrophy. The nomogram predicting OLGA any-stage had an area under the curve (AUC) of 0.610 for the training sample and 0.615 for the validation sample, with favorable calibrations in the overall population. Similarly, the nomogram predicting OLGA stages III-IV had an AUC of 0.702 and 0.714 for the training and validation samples, respectively, with favorable calibrations in the overall population. Conclusions: The prediction model can early identify the occurrence of gastric atrophy and the severity stage of gastric atrophy to some extent.
Collapse
Affiliation(s)
- Song Wang
- Department of Gastroenterology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Fei Ye
- Department of Gastroenterology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Yuan Sheng
- Department of Gastroenterology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Wenyong Yu
- Department of Gastroenterology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Yingling Liu
- Department of Gastroenterology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Dehua Liu
- Department of Gastroenterology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Kaiguang Zhang
- Department of Gastroenterology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| |
Collapse
|
19
|
Gonzalez RS, Raza A, Propst R, Adeyi O, Bateman J, Sopha SC, Shaw J, Auerbach A. Recent Advances in Digestive Tract Tumors: Updates From the 5th Edition of the World Health Organization "Blue Book". Arch Pathol Lab Med 2021; 145:607-626. [PMID: 32886739 DOI: 10.5858/arpa.2020-0047-ra] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/12/2020] [Indexed: 11/06/2022]
Abstract
CONTEXT.— The World Health Organization Classification of Tumours: Digestive System Tumors, 5th edition, was published in 2019 and shows several impactful changes as compared with the 4th edition published in 2010. Changes include a revised nomenclature of serrated lesions and revamping the classification of neuroendocrine neoplasms. Appendiceal goblet cell adenocarcinoma is heavily revised, and intrahepatic cholangiocarcinoma is split into 2 subtypes. New subtypes of colorectal carcinoma and hepatocellular carcinoma are described. Precursor lesions are emphasized with their own entries, and both dysplastic and invasive lesions are generally recommended to be graded using a 2-tier system. Hematolymphoid tumors, mesenchymal tumors, and genetic tumor syndromes each have their own sections in the 5th edition. New hematolymphoid lesions include monomorphic epitheliotropic intestinal T-cell lymphoma; duodenal-type follicular lymphoma; intestinal T-cell lymphoma, not otherwise specified; and indolent T-cell lymphoproliferative disorder of the gastrointestinal tract. This paper will provide an in-depth look at the changes in the 5th edition as compared with the 4th edition. OBJECTIVE.— To provide a comprehensive, in-depth update on the World Health Organization classification of digestive tumors, including changes to nomenclature, updated diagnostic criteria, and newly described entities. DATA SOURCES.— The 5th edition of the World Health Organization Classification of Tumours: Digestive System Tumours, as well as the 4th edition. CONCLUSIONS.— The World Health Organization has made many key changes in its newest update on tumors of the digestive system. Pathologists should be aware of these changes and incorporate them into their practice as able or necessary.
Collapse
Affiliation(s)
- Raul S Gonzalez
- The Department of Pathology, Beth Israel Deaconess Medical Center, Boston, Massachusetts (Gonzalez)
| | - Anwar Raza
- The Department of Pathology and Human Anatomy, Loma Linda University, Loma Linda, California (Raza, Propst)
| | - Robert Propst
- The Department of Pathology and Human Anatomy, Loma Linda University, Loma Linda, California (Raza, Propst)
| | - Oyedele Adeyi
- The Department of Pathology, University of Minnesota, Minneapolis (Adeyi, Bateman)
| | - Justin Bateman
- The Department of Pathology, University of Minnesota, Minneapolis (Adeyi, Bateman)
| | - Sabrina C Sopha
- The Department of Pathology, University of Maryland Baltimore Washington Medical Center, Glen Burnie (Sopha)
| | - Janet Shaw
- The Joint Pathology Center, Silver Spring, Maryland (Shaw, Auerbach)
| | - Aaron Auerbach
- The Joint Pathology Center, Silver Spring, Maryland (Shaw, Auerbach)
| |
Collapse
|
20
|
Yan T, Wong PK, Qin YY. Deep learning for diagnosis of precancerous lesions in upper gastrointestinal endoscopy: A review. World J Gastroenterol 2021; 27:2531-2544. [PMID: 34092974 PMCID: PMC8160615 DOI: 10.3748/wjg.v27.i20.2531] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2021] [Revised: 03/27/2021] [Accepted: 04/09/2021] [Indexed: 02/06/2023] Open
Abstract
Upper gastrointestinal (GI) cancers are the leading cause of cancer-related deaths worldwide. Early identification of precancerous lesions has been shown to minimize the incidence of GI cancers and substantiate the vital role of screening endoscopy. However, unlike GI cancers, precancerous lesions in the upper GI tract can be subtle and difficult to detect. Artificial intelligence techniques, especially deep learning algorithms with convolutional neural networks, might help endoscopists identify the precancerous lesions and reduce interobserver variability. In this review, a systematic literature search was undertaken of the Web of Science, PubMed, Cochrane Library and Embase, with an emphasis on the deep learning-based diagnosis of precancerous lesions in the upper GI tract. The status of deep learning algorithms in upper GI precancerous lesions has been systematically summarized. The challenges and recommendations targeting this field are comprehensively analyzed for future research.
Collapse
Affiliation(s)
- Tao Yan
- School of Mechanical Engineering, Hubei University of Arts and Science, Xiangyang 441053, Hubei Province, China
- Department of Electromechanical Engineering, University of Macau, Taipa 999078, Macau, China
| | - Pak Kin Wong
- Department of Electromechanical Engineering, University of Macau, Taipa 999078, Macau, China
| | - Ye-Ying Qin
- Department of Electromechanical Engineering, University of Macau, Taipa 999078, Macau, China
| |
Collapse
|
21
|
Research progress on gut microbiota in patients with gastric cancer, esophageal cancer, and small intestine cancer. Appl Microbiol Biotechnol 2021; 105:4415-4425. [PMID: 34037843 DOI: 10.1007/s00253-021-11358-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2021] [Revised: 05/11/2021] [Accepted: 05/16/2021] [Indexed: 12/12/2022]
Abstract
The pathogenesis of gut microbiota in humans can be indicated due to the wide application of techniques, such as 16S rRNA sequencing. Presently, several studies have found a significant difference in fecal flora between normal individuals and patients with gastric cancer. Although clinical research on the feedback mechanism of gastric flora and gut microbiota is lacking, clarifying the relationship between gut microbiota and the characteristics of cancer is significant for the early diagnosis of gastric cancer. This study was conducted to review the results of several studies in the past 5 years and analyze the intestinal bacteria in patients with gastric cancer and compare them with those in patients with esophageal and small intestine cancers. It was found that the gut microbiota in patients with gastric cancer was similar to that in patients with esophageal cancer. However, making an analysis and comparing the gut microbiota in patients with small intestine and gastric cancers was impossible due to the low incidence of small intestinal cancer. Our review summarized the research progress on using the gut microbiota for early screening for gastric cancer, and the results of this study will provide a further direction in this field. KEY POINTS: • We reviewed several relative mechanisms of the gut microbiota related to gastric cancer. • The gut microbiota in gastric, esophageal, and small intestine cancers are significantly different in types and quantity, and we have provided some tips for further research. • A prospective review of sequencing methods and study results on the gut microbiota in gastric, esophageal, and small intestine cancers was described.
Collapse
|
22
|
High-risk individuals for gastric cancer would be missed for surveillance without subtyping of intestinal metaplasia. Virchows Arch 2021; 479:679-686. [PMID: 33990867 DOI: 10.1007/s00428-021-03116-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Revised: 05/02/2021] [Accepted: 05/04/2021] [Indexed: 10/21/2022]
Abstract
The use of Operative Link on Gastritis Assessment (OLGA) and Operative Link on Gastritis Assessment based on Intestinal Metaplasia (OLGIM) staging system is recommended for identifying subjects at risk for developing gastric cancer; usually high-risk lesions are considered only as stages III and IV. Accumulating evidence suggests that incomplete intestinal metaplasia (IM) is important in the development of gastric cancer. Our aim has been to identify the prevalence of incomplete IM in patients with low-risk OLGA/OLGIM stages among a high-risk general population. Healthy adult volunteers aged 40-64 years were invited to undergo upper endoscopy within a regional GISTAR pilot study in Kazakhstan (n = 166). Gastric lesions were staged according to OLGA/OLGIM staging system. High iron diamine-alcian blue (HID-AB) was used for subtyping IM. IM prevalence overall was 45.8%. Incomplete IM was present in 52.6% (type II in 30.3% and type III in 22.3%), whereas complete IM was found in 47.4% individuals. The prevalence of OLGIM I and II stage were 39.8 and 4.8%, respectively, whereas OLGIM III was observed in 1.2%. The prevalence of incomplete IM in patients stratified to OLGIM I was 54.5% (type II in 31.8% and type III in 22.7%). High prevalence of incomplete IM was detected not only in subjects with extensive IM, but in those stratified as at the OLGIM I stage. Without IM subtyping, patients with high risk of gastric cancer development would be missed for surveillance.
Collapse
|
23
|
ERDAL H, SAKİN YS, ÇETİN M, ÇELİK B, ÖNMEZ A, UYGUN A, GÜLŞEN M. The Relationship Between Hyperplastic Gastric Polyps And Helicobacter Pylori. KONURALP TIP DERGISI 2021. [DOI: 10.18521/ktd.834666] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
|
24
|
Ebigbo A, Römmele C, Meisinger C, Probst A, Beyer A, Arnold J, Beer F, Endlicher E, Schiffelholz W, Linseisen J, Messmann H. Vorsorgegastroskopie in Deutschland – erste Ergebnisse der Pilotphase der Prä-GIT-Studie. TUMORDIAGNOSTIK & THERAPIE 2020; 41:604-609. [DOI: 10.1055/a-1267-3046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
Abstract
ZusammenfassungIm Rahmen der Prä-GIT-Studie sollen Informationen zur Prävalenz und zum Verlauf von präkanzerösen und frühmalignen Läsionen im oberen Gastrointestinaltrakt (OGIT) gewonnen sowie Hochrisikogruppen identifiziert werden. Studienteilnehmer erhalten im Rahmen einer regulären Vorsorgekoloskopie nach vorangegangener Einverständniserklärung in derselben Sitzung eine standardisierte hochauflösende Gastroskopie. Für die Hauptphase der Studie ist geplant, 5000 Teilnehmer in 30 gastroenterologischen Praxen in Bayern einzuschließen. Zur Vorbereitung wurde diese Pilotstudie durchgeführt, um die Machbarkeit des Studienprotokolls, die Akzeptanz der Untersuchung sowie die Durchführung in der „Praxis“-Realität zu prüfen.
Methodik Drei gastroenterologische Praxen in Bayern nahmen an der dreimonatigen Pilotphase von 10/2018 bis 12/2018 teil. Nach entsprechender Aufklärung wurde Patienten, die sich zur Vorsorgekoloskopie in diesen Praxen vorstellten, zusätzlich eine diagnostische Gastroskopie angeboten. Die Gastroskopien erfolgten nach einer vorgegebenen Arbeitsanweisung (Standard Operating Procedure, SOP). Zusätzlich erfolgten eine Datenerhebung zu Ernährung und Lebensstil anhand eines Selbstausfüllfragebogens sowie die Sammlung von Bioproben.
Ergebnisse Innerhalb von 3 Monaten wurden 52 Probanden in drei Praxen in Bayern in die Studie eingeschlossen. Das durchschnittliche Alter lag bei 63,4 Jahren. Die durchschnittliche Untersuchungszeit lag bei 11 Minuten, Komplikationen traten keine auf. Ein Teilnehmer zeigte einen Polyp im Bereich des Recessus piriformis, 21 Teilnehmer zeigten eine Refluxösophagitis, vier Teilnehmer einen Barrett-Ösophagus, davon einer histologisch einen Barrett-Ösophagus mit LGIEN, ein Teilnehmer wies einen Ösophaguspolyp mit leichtgradiger Plattenepitheldysplasie auf, 12 Teilnehmer eine mit Helicobacter pylori assoziierte Gastritis. Eine korpusdominante Atrophie wurde in einem Fall und ein Duodenal-Adenom mit LGIEN in zwei Fällen beschrieben. 100 % bzw. 89 % der Teilnehmer stellten eine Blut- bzw. eine Stuhlprobe für weitere Untersuchungen zur Verfügung. Alle Teilnehmer füllten zudem den Patientenfragebogen aus.
Zusammenfassung Durch die Prä-GIT-Studie soll erstmals für Deutschland eine solide Datenbasis generiert werden, um die Häufigkeiten verschiedener prämaligner und frühkanzeröser Veränderungen im oberen Gastrointestinaltrakt valide schätzen zu können. Die jetzige Pilotphase liefert wichtige Erkenntnisse zur Machbarkeit sowie zur Verbesserung der Prozesse im Rahmen der Hauptphase der Studie. Erfreulicherweise zeigte sich eine sehr große Akzeptanz der Teilnahme an der Studie.
Collapse
Affiliation(s)
- Alanna Ebigbo
- III. Medizinische Klinik, Universitätsklinikum Augsburg
| | | | | | | | - Albert Beyer
- Praxis für Gastroenterologie und gastrointestinale Onkologie, Altötting
| | - Jochen Arnold
- Praxis Dr. Arnold, Dr. Beer, Prof. Dr. Endlicher, Prof. Dr. Kreuser, Regensburg
| | - Franz Beer
- Praxis Dr. Arnold, Dr. Beer, Prof. Dr. Endlicher, Prof. Dr. Kreuser, Regensburg
| | - Esther Endlicher
- Praxis Dr. Arnold, Dr. Beer, Prof. Dr. Endlicher, Prof. Dr. Kreuser, Regensburg
| | | | | | | |
Collapse
|
25
|
Gastric cancer screening in low incidence populations: Position statement of AEG, SEED and SEAP. GASTROENTEROLOGIA Y HEPATOLOGIA 2020; 44:67-86. [PMID: 33252332 DOI: 10.1016/j.gastrohep.2020.08.004] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 08/06/2020] [Accepted: 08/31/2020] [Indexed: 02/06/2023]
Abstract
This positioning document, sponsored by the Asociación Española de Gastroenterología, the Sociedad Española de Endoscopia Digestiva and the Sociedad Española de Anatomía Patológica, aims to establish recommendations for the screening of gastric cancer (GC) in low incidence populations, such as the Spanish. To establish the quality of the evidence and the levels of recommendation, we used the methodology based on the GRADE system (Grading of Recommendations Assessment, Development and Evaluation). We obtained a consensus among experts using a Delphi method. The document evaluates screening in the general population, individuals with relatives with GC and subjects with GC precursor lesions (GCPL). The goal of the interventions should be to reduce GC related mortality. We recommend the use of the OLGIM classification and determine the intestinal metaplasia (IM) subtype in the evaluation of GCPL. We do not recommend to establish endoscopic mass screening for GC or Helicobacter pylori. However, the document strongly recommends to treat H.pylori if the infection is detected, and the investigation and treatment in individuals with a family history of GC or with GCPL. Instead, we recommend against the use of serological tests to detect GCPL. Endoscopic screening is suggested only in individuals that meet familial GC criteria. As for individuals with GCPL, endoscopic surveillance is only suggested in extensive IM associated with additional risk factors (incomplete IM and/or a family history of GC), after resection of dysplastic lesions or in patients with dysplasia without visible lesion after a high quality gastroscopy with chromoendoscopy.
Collapse
|
26
|
Lahner E, Marzinotto I, Lampasona V, Dottori L, Bazzigaluppi E, Brigatti C, Secchi M, Piemonti L, Conti L, Pilozzi E, Davidson HW, Wenzlau JM, Annibale B. Autoantibodies Toward ATP4A and ATP4B Subunits of Gastric Proton Pump H+,K+-ATPase Are Reliable Serological Pre-endoscopic Markers of Corpus Atrophic Gastritis. Clin Transl Gastroenterol 2020; 11:e00240. [PMID: 33031196 PMCID: PMC7544173 DOI: 10.14309/ctg.0000000000000240] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
INTRODUCTION Noninvasive assessment of corpus atrophic gastritis (CAG), a condition at increased risk of gastric cancer, is based on the measurement of pepsinogens, gastrin, and Helicobacter pylori antibodies. Parietal cell autoantibodies (PCAs) against the gastric proton pump (ATP4) are potential serological biomarkers of CAG. The purpose of this study was to compare the diagnostic performance of PCA and pepsinogen I tests in patients with clinical suspicion of CAG with the histopathological evaluation of gastric biopsies as reference standard. METHODS A prospective case-finding study was performed on 218 naive adult patients (131 women, median age 65 years) who underwent gastric biopsies to confirm/exclude CAG. Patients with histopathological CAG were defined as cases, conversely as controls. Autoantibodies against the individual alpha (ATP4A) and beta (ATP4B) subunits of ATP4 were measured by luciferase immunoprecipitation, and global PCA and pepsinogen I by enzyme-linked immunosorbent assay. RESULTS Histopathology classified 107 subjects (49%) as cases (CAG+, autoimmune 81.2%, and multifocal extensive 18.8%) and 111 subjects (51%) as controls (CAG-). In cases, ATP4A, ATP4B, and PCA titers were increased compared with controls, whereas pepsinogen I was reduced (P < 0.0001 for all). ATP4B, ATP4A, and pepsinogen I tests showed sensitivities of 77%, 75%, and 73% and specificities of 88%, 88%, and 80%, respectively. The receiver operating characteristic (ROC) area under the ROC curve (AUC) of these serological biomarkers confirmed their ability to discriminate cases from controls (ATP4B = 0.838, ATP4A = 0.826, pepsinogen I = 0.775, and PCA = 0.805), whereas the partial ROC-pAUC90 analysis showed that the ATP4B test had the best diagnostic performance (P = 0.008 vs ATP4; P = 0.0002 vs pepsinogen I). The presence of autoimmune or extensive gastritis was not significantly different between ATP4B positive or negative cases (P = 0.217). DISCUSSION PCAs are promising serological biomarkers for the identification of CAG in high-risk individuals, particularly in an autoimmune pattern but also in an extensive-multifocal atrophy pattern.
Collapse
Affiliation(s)
- Edith Lahner
- Medical-Surgical Department of Clinical Sciences and Translational Medicine, Sant'Andrea Hospital, University Sapienza, Rome, Italy
| | - Ilaria Marzinotto
- San Raffaele Diabetes Research Institute, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Vito Lampasona
- San Raffaele Diabetes Research Institute, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Ludovica Dottori
- Medical-Surgical Department of Clinical Sciences and Translational Medicine, Sant'Andrea Hospital, University Sapienza, Rome, Italy
| | - Elena Bazzigaluppi
- San Raffaele Diabetes Research Institute, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Cristina Brigatti
- San Raffaele Diabetes Research Institute, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Massimiliano Secchi
- San Raffaele Diabetes Research Institute, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Lorenzo Piemonti
- San Raffaele Diabetes Research Institute, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Laura Conti
- Medical-Surgical Department of Clinical Sciences and Translational Medicine, Sant'Andrea Hospital, University Sapienza, Rome, Italy
| | | | - Howard W. Davidson
- Barbara Davis Center for Diabetes, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Janet M. Wenzlau
- Barbara Davis Center for Diabetes, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Bruno Annibale
- Medical-Surgical Department of Clinical Sciences and Translational Medicine, Sant'Andrea Hospital, University Sapienza, Rome, Italy
| |
Collapse
|
27
|
Ge X, Zhang X, Ma Y, Chen S, Chen Z, Li M. Diagnostic value of macrophage inhibitory cytokine 1 as a novel prognostic biomarkers for early gastric cancer screening. J Clin Lab Anal 2020; 35:e23568. [PMID: 32918498 PMCID: PMC7843257 DOI: 10.1002/jcla.23568] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 08/11/2020] [Accepted: 08/19/2020] [Indexed: 01/13/2023] Open
Abstract
BACKGROUND Early diagnosis is very important to improve the survival rate of patients with gastric cancer (GC), especially in asymptomatic participants. However, low sensitivity of common biomarkers has caused difficulties in early screening of GC. In this study, we explored whether MIC-1 can improve the detection rate of early GC. METHODS We screened 8257 participants based on risk factors such as age, gender, and family history for physical examination including gastroscopy. Participant blood samples were taken for measure MIC-1, CA-199, CA72-4, and PG1/PG2 levels. The diagnostic performance of MIC-1 was assessed and compared with CA-199, CA72-4, and PG1/PG2, and its role in early GC diagnosis and the assessment of the risk of precancerous lesions have also been studied. RESULTS Based on endoscopic and histopathological findings, 55 participants had GC, 566 participants had low-grade neoplasia, and 2605 participants had chronic gastritis. MIC-1 levels were significantly elevated in GC serum samples as compared to controls (P < .001). The sensitivity of serum MIC-1 for GC diagnosis was much higher than that of CA-199 (49.1% vs 20.0%) with similar specificities. Moreover, receiver operating characteristic (ROC) curve analysis also showed that serum MIC-1 had a better performance compared with CA-199, CA72-4, and PG1/PG2 in distinguishing early-stage GC (AUC: 72.9% vs 69.5%, 67.5%, 44.0%, respectively). CONCLUSIONS Serum MIC-1 is significantly elevated in most patients with early GC. MIC-1 can serve as a novel diagnostic marker of early GC and value the risk of GC.
Collapse
Affiliation(s)
- Xin Ge
- Department of Laboratory Diagnostics, The First Affiliated Hospital of University of Science and Technology of China, Hefei, P.R. China
| | - Xiaolei Zhang
- Department of Laboratory Diagnostics, The First Affiliated Hospital of University of Science and Technology of China, Hefei, P.R. China
| | - Yanling Ma
- Anhui Office for Cancer Control and Research, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, P.R. China
| | - Shaohua Chen
- Department of Laboratory Diagnostics, The First Affiliated Hospital of University of Science and Technology of China, Hefei, P.R. China
| | - Zhaowu Chen
- Department of Laboratory Diagnostics, The First Affiliated Hospital of University of Science and Technology of China, Hefei, P.R. China
| | - Ming Li
- Department of Laboratory Diagnostics, The First Affiliated Hospital of University of Science and Technology of China, Hefei, P.R. China
| |
Collapse
|
28
|
Lenti MV, Rugge M, Lahner E, Miceli E, Toh BH, Genta RM, De Block C, Hershko C, Di Sabatino A. Autoimmune gastritis. Nat Rev Dis Primers 2020; 6:56. [PMID: 32647173 DOI: 10.1038/s41572-020-0187-8] [Citation(s) in RCA: 167] [Impact Index Per Article: 33.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/26/2020] [Indexed: 02/07/2023]
Abstract
Autoimmune gastritis (AIG) is an increasingly prevalent, organ-specific, immune-mediated disorder characterized by the destruction of gastric parietal cells, leading to the loss of intrinsic factor and reduced acid output. These alterations result in malabsorption of iron, vitamin B12 (pernicious anaemia) and potentially other micronutrients. For several years, most studies have focused on pernicious anaemia only, generating confusion between the two entities. In AIG, the gastric proton pump, H+/K+ ATPase, is the major autoantigen recognized by autoreactive T cells. The T cell-dependent activation of B cells stimulates the production of anti-parietal cell antibodies, the serological hallmark of AIG. The role of Helicobacter pylori infection in activating or favouring the autoimmune process is still uncertain. Early histopathological alterations allowing a more precise and prompt recognition have recently been described. AIG is burdened by a substantial diagnostic delay as it can present with varied clinical signs including, among others, gastrointestinal symptoms and neuropsychiatric manifestations. In advanced stages, AIG might progress to neuroendocrine tumours and gastric adenocarcinoma. Management includes early detection through a proactive case-finding strategy, micronutrient supplementation and endoscopic surveillance. This Primer comprehensively describes the most important insights regarding the epidemiology, pathophysiology, diagnosis and management of AIG, focusing on the most controversial, outstanding issues and future directions.
Collapse
Affiliation(s)
- Marco Vincenzo Lenti
- Department of Internal Medicine, IRCCS San Matteo Hospital Foundation, University of Pavia, Pavia, Italy
| | - Massimo Rugge
- Department of Medicine (DIMED), Surgical Pathology & Cytopathology Unit, University of Padua, Padua, Italy
| | - Edith Lahner
- Department of Surgical-Medical Sciences and Translational Medicine, Digestive and Liver Disease Unit, Sant'Andrea Hospital, Sapienza University of Rome, Rome, Italy
| | - Emanuela Miceli
- Department of Internal Medicine, IRCCS San Matteo Hospital Foundation, University of Pavia, Pavia, Italy
| | - Ban-Hock Toh
- Centre for Inflammatory Diseases, Monash Health, Faculty of Medicine, Nursing and Health Sciences, Monash University, Clayton, Victoria, Australia
| | - Robert M Genta
- Department of Medicine, Michael E. DeBakey VA Medical Center, Baylor College of Medicine, Houston, Texas, USA
| | - Christophe De Block
- Department of Endocrinology, Diabetology and Metabolism, Faculty of Medicine, Antwerp University Hospital and University of Antwerp, Antwerpen, Belgium
| | - Chaim Hershko
- Department of Hematology, Shaare Zedek Medical Center, Hebrew University of Jerusalem, Jerusalem, Israel.,Hematology Clinic and Central Clinical Laboratories, Clalit Health Services, Jerusalem, Israel
| | - Antonio Di Sabatino
- Department of Internal Medicine, IRCCS San Matteo Hospital Foundation, University of Pavia, Pavia, Italy.
| |
Collapse
|
29
|
Ebigbo A, Römmele C, Meisinger C, Probst A, Beyer A, Arnold J, Beer F, Endlicher E, Schiffelholz W, Linseisen J, Messmann H. [Screening gastroscopy in Germany - first results of the pilot phase of the Prä-GIT study]. ZEITSCHRIFT FUR GASTROENTEROLOGIE 2020; 58:761-766. [PMID: 32572870 DOI: 10.1055/a-1190-5332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
The primary aim of the Prä-GIT study is to collect information on the prevalence and course of precancerous and early malignant lesions in the upper GIT (uGIT) as well as risk factors associated with these lesions in asymptomatic individuals. Study participants presenting for a screening colonoscopy will undergo an additional endoscopic examination of the uGIT in the same session. 5000 participants in 30 endoscopy clinics in Bayern will be included. The pilot study presented here was performed to test the main study protocol as well as the acceptance of an additional uGIT endoscopic examination. METHODS Three endoscopy clinics in Bayern took part in the three-month pilot study between October and December 2018. Patients presenting for a screening colonoscopy in these clinics were offered an additional endoscopic examination of the uGIT which was performed according to a standard operating procedure (SOP). Furthermore, data on dietary habits and lifestyle, as well as biological samples, were collected. RESULTS 52 participants were included in three clinics in Altötting, Augsburg and Regensburg. The average age was 63.4 years. The average time spent per uGIT endoscopy was 11 minutes. No complications occurred. One participant showed a polyp of the recessus piriformis. Refluxesophagitis was seen in 21 participants, four participants had Barrett's esophagus with histological evidence of low-grade dysplasia in one patient, and an esophageal polyp with low-grade dysplasia was seen in one participant. Helicobacter-pylori gastritis was documented in 12 participants. Corpus-dominant atrophy was described in one participant, while a duodenal adenoma with low-grade dysplasia was seen in two participants. 100 % and 89 % of study participants gave a blood or a stool sample, respectively. All participants answered the questionnaire on dietary and lifestyle habits. CONCLUSION The Prä-GIT study was designed to generate a database for premalignant and early cancerous lesions of the uGIT in Germany in asymptomatic individuals presenting for a regular screening colonoscopy. The current pilot study has delivered important insights into the feasibility of the main study protocol. Additionally, the pilot study has shown a high rate of acceptance of an additional uGIT examination in this study population.
Collapse
Affiliation(s)
- Alanna Ebigbo
- III. Medizinische Klinik, Universitätsklinikum Augsburg
| | | | | | | | - Albert Beyer
- Praxis für Gastroenterologie und gastrointestinale Onkologie, Altötting
| | - Jochen Arnold
- Praxis Dr. Arnold, Dr. Beer, Prof. Dr. Endlicher, Prof. Dr. Kreuser, Regensburg
| | - Franz Beer
- Praxis Dr. Arnold, Dr. Beer, Prof. Dr. Endlicher, Prof. Dr. Kreuser, Regensburg
| | - Esther Endlicher
- Praxis Dr. Arnold, Dr. Beer, Prof. Dr. Endlicher, Prof. Dr. Kreuser, Regensburg
| | | | | | | |
Collapse
|
30
|
Pepsinogen Serology and Gastritis OLGA Staging in Mucosal Atrophy Assessment: A Cross-Sectional Study Involving East China Endoscopy Population. Gastroenterol Res Pract 2020; 2020:2324505. [PMID: 32351553 PMCID: PMC7174926 DOI: 10.1155/2020/2324505] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Accepted: 12/24/2019] [Indexed: 01/17/2023] Open
Abstract
The histological gastric stage (OLGA) plays an important role in evaluating gastric atrophy, a symptom which suggests a risk of gastric cancer (GC). However, gastroscopy is an invasive examination, which has limited application in populations undergoing physical examination. Serum pepsinogen (PG) and gastin-17 (G-17) levels are noninvasive indexes which are recommended when screening for GC. We aim to explore the correlation between PG/G-17 and OLGA stage, in order to provide reliable indexes for GC screening. The study included 453 asymptomatic individuals from East China undergoing physical examination, who then underwent endoscopy including collection of biopsy samples. Assays for serum PG, G-17, and Helicobacter pylori (Hp) were performed. Atrophy of gastric mucosa was graded according to OLGA for each individual. 453 participants, average age 52.46 ± 10.30 years, 253 male and 200 female, were included. In the asymptomatic physical examination population, serum PGI, PGII, and PGR levels decreased with increasing OLGA scores. PGI and PGR were inversely correlated with increasing OLGA stage in both Hp-positive and Hp-negative groups. The levels of serum PGI, PGII, and G-17 in the Hp-positive group were higher than those in the Hp-negative group; conversely, the PGR levels were lower. Furthermore, OLGA scores increased with age in the Hp-positive group. In conclusion, there is a significant correlation between OLGA stage and serum PG in populations from East China undergoing physical examination. Serum PG and G-17 combined with Hp test plays an important role in evaluating gastric atrophy.
Collapse
|
31
|
Huang RJ, Choi AY, Truong CD, Yeh MM, Hwang JH. Diagnosis and Management of Gastric Intestinal Metaplasia: Current Status and Future Directions. Gut Liver 2020; 13:596-603. [PMID: 31394893 PMCID: PMC6860040 DOI: 10.5009/gnl19181] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Revised: 07/08/2019] [Accepted: 07/08/2019] [Indexed: 12/13/2022] Open
Abstract
Gastric intestinal metaplasia (GIM) is a known premalignant condition of the human stomach along the pathway to gastric cancer (GC). Histologically, GIM represents the replacement of normal gastric mucosa by mucin-secreting intestinal mucosa. Helicobacter pylori infection is the most common etiologic agent of GIM development worldwide. The prevalence of GIM is heterogeneous among different regions of the world and correlates with the population endemicity of H. pylori carriage, among other environmental factors. GC remains the third leading cause of cancer-related mortality globally. GIM is usually diagnosed by upper endoscopy with biopsy, and histologic scoring systems have been developed to risk-stratify patients at highest risk for progression to GC. Several recent endoscopic imaging modalities may improve the optical detection of GIM and early GC. Appropriate surveillance of GIM may be cost effective and represents an opportunity for the early diagnosis and therapy of GC. Certain East Asian nations have established population-level programs for the screening and surveillance of GIM; guidelines regarding GIM surveillance have also recently been published in Europe. By contrast, few data exist regarding the appropriateness of surveillance of GIM in the United States. In this review, we discuss the pathogenesis, epidemiology, diagnosis, and management of GIM with an emphasis on the role of appropriate endoscopic surveillance.
Collapse
Affiliation(s)
- Robert J Huang
- Division of Gastroenterology and Hepatology, Stanford University, Stanford, CA, USA
| | - Alyssa Y Choi
- Departments of Medicine, University of Washington, Seattle, WA, USA
| | - Camtu D Truong
- Departments of Pathology, University of Washington, Seattle, WA, USA
| | - Matthew M Yeh
- Departments of Pathology, University of Washington, Seattle, WA, USA
| | - Joo Ha Hwang
- Division of Gastroenterology and Hepatology, Stanford University, Stanford, CA, USA
| |
Collapse
|
32
|
Abstract
Introduction: Atrophic gastritis (AG) is a complex syndrome which arises as a consequence of H. pylori infection or in the context of gastric autoimmunity. It often deserves a benign course but may lead to potentially life-threatening complications: cancer and anemia. This review aims to address traditional and innovative knowledge on this often under-diagnosed disorder.Areas covered: This review covers clinical presentation, risk factors, diagnosis, and management of AG and provides an updated resource for clinicians to get insight into this challenging disorder. Updated literature was searched in PubMed. Manual search from reference lists of publications was performed.Expert opinion: A case-finding strategy may be beneficial in individuals with anemia, dyspepsia, autoimmune thyropaties and type 1 diabetes, and family history of gastric cancer. AG is linked to gastric cancer risk and endoscopic surveillance is indicated according to topography of gastric atrophy and risk factors. The direction for future research in AG is summarized.
Collapse
Affiliation(s)
- Bruno Annibale
- Department of Medical-Surgical Sciences and Translational Medicine, Sant'Andrea Hospital, Sapienza University of Rome, Roma, Italy
| | - Gianluca Esposito
- Department of Medical-Surgical Sciences and Translational Medicine, Sant'Andrea Hospital, Sapienza University of Rome, Roma, Italy
| | - Edith Lahner
- Department of Medical-Surgical Sciences and Translational Medicine, Sant'Andrea Hospital, Sapienza University of Rome, Roma, Italy
| |
Collapse
|
33
|
Mezmale L, Isajevs S, Bogdanova I, Polaka I, Krigere A, Rudzite D, Rudule A, Kikuste I, Parshutin S, Tazhedinov A, Mushinskiy D, Sametayev D, Belikhina T, Igissinov N, Park JY, Herrero R, Leja M. Prevalence of Atrophic Gastritis in Kazakhstan and the Accuracy of Pepsinogen Tests to Detect Gastric Mucosal Atrophy. Asian Pac J Cancer Prev 2019; 20:3825-3829. [PMID: 31870128 PMCID: PMC7173362 DOI: 10.31557/apjcp.2019.20.12.3825] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Accepted: 12/08/2019] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Atrophic gastritis is considered precursor condition for gastric cancer. There is so far limited evidence on the performance of pepsinogens for atrophy detection in Central Asia. The aim of our study was to detect the prevalence of atrophic gastritis in the asymptomatic adult population in Kazakhstan as well as address the accuracy of pepsinogen testing in atrophy detection. METHODS Healthy individuals aged 40-64 were included. Upper endoscopy and pepsinogens (PG) evaluation were performed. PG were analysed in plasma by latex agglutination. Cut off values were used to define decreased PG values (PGR ≤ 3 and PG I ≤ 70 ng/mL); severely decreased PG values (PGR ≤ 2 and PG I ≤ 30 ng/mL). Biopsies were analyzed and obtained according to the updated Sydney System. PG test sensitivity, specificity and overall accuracy were assessed using the histological diagnosis as the "gold standard". RESULTS Altogether 157 individuals - female 40,1% and male 59,9% were included. Histologically, moderate to severe corpus atrophy, was present only in 1,3% cases. From all study subjects, 26,8% had decreased plasma PG values with cut-off values PGR ≤ 3 and PG I ≤ 70 ng/mL. The sensitivity of the PG test with this cut-off values was 50,0%, specificity 73,5%, overall accuracy 73,2% for detection of moderate to severe atrophy in the corpus. The sensitivity of PG test with cut-off values PGR ≤ 2 and PG I ≤30 ng/mL was 50,0%, specificity 90,9% and overall accuracy 90,4%. CONCLUSIONS The prevalence of gastric mucosal atrophy was low in the Kazakh population. Serological PG test screening nevertheless can play an important role in the diagnosis of gastric precancerous lesions. However, the diagnostic accuracy of the PG test is mainly dependent on the cut-off values for positive results.
Collapse
Affiliation(s)
- Linda Mezmale
- Institute of Clinical and Preventive Medicine,
- Faculty of Medicine, University of Latvia,
| | - Sergejs Isajevs
- Institute of Clinical and Preventive Medicine,
- Academic Histology Laboratory,
| | - Inga Bogdanova
- Institute of Clinical and Preventive Medicine,
- Academic Histology Laboratory,
| | | | | | | | - Aiga Rudule
- Institute of Clinical and Preventive Medicine,
| | - Ilze Kikuste
- Institute of Clinical and Preventive Medicine,
- Faculty of Medicine, University of Latvia,
- Digestive Disease Centre GASTRO, Riga, Latvia,
| | | | | | | | | | | | - Nurbek Igissinov
- Astana Medical University,
- Central Asian Cancer Institute, Nur-Sultan, Kazakhstan,
- International High School of Medicine,
- Eurasian Institute For Cancer Research, Bishkek, Kyrgyzstan,
| | - Jin Young Park
- Prevention and Implementation Group, Section of Early Detection and Prevention, International Agency for Research on Cancer, Lyon, France.
| | - Rolando Herrero
- Prevention and Implementation Group, Section of Early Detection and Prevention, International Agency for Research on Cancer, Lyon, France.
| | - Marcis Leja
- Institute of Clinical and Preventive Medicine,
- Faculty of Medicine, University of Latvia,
- Digestive Disease Centre GASTRO, Riga, Latvia,
| |
Collapse
|
34
|
Lahner E, Zagari RM, Zullo A, Di Sabatino A, Meggio A, Cesaro P, Lenti MV, Annibale B, Corazza GR. Chronic atrophic gastritis: Natural history, diagnosis and therapeutic management. A position paper by the Italian Society of Hospital Gastroenterologists and Digestive Endoscopists [AIGO], the Italian Society of Digestive Endoscopy [SIED], the Italian Society of Gastroenterology [SIGE], and the Italian Society of Internal Medicine [SIMI]. Dig Liver Dis 2019; 51:1621-1632. [PMID: 31635944 DOI: 10.1016/j.dld.2019.09.016] [Citation(s) in RCA: 99] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Revised: 09/03/2019] [Accepted: 09/23/2019] [Indexed: 02/08/2023]
Abstract
Chronic atrophic gastritis (CAG) is an underdiagnosed condition characterised by translational features going beyond the strict field of gastroenterology as it may manifest itself by a variable spectrum of gastric and extra-gastric symptoms and signs. It is relatively common among older adults in different parts of the world, but large variations exist. Helicobacter pylori-related CAG [multifocal] and autoimmune CAG (corpus-restricted) are apparently two different diseases, but they display overlapping features. Patients with cobalamin and/or iron deficiency anaemia or autoimmune disorders, including autoimmune thyroiditis and type 1 diabetes mellitus, should be offered screening for CAG. Pepsinogens, gastrin-17, and anti-H. pylori antibodies serum assays seem to be reliable non-invasive screening tools for the presence of CAG, helpful to identify individuals to refer to gastroscopy with five standard gastric biopsies in order to obtain histological confirmation of diagnosis. Patients with CAG are at increased risk of developing gastric cancer, and they should be estimated with histological staging systems (OLGA or OLGIM). H. pylori eradication may be beneficial by modifying the natural history of atrophy, but not that of intestinal metaplasia. Patients with advanced stages of CAG (Stage III/IV OLGA or OLGIM) should undergo endoscopic surveillance every three years, those with autoimmune CAG every three-five years. In patients with CAG, a screening for autoimmune thyroid disease and micronutrient deficiencies, including iron and vitamin B12, should be performed. The optimal treatment for dyspeptic symptoms in patients with CAG remains to be defined. Proton pump inhibitors are not indicated in hypochlorhydric CAG patients.
Collapse
Affiliation(s)
- Edith Lahner
- Department of Surgical-Medical Sciences and Translational Medicine, Digestive and Liver Disease Unit, Sant'Andrea Hospital, Sapienza University of Rome, Rome, Italy.
| | | | - Angelo Zullo
- Gastroenterology and Digestive Endoscopy, 'Nuovo Regina Margherita' Hospital, Rome, Italy
| | - Antonio Di Sabatino
- First Department of Internal Medicine, IRCCS San Matteo Hospital Foundation, University of Pavia, Pavia, Italy
| | - Alberto Meggio
- Department of Gastroenterology, Trento and Rovereto Hospital, Trento, Italy
| | - Paola Cesaro
- Digestive Endoscopy Unit and Gastroenterology, Fondazione Poliambulanza, Brescia, Italy
| | - Marco Vincenzo Lenti
- First Department of Internal Medicine, IRCCS San Matteo Hospital Foundation, University of Pavia, Pavia, Italy
| | - Bruno Annibale
- Department of Surgical-Medical Sciences and Translational Medicine, Digestive and Liver Disease Unit, Sant'Andrea Hospital, Sapienza University of Rome, Rome, Italy
| | - Gino Roberto Corazza
- First Department of Internal Medicine, IRCCS San Matteo Hospital Foundation, University of Pavia, Pavia, Italy
| |
Collapse
|
35
|
Liao Z, Tan ZW, Zhu P, Tan NS. Cancer-associated fibroblasts in tumor microenvironment – Accomplices in tumor malignancy. Cell Immunol 2019; 343:103729. [DOI: https:/doi.org/10.1016/j.cellimm.2017.12.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/02/2023]
|
36
|
Negovan A, Iancu M, Fülöp E, Bănescu C. Helicobacter pylori and cytokine gene variants as predictors of premalignant gastric lesions. World J Gastroenterol 2019; 25:4105-4124. [PMID: 31435167 PMCID: PMC6700706 DOI: 10.3748/wjg.v25.i30.4105] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2019] [Revised: 07/12/2019] [Accepted: 07/19/2019] [Indexed: 02/06/2023] Open
Abstract
Gastric cancer remains the third leading cause of mortality from cancer worldwide and carries a poor prognosis, due largely to late diagnosis. The importance of the interaction between Helicobacter pylori (H. pylori) infection, the main risk factor, and host-related genetic factors has been studied intensively in recent years. The genetic predisposition for non-hereditary gastric cancer is difficult to assess, as neither the real prevalence of premalignant gastric lesions in various populations nor the environmental risk factors for cancer progression are clearly defined. For non-cardiac intestinal-type cancer, identifying the factors that modulate the progression from inflammation toward cancer is crucial in order to develop preventive strategies. The role of cytokines and their gene variants has been questioned in regard to non-self-limiting H. pylori gastritis and its evolution to gastric atrophy and intestinal metaplasia; the literature now includes various and non-conclusive results on this topic. The influence of the majority of cytokine single nucleotide polymorphisms has been investigated for gastric cancer but not for preneoplastic gastric lesions. Among the investigated gene variants onlyIL10T-819C, IL-8-251, IL-18RAP917997, IL-22 rs1179251, IL1-B-511, IL1-B-3954, IL4R-398 and IL1RN were identified as predictors for premalignant gastric lesions risk. One of the most important limiting factors is the inhomogeneity of the studies (e.g., the lack of data on concomitant H. pylori infection, methods used to assess preneoplastic lesions, and source population). Testing the modifying effect of H. pylori infection upon the relationship between cytokine gene variants and premalignant gastric lesions, or even testing the interaction between H. pylori and cytokine gene variants in multivariable models adjusted for potential covariates, could increase generalizability of results.
Collapse
Affiliation(s)
- Anca Negovan
- Department of Clinical Science-Internal Medicine, University of Medicine, Pharmacy, Sciences and Technology of Târgu Mureș, Mureș 540139, Romania
| | - Mihaela Iancu
- Department of Medical Informatics and Biostatistics, “Iuliu Hațieganu” University of Medicine and Pharmacy, Cluj-Napoca, Cluj 400349, Romania
| | - Emőke Fülöp
- Department of Morphological Sciences, Histology, University of Medicine, Pharmacy, Sciences and Technology of Târgu Mureș, Mureș 540139, Romania
| | - Claudia Bănescu
- Genetics Laboratory, Center for Advanced Medical and Pharmaceutical Research, University of Medicine, Pharmacy, Sciences and Technology of Târgu Mureș, Mureș 540139, Romania
| |
Collapse
|
37
|
Ebigbo A, Messmann H, Römmele C. Endoscopic Upper GI Screening. Visc Med 2019; 35:240-244. [PMID: 31602385 DOI: 10.1159/000501889] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Accepted: 07/03/2019] [Indexed: 12/11/2022] Open
Abstract
The rationale behind endoscopic screening of the upper gastrointestinal tract (GIT) in the West is different from Asian countries with a high prevalence of gastric cancer. If epidemiological data on upper GIT cancer in the Western world is considered, then endoscopic screening of the upper GIT must target premalignant conditions as well and, furthermore, lay emphasis on the assessment of the entire upper GIT in high-risk individuals from the mouth till the duodenum. Although data on the incidence, prevalence, and further development of premalignant conditions in the upper GIT is lacking, a risk-based approach to endoscopic screening will be more cost-effective than a general screening approach. Finally, endoscopists need to be educated and trained in the assessment of the upper GIT during screening endoscopy.
Collapse
Affiliation(s)
- Alanna Ebigbo
- Department of Gastroenterology, III. Medizinische Klinik, Universitätsklinikum Augsburg, Augsburg, Germany
| | - Helmut Messmann
- Department of Gastroenterology, III. Medizinische Klinik, Universitätsklinikum Augsburg, Augsburg, Germany
| | - Christoph Römmele
- Department of Gastroenterology, III. Medizinische Klinik, Universitätsklinikum Augsburg, Augsburg, Germany
| |
Collapse
|
38
|
Rugge M, Sugano K, Scarpignato C, Sacchi D, Oblitas WJ, Naccarato AG. Gastric cancer prevention targeted on risk assessment: Gastritis OLGA staging. Helicobacter 2019; 24:e12571. [PMID: 30773732 DOI: 10.1111/hel.12571] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Revised: 01/08/2019] [Accepted: 01/10/2019] [Indexed: 12/24/2022]
Abstract
Gastric cancer (GC) ranks among the most lethal epithelial malignancies, and its striking mortality rate prompts a global prevention strategy. Helicobacter pylori (H. pylori) gastritis is the main GC promoter, and the 2014 Global Kyoto conference recognized H. pylori gastritis as a (treatable) infectious disease. It is therefore plausible that any large-scale intervention for H. pylori eradication would result in cleansing the world of the fifth cause of cancer-related death. Atrophic gastritis is the cancerization field in which GCs (both intestinal and diffuse histotypes) mainly develop. Discontinuing the inflammatory cascade triggered by H. pylori is tantamount to preventing GC. For patients (still infected or eradicated) who have already developed gastric atrophy, the severity/topography of the atrophic changes correlates with their cancer risk. Gastritis OLGA (Operative Link for Gastritis Assessment) staging consistently ranks the atrophy-associated cancer risk, providing a solid clinical/biological rationale for establishing patient-specific surveillance programs. By combining primary and secondary prevention strategies, gastric cancer is a preventable disease.
Collapse
Affiliation(s)
- Massimo Rugge
- Department of Medicine (DIMED), Surgical Pathology and Cytopathology Unit, University of Padova, Padova, Italy.,Veneto Tumor Registry (RTV), Veneto Regional Authority, Padova, Italy
| | - Kentaro Sugano
- Department of Medicine, Jichi Medical University, Tochigi, Japan
| | - Carmelo Scarpignato
- Department of Medicine and Surgery, Clinical Pharmacology and Pathophysiology Unit, School of Medicine and Dentistry, University of Parma, Parma, Italy
| | - Diana Sacchi
- Department of Medicine (DIMED), Surgical Pathology and Cytopathology Unit, University of Padova, Padova, Italy
| | | | - Antonio Giuseppe Naccarato
- Department of Translational Research and of New Surgical and Medical Technologies, Pathology Section, University of Pisa, Pisa, Italy
| |
Collapse
|
39
|
Li T, Shao W, Li S, Ma L, Zheng L, Shang W, Jia X, Sun P, Liang X, Jia J. H. pylori infection induced BMAL1 expression and rhythm disorder aggravate gastric inflammation. EBioMedicine 2019; 39:301-314. [PMID: 30502053 PMCID: PMC6354571 DOI: 10.1016/j.ebiom.2018.11.043] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2018] [Revised: 11/14/2018] [Accepted: 11/20/2018] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Rhythm abnormalities are crucial for diverse diseases. However, their role in disease progression induced by Helicobacter pylori (H. pylori) remains elusive. METHODS H. pylori infection was used in in vivo and in vitro experiments to examine its effect on rhythmic genes. The GEO database was used to screen H. pylori affecting rhythm genes, and the effect of rhythm genes on inflammatory factors. Chromatin immunoprecipitation and dual luciferase assays were used to further find out the regulation between molecules. Animal models were used to confirm the relationship between rhythm genes and H. pylori-induced inflammation. FINDINGS BMAL1 disorders aggravate inflammation induced by H. pylori. Specifically, H. pylori induce BMAL1 expression in vitro and in vivo through transcriptional activation of LIN28A, breaking the circadian rhythm. Mechanistically, LIN28A binds to the promoter region of BMAL1 and directly activates its transcription under H. pylori infection. BMAL1 in turn functions as a transcription factor and enhances the expression of proinflammatory cytokine TNF-α, thereby promoting inflammation. Of note, BMAL1 dysfunction in the rhythm disorder animal model aggravates inflammatory response induced by H. pylori infection in vivo. INTERPRETATION These findings in this study imply the pathogenic relationship between BMAL1 and H. pylori. BMAL1 may serve as a potential diagnostic marker and therapeutic target for the early diagnosis and treatment of diseases related to H. pylori infection. FUND: National Natural Science Foundation of China.
Collapse
Affiliation(s)
- Tongyu Li
- Department of Microbiology, Key Laboratory for Experimental Teratology of the Chinese Ministry of Education, School of Basic Medical Science, Shandong University, Jinan, Shandong 250012, PR China; Key Laboratory of Infection and Immunity of Shandong Province, School of Basic Medical Science, Shandong University, Jinan, Shandong 250012, PR China
| | - Wei Shao
- Department of Microbiology, Key Laboratory for Experimental Teratology of the Chinese Ministry of Education, School of Basic Medical Science, Shandong University, Jinan, Shandong 250012, PR China
| | - Shuyan Li
- Department of Microbiology, Key Laboratory for Experimental Teratology of the Chinese Ministry of Education, School of Basic Medical Science, Shandong University, Jinan, Shandong 250012, PR China
| | - Lin Ma
- Department of Microbiology, Key Laboratory for Experimental Teratology of the Chinese Ministry of Education, School of Basic Medical Science, Shandong University, Jinan, Shandong 250012, PR China
| | - Lixin Zheng
- Department of Microbiology, Key Laboratory for Experimental Teratology of the Chinese Ministry of Education, School of Basic Medical Science, Shandong University, Jinan, Shandong 250012, PR China
| | - Wenjing Shang
- Department of Microbiology, Key Laboratory for Experimental Teratology of the Chinese Ministry of Education, School of Basic Medical Science, Shandong University, Jinan, Shandong 250012, PR China
| | - Xiaxia Jia
- Department of Microbiology, Key Laboratory for Experimental Teratology of the Chinese Ministry of Education, School of Basic Medical Science, Shandong University, Jinan, Shandong 250012, PR China
| | - Pengpeng Sun
- Department of Microbiology, Key Laboratory for Experimental Teratology of the Chinese Ministry of Education, School of Basic Medical Science, Shandong University, Jinan, Shandong 250012, PR China
| | - Xiuming Liang
- Department of Microbiology, Key Laboratory for Experimental Teratology of the Chinese Ministry of Education, School of Basic Medical Science, Shandong University, Jinan, Shandong 250012, PR China; Cancer Research Laboratory, Shandong University, Karolinska Institutet collaborative Laboratory, School of Basic Medical Science, Shandong University, Jinan, Shandong 250012, PR China.
| | - Jihui Jia
- Department of Microbiology, Key Laboratory for Experimental Teratology of the Chinese Ministry of Education, School of Basic Medical Science, Shandong University, Jinan, Shandong 250012, PR China; Key Laboratory of Infection and Immunity of Shandong Province, School of Basic Medical Science, Shandong University, Jinan, Shandong 250012, PR China; Cancer Research Laboratory, Shandong University, Karolinska Institutet collaborative Laboratory, School of Basic Medical Science, Shandong University, Jinan, Shandong 250012, PR China.
| |
Collapse
|
40
|
Rugge M, Meggio A, Pravadelli C, Barbareschi M, Fassan M, Gentilini M, Zorzi M, Pretis GD, Graham DY, Genta RM. Gastritis staging in the endoscopic follow-up for the secondary prevention of gastric cancer: a 5-year prospective study of 1755 patients. Gut 2019; 68:11-17. [PMID: 29306868 DOI: 10.1136/gutjnl-2017-314600] [Citation(s) in RCA: 113] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2017] [Revised: 11/21/2017] [Accepted: 11/26/2017] [Indexed: 12/11/2022]
Abstract
OBJECTIVE Operative link on gastritis assessment (OLGA) staging for gastritis ranks the risk for gastric cancer (GC) in progressive stages (0-IV). This prospective study aimed at quantifying the cancer risk associated with each gastritis stage. DESIGN A cohort of 1755 consecutive patients with dyspepsia underwent initial (T-0) oesophagogastroduodenoscopy with mapped gastric biopsies, OLGA staging and assessment of Helicobacter pylori infection. Patients were followed for 55 months (median); patients with stages II III and IV underwent a second endoscopy/restaging (T-1), and those with stages 0 and I were followed clinically and through in-depth clinical and record checking. Endpoints were OLGA stage at T-1 and development of gastric epithelial neoplasia. RESULTS At T-0, 77.6% of patients had stage 0, 14.4% stage I, 5.1% stage II, 2.1% stage III and 0.85% stage IV. H. pylori infection was detected in 603 patients at T-0 and successfully eradicated in 602 of them; 220 had a documented history of H. pylori eradication; and 932 were H. pylori naïve-negative. Incident neoplastic lesions (prevalence=0.4%; low-grade intraepithelial neoplasia (IEN)=4; high-grade IEN=1; GC=2) developed exclusively in patients with stages III-IV. The risk for epithelial neoplasia was null in patients at stages 0, I and II (95% CI 0 to 0.4), 36.5 per 1000 person-years in patients at stage III (95% CI 13.7 to 97.4) and 63.1 per 1000 person-years in patients at stage IV (95% CI 20.3 to 195.6). CONCLUSIONS This prospective study confirms that OLGA staging reliably predicts the risk for development of gastric epithelial neoplasia. Although no neoplastic lesions arose in H. pylori-naïve patients, the H. pylori eradication in subjects with advanced stages (III-IV) did not abolish the risk for neoplastic progression.
Collapse
Affiliation(s)
- Massimo Rugge
- Department of Medicine (DIMED), Pathology Unit, University of Padua, Padova, Italy.,Veneto Tumor Registry, Veneto Region, Padova, Italy
| | - Alberto Meggio
- Department of Gastroenterology, Trento and Rovereto Hospital, Trento, Italy
| | - Cecilia Pravadelli
- Department of Gastroenterology, Trento and Rovereto Hospital, Trento, Italy
| | | | - Matteo Fassan
- Department of Medicine (DIMED), Pathology Unit, University of Padua, Padova, Italy
| | | | - Manuel Zorzi
- Veneto Tumor Registry, Veneto Region, Padova, Italy
| | - Giovanni De Pretis
- Department of Gastroenterology, Trento and Rovereto Hospital, Trento, Italy
| | - David Y Graham
- Department of Medicine, Michael E DeBakey VA Medical Center, Baylor College of Medicine, Houston, Texas, USA
| | - Robert M Genta
- Miraca Life Sciences Research Institute, Irving, Texas, USA.,Departments of Pathology and Medicine, Baylor College of Medicine, Houston, Texas, USA
| |
Collapse
|
41
|
Rugge M, Genta RM, Fassan M, Valentini E, Coati I, Guzzinati S, Savarino E, Zorzi M, Farinati F, Malfertheiner P. OLGA Gastritis Staging for the Prediction of Gastric Cancer Risk: A Long-term Follow-up Study of 7436 Patients. Am J Gastroenterol 2018; 113:1621-1628. [PMID: 30333540 DOI: 10.1038/s41395-018-0353-8] [Citation(s) in RCA: 83] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Accepted: 07/20/2018] [Indexed: 12/11/2022]
Abstract
OBJECTIVES Gastritis OLGA-staging ranks the risk for gastric cancer (GC) in progressive stages (0-IV). This long-term follow-up study quantifies the GC risk associated with each OLGA stage. METHODS Consecutive patients (7436) underwent esophagogastroscopy (T-0), with mapped gastric biopsies, OLGA staging, and H. pylori status assessment. Patients with neoplastic lesion (invasive or non-invasive) at the index endoscopy (and/or within 12 months) were excluded. All patients were followed-up (T-1) by combining different sources of clinical/pathological information (Regional Registries of: (i) esophagogastroduodenoscopies; (ii) pathology reports; (iii) cancer, (iv) mortality). The endpoint was histologically documented development of gastric epithelial neoplasia. RESULTS At T-0, the patients' distribution by OLGA stage was: Stage 0 = 80.8%; Stage I = 12.6%; Stage II = 4.3%; Stage III = 2.0%; Stage IV = 0.3%; H. pylori infection was detected in 25.9% of patients. At the end of the follow-up (mean/median = 6.3/6.6 years), 28 incident neoplasia were documented (overall prevalence = 0.60 per 103/person-years; low-grade intraepithelial neoplasia = 17/28; high-grade intraepithelial neoplasia = 4/28; GC = 7/28). By OLGA stage at the enrollment, the rate of incident neoplasia was: Stage 0 = 1 case; rate/103 person-years = 0.03; 95%CI: 0.004-0.19; Stage I = 2 cases; rate/103 person-years = 0.34; 95%CI: 0.09-1.36; Stage II = 3 cases; rate/103 person-years = 1.48; 95%CI: 0.48-4.58; Stage III = 17 cases; rate/103 person-years = 19.1; 95%CI: 11.9-30.7; Stage IV = 5 cases; rate/103 person-years = 41.2; 95%CI: 17.2-99.3. Multivariate analysis including gender, age, H. pylori status, and OLGA stage at enrollment only disclosed OLGA stage as predictor of neoplastic progression (OLGA stage III: HR = 712.4, 95%CI = 92.543-5484.5; OLGA stage IV: HR = 1450.7, 95%CI = 166.7-12626.0). CONCLUSIONS Among 7436 patients, OLGA stages at the enrollment correlated significantly with different risk for gastric neoplasia. Based on the obtained results, gastritis staging is a critical adjunct in endoscopy follow-up protocols aimed at GC secondary prevention.
Collapse
Affiliation(s)
- Massimo Rugge
- Department of Medicine (DIMED), Surgical pathology & Cytopathology Unit, University of Padova-Azienda Ospedaliera di Padova, Padova, Italy. Veneto Tumor Registry, Veneto Region, Padova, Italy. Departments of Pathology and Medicine, Baylor College of Medicine, Houston, TX, USA. Inform Diagnostics Research Institute, Irving, TX, USA. Department of Oncology and Gastroenterology (DISGOG), Gastroenterology Unit, University of Padova-Azienda Ospedaliera di Padova, Padova, Italy. Department of Gastroenterology, Hepatology and Infectious Diseases, Otto-von-Guericke University Hospital, Magdeburg, Germany.,Department of Medicine (DIMED), Surgical pathology & Cytopathology Unit, University of Padova-Azienda Ospedaliera di Padova, Padova, Italy. Veneto Tumor Registry, Veneto Region, Padova, Italy. Departments of Pathology and Medicine, Baylor College of Medicine, Houston, TX, USA. Inform Diagnostics Research Institute, Irving, TX, USA. Department of Oncology and Gastroenterology (DISGOG), Gastroenterology Unit, University of Padova-Azienda Ospedaliera di Padova, Padova, Italy. Department of Gastroenterology, Hepatology and Infectious Diseases, Otto-von-Guericke University Hospital, Magdeburg, Germany
| | - Robert M Genta
- Department of Medicine (DIMED), Surgical pathology & Cytopathology Unit, University of Padova-Azienda Ospedaliera di Padova, Padova, Italy. Veneto Tumor Registry, Veneto Region, Padova, Italy. Departments of Pathology and Medicine, Baylor College of Medicine, Houston, TX, USA. Inform Diagnostics Research Institute, Irving, TX, USA. Department of Oncology and Gastroenterology (DISGOG), Gastroenterology Unit, University of Padova-Azienda Ospedaliera di Padova, Padova, Italy. Department of Gastroenterology, Hepatology and Infectious Diseases, Otto-von-Guericke University Hospital, Magdeburg, Germany.,Department of Medicine (DIMED), Surgical pathology & Cytopathology Unit, University of Padova-Azienda Ospedaliera di Padova, Padova, Italy. Veneto Tumor Registry, Veneto Region, Padova, Italy. Departments of Pathology and Medicine, Baylor College of Medicine, Houston, TX, USA. Inform Diagnostics Research Institute, Irving, TX, USA. Department of Oncology and Gastroenterology (DISGOG), Gastroenterology Unit, University of Padova-Azienda Ospedaliera di Padova, Padova, Italy. Department of Gastroenterology, Hepatology and Infectious Diseases, Otto-von-Guericke University Hospital, Magdeburg, Germany
| | - Matteo Fassan
- Department of Medicine (DIMED), Surgical pathology & Cytopathology Unit, University of Padova-Azienda Ospedaliera di Padova, Padova, Italy. Veneto Tumor Registry, Veneto Region, Padova, Italy. Departments of Pathology and Medicine, Baylor College of Medicine, Houston, TX, USA. Inform Diagnostics Research Institute, Irving, TX, USA. Department of Oncology and Gastroenterology (DISGOG), Gastroenterology Unit, University of Padova-Azienda Ospedaliera di Padova, Padova, Italy. Department of Gastroenterology, Hepatology and Infectious Diseases, Otto-von-Guericke University Hospital, Magdeburg, Germany
| | - Elisa Valentini
- Department of Medicine (DIMED), Surgical pathology & Cytopathology Unit, University of Padova-Azienda Ospedaliera di Padova, Padova, Italy. Veneto Tumor Registry, Veneto Region, Padova, Italy. Departments of Pathology and Medicine, Baylor College of Medicine, Houston, TX, USA. Inform Diagnostics Research Institute, Irving, TX, USA. Department of Oncology and Gastroenterology (DISGOG), Gastroenterology Unit, University of Padova-Azienda Ospedaliera di Padova, Padova, Italy. Department of Gastroenterology, Hepatology and Infectious Diseases, Otto-von-Guericke University Hospital, Magdeburg, Germany
| | - Irene Coati
- Department of Medicine (DIMED), Surgical pathology & Cytopathology Unit, University of Padova-Azienda Ospedaliera di Padova, Padova, Italy. Veneto Tumor Registry, Veneto Region, Padova, Italy. Departments of Pathology and Medicine, Baylor College of Medicine, Houston, TX, USA. Inform Diagnostics Research Institute, Irving, TX, USA. Department of Oncology and Gastroenterology (DISGOG), Gastroenterology Unit, University of Padova-Azienda Ospedaliera di Padova, Padova, Italy. Department of Gastroenterology, Hepatology and Infectious Diseases, Otto-von-Guericke University Hospital, Magdeburg, Germany
| | - Stefano Guzzinati
- Department of Medicine (DIMED), Surgical pathology & Cytopathology Unit, University of Padova-Azienda Ospedaliera di Padova, Padova, Italy. Veneto Tumor Registry, Veneto Region, Padova, Italy. Departments of Pathology and Medicine, Baylor College of Medicine, Houston, TX, USA. Inform Diagnostics Research Institute, Irving, TX, USA. Department of Oncology and Gastroenterology (DISGOG), Gastroenterology Unit, University of Padova-Azienda Ospedaliera di Padova, Padova, Italy. Department of Gastroenterology, Hepatology and Infectious Diseases, Otto-von-Guericke University Hospital, Magdeburg, Germany
| | - Edoardo Savarino
- Department of Medicine (DIMED), Surgical pathology & Cytopathology Unit, University of Padova-Azienda Ospedaliera di Padova, Padova, Italy. Veneto Tumor Registry, Veneto Region, Padova, Italy. Departments of Pathology and Medicine, Baylor College of Medicine, Houston, TX, USA. Inform Diagnostics Research Institute, Irving, TX, USA. Department of Oncology and Gastroenterology (DISGOG), Gastroenterology Unit, University of Padova-Azienda Ospedaliera di Padova, Padova, Italy. Department of Gastroenterology, Hepatology and Infectious Diseases, Otto-von-Guericke University Hospital, Magdeburg, Germany
| | - Manuel Zorzi
- Department of Medicine (DIMED), Surgical pathology & Cytopathology Unit, University of Padova-Azienda Ospedaliera di Padova, Padova, Italy. Veneto Tumor Registry, Veneto Region, Padova, Italy. Departments of Pathology and Medicine, Baylor College of Medicine, Houston, TX, USA. Inform Diagnostics Research Institute, Irving, TX, USA. Department of Oncology and Gastroenterology (DISGOG), Gastroenterology Unit, University of Padova-Azienda Ospedaliera di Padova, Padova, Italy. Department of Gastroenterology, Hepatology and Infectious Diseases, Otto-von-Guericke University Hospital, Magdeburg, Germany
| | - Fabio Farinati
- Department of Medicine (DIMED), Surgical pathology & Cytopathology Unit, University of Padova-Azienda Ospedaliera di Padova, Padova, Italy. Veneto Tumor Registry, Veneto Region, Padova, Italy. Departments of Pathology and Medicine, Baylor College of Medicine, Houston, TX, USA. Inform Diagnostics Research Institute, Irving, TX, USA. Department of Oncology and Gastroenterology (DISGOG), Gastroenterology Unit, University of Padova-Azienda Ospedaliera di Padova, Padova, Italy. Department of Gastroenterology, Hepatology and Infectious Diseases, Otto-von-Guericke University Hospital, Magdeburg, Germany
| | - Peter Malfertheiner
- Department of Medicine (DIMED), Surgical pathology & Cytopathology Unit, University of Padova-Azienda Ospedaliera di Padova, Padova, Italy. Veneto Tumor Registry, Veneto Region, Padova, Italy. Departments of Pathology and Medicine, Baylor College of Medicine, Houston, TX, USA. Inform Diagnostics Research Institute, Irving, TX, USA. Department of Oncology and Gastroenterology (DISGOG), Gastroenterology Unit, University of Padova-Azienda Ospedaliera di Padova, Padova, Italy. Department of Gastroenterology, Hepatology and Infectious Diseases, Otto-von-Guericke University Hospital, Magdeburg, Germany
| |
Collapse
|
42
|
Lahner E, Carabotti M, Esposito G, Hassan C, Zullo A, Annibale B. Occurrence and predictors of metaplastic atrophic gastritis in a nation-wide consecutive endoscopic population presenting with upper gastrointestinal symptoms. Eur J Gastroenterol Hepatol 2018; 30:1291-1296. [PMID: 30161028 DOI: 10.1097/meg.0000000000001246] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
OBJECTIVES The frequency of metaplastic atrophic gastritis in noninvestigated dyspepsia and its associated symptoms pattern are not known. This study aimed to assess the occurrence and clinical predictors of metaplastic atrophic gastritis in a consecutive endoscopic population presenting with upper gastrointestinal symptoms. PATIENTS AND METHODS A post-hoc cross-sectional study was carried out on endoscopic-histological data from 668 patients (64% women, age: 58 years) with upper gastrointestinal symptoms obtained during a multicenter study. Patients with dyspeptic (46%), reflux-like symptoms (22%), and both (32%) as indications for gastroscopy were included. Gastroscopy with biopsies (Sydney System) was performed. Clinical data and symptoms were collected using a structured questionnaire. RESULTS Metaplastic atrophic gastritis was detected in 201 (30.1%) patients: 22.3% antrum-restricted, 4.5% corpus-restricted, and 3.3% extensive. Postprandial fullness was more frequent in patients with metaplastic atrophic gastritis compared with those without metaplastic atrophic gastritis (29.8 vs. 18.8%, P=0.002); epigastric pain/burning, early satiety, and reflux symptoms occurred similarly in both groups. At multivariate logistic regression, metaplastic atrophic gastritis (any site) was associated with age older than 55 years [odds ratio (OR): 1.86, 95% confidence interval (CI): 1.26-2.73], Helicobacter pylori (OR: 1.66, 95% CI: 1.1-2.39), postprandial fullness (OR: 1.62, 95% CI: 1.03-2.54), and smoking (OR: 1.53, 95% CI: 1.02-2.31). Corpus-restricted atrophic gastritis was associated with postprandial fullness (OR: 4.16, 95% CI: 1.79-9.63) and NSAIDs (OR: 2.98, 95% CI: 1.01-8.88), extensive and antrum-restricted atrophic gastritis was associated with age older than 55 (OR 3.6, 95% CI: 1.20-12.67 and OR: 1.63, 95% CI: 1.06-2.49). CONCLUSION Metaplastic atrophic gastritis, a preneoplastic condition, may be found in about one of three patients undergoing endoscopy for upper gastrointestinal symptoms, and clinical predictors are age older than 55 years, smoking, H. pylori infection, and postprandial fullness, especially for corpus-restricted metaplastic atrophic gastritis. An accurate pre-endoscopic characterization of patients with upper gastrointestinal symptoms with easily achievable clinical information may help to better address an endoscopic investigation.
Collapse
Affiliation(s)
- Edith Lahner
- Medical-Surgical Department of Clinical Sciences and Translational Medicine, Sant'Andrea Hospital, University Sapienza
| | - Marilia Carabotti
- Medical-Surgical Department of Clinical Sciences and Translational Medicine, Sant'Andrea Hospital, University Sapienza
| | - Gianluca Esposito
- Medical-Surgical Department of Clinical Sciences and Translational Medicine, Sant'Andrea Hospital, University Sapienza
| | - Cesare Hassan
- Department of Gastroenterology and Digestive Endoscopy, 'Nuovo Regina Margherita' Hospital, Rome, Italy
| | - Angelo Zullo
- Department of Gastroenterology and Digestive Endoscopy, 'Nuovo Regina Margherita' Hospital, Rome, Italy
| | - Bruno Annibale
- Medical-Surgical Department of Clinical Sciences and Translational Medicine, Sant'Andrea Hospital, University Sapienza
| |
Collapse
|
43
|
Wan JJ, Fei SJ, Lv SX, Han ST, Ma XG, Xu DS, Chen HM, Zhang J. Role of gastroscopic biopsy of gastric ulcer margins and healed sites in the diagnosis of early gastric cancer: A clinical controlled study of 513 cases. Oncol Lett 2018; 16:4211-4218. [PMID: 30250534 PMCID: PMC6144944 DOI: 10.3892/ol.2018.9156] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2016] [Accepted: 05/04/2017] [Indexed: 01/09/2023] Open
Abstract
The present case study aimed to evaluate the effect of gastroscopic biopsy of gastric ulcer margins and healed sites in the diagnosis of early gastric cancer. A total of 513 patients who were diagnosed with gastric ulcers using gastroscopy between January 1999 and December 2013 were included in the present study and were divided into either the experimental or the control group. In the control group, samples were only taken from the ulcer margin, whereas in the experimental group samples were taken from the ulcer margin and from the ulcer base. In the experimental group, a routine biopsy of the ulcer margin was performed on first examination, and recheck by gastroscopy was performed from the second week. For ulcers that remained unhealed, a biopsy of the ulcer margin was subsequently conducted; however, for healed or almost healed ulcers, a biopsy of the ulcer base was conducted. The duration of follow-up by gastroscopy ranged between 1 week and 24 months. For the control group, a biopsy of the ulcer margin was conducted using the conventional method. The detection rate of the experimental group was 3.88% (9/232), with 4 cases of gastric cancer confirmed from examinations of the ulcer base. The detection rate of the control group was 1.07% (3/281), which was significantly decreased compared with that of the experimental group (P=0.0345). Overall, patients who underwent regular follow-up gastroscopy following treatment exhibited a markedly increased detection rate of early gastric cancer, suggesting that early cancer may occur in healed gastric ulcer sites.
Collapse
Affiliation(s)
- Jing-Jing Wan
- Digestive Endoscopy Center, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210023, P.R. China.,Department of Gastroenterology, Huaian Hospital Affiliated to Xuzhou Medical University, Huai'an, Jiangsu 223002, P.R. China
| | - Su-Juan Fei
- Department of Gastroenterology, Xuzhou Medical University Affiliated Hospital, Xuzhou, Jiangsu 221004, P.R. China
| | - Sheng-Xiang Lv
- Department of Gastroenterology, Huaian Hospital Affiliated to Xuzhou Medical University, Huai'an, Jiangsu 223002, P.R. China
| | - Shu-Tang Han
- Digestive Endoscopy Center, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210023, P.R. China
| | - Xing-Gang Ma
- Department of Gastroenterology, Huaian Hospital Affiliated to Xuzhou Medical University, Huai'an, Jiangsu 223002, P.R. China
| | - Dong-Sheng Xu
- Department of Gastroenterology, Huaian Hospital Affiliated to Xuzhou Medical University, Huai'an, Jiangsu 223002, P.R. China
| | - Hong-Mei Chen
- Department of Gastroenterology, Huaian Hospital Affiliated to Xuzhou Medical University, Huai'an, Jiangsu 223002, P.R. China
| | - Ju Zhang
- Department of Gastroenterology, Huaian Hospital Affiliated to Xuzhou Medical University, Huai'an, Jiangsu 223002, P.R. China
| |
Collapse
|
44
|
Lahner E, Carabotti M, Annibale B. Treatment of Helicobacter pylori infection in atrophic gastritis. World J Gastroenterol 2018; 24:2373-2380. [PMID: 29904244 PMCID: PMC6000293 DOI: 10.3748/wjg.v24.i22.2373] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2018] [Revised: 05/16/2018] [Accepted: 05/28/2018] [Indexed: 02/06/2023] Open
Abstract
Helicobacter pylori (Hp) is a major human pathogen causing chronic, progressive gastric mucosal damage and is linked to gastric atrophy and cancer. Hp-positive individuals constitute the major reservoir for transmission of infection. There is no ideal treatment for Hp. Hp infection is not cured by a single antibiotic, and sometimes, a combined treatment with three or more antibiotics is ineffective. Atrophic gastritis (AG) is a chronic disease whose main features are atrophy and/or intestinal metaplasia of the gastric glands, which arise from long-standing Hp infection. AG is reportedly linked to an increased risk for gastric cancer, particularly when extensive intestinal metaplasia is present. Active or past Hp infection may be detected by conventional methods in about two-thirds of AG patients. By immunoblotting of sera against Hp whole-cell protein lysates, a previous exposure to Hp infection is detected in all AG patients. According to guidelines, AG patients with Hp positivity should receive eradication treatment. The goals of treatment are as follows: (1) Cure of infection, resolution of inflammation and normalization of gastric functions; (2) possible reversal of atrophic and metaplastic changes of the gastric mucosa; and (3) prevention of gastric cancer. An ideal antibiotic regimen for Hp should achieve eradication rates of approximately 90%, and complex multidrug regimens are required to reach this goal. Amongst the factors associated with treatment failure are high bacterial load, high gastric acidity, Hp strain, smoking, low compliance, overweight, and increasing antibiotic resistance. AG, when involving the corporal mucosa, is linked to reduced gastric acid secretion. At a non-acidic intra-gastric pH, the efficacy of the common treatment regimens combining proton pump inhibitors with one or more antibiotics may not be the same as that observed in patients with Hp gastritis in an acid-producing stomach. Although the efficacy of these therapeutic regimens has been thoroughly tested in subjects with Hp infection, there is a paucity of evidence in the subgroup of patients with AG. Bismuth-based therapy may be an attractive treatment in the specific setting of AG, and specific studies on the efficacy of bismuth-based therapies are needed in patients with AG.
Collapse
Affiliation(s)
- Edith Lahner
- Medical-Surgical Department of Clinical Sciences and Translational Medicine, Sant’Andrea Hospital, University Sapienza, Rome 00189, Italy
| | - Marilia Carabotti
- Medical-Surgical Department of Clinical Sciences and Translational Medicine, Sant’Andrea Hospital, University Sapienza, Rome 00189, Italy
| | - Bruno Annibale
- Medical-Surgical Department of Clinical Sciences and Translational Medicine, Sant’Andrea Hospital, University Sapienza, Rome 00189, Italy
| |
Collapse
|
45
|
Cancer-associated fibroblasts in tumor microenvironment - Accomplices in tumor malignancy. Cell Immunol 2018; 343:103729. [PMID: 29397066 DOI: 10.1016/j.cellimm.2017.12.003] [Citation(s) in RCA: 200] [Impact Index Per Article: 28.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2017] [Revised: 11/15/2017] [Accepted: 12/04/2017] [Indexed: 12/12/2022]
Abstract
There is much cellular heterogeneity in the tumor microenvironment. The tumor epithelia and stromal cells co-evolve, and this reciprocal relationship dictates almost every step of cancer development and progression. Despite this, many anticancer therapies are designed around druggable features of tumor epithelia, ignoring the supportive role of stromal cells. Cancer-associated fibroblasts (CAFs) are the dominant cell type within the reactive stroma of many tumor types. Numerous previous studies have highlighted a pro-tumorigenic role for CAFs via secretion of various growth factors, cytokines, chemokines, and the degradation of extracellular matrix. Recent works showed that CAFs secrete H2O2 to effect stromal-mediated field cancerization, transform primary epithelial cells, and aggravate cancer cell aggressiveness, in addition to inflammatory and mitogenic factors. Molecular characterization of CAFs also underscores the importance of Notch and specific nuclear receptor signaling in the activation of CAFs. This review consolidates recent findings of CAFs and highlights areas for future investigations.
Collapse
|
46
|
Abstract
The elective background for gastric adenocarcinoma is the atrophic transformation of the gastric mucosa. The extent of mucosal atrophy basically parallels the risk of developing gastric cancer. This means that either noninvasive (serology) or invasive (endoscopy/histology) methods enabling the atrophic transformation to be quantified can be used theoretically to assess a given patient's gastric cancer risk. This review aims to focus on the reliability of histology gastritis Operative Link for Gastritis Assessment -staging system for assessing the 'personalized' cancer risk in individuals with (atrophic) gastritis.
Collapse
|
47
|
Baker KT, Salk JJ, Brentnall TA, Risques RA. Precancer in ulcerative colitis: the role of the field effect and its clinical implications. Carcinogenesis 2018; 39:11-20. [PMID: 29087436 PMCID: PMC6248676 DOI: 10.1093/carcin/bgx117] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Revised: 09/22/2017] [Accepted: 10/26/2017] [Indexed: 12/13/2022] Open
Abstract
Cumulative evidence indicates that a significant proportion of cancer evolution may occur before the development of histological abnormalities. While recent improvements in DNA sequencing technology have begun to reveal the presence of these early preneoplastic clones, the concept of 'premalignant field' was already introduced by Slaughter more than half a century ago. Also referred to as 'field effect', 'field defect' or 'field cancerization', these terms describe the phenomenon by which molecular alterations develop in normal-appearing tissue and expand to form premalignant patches with the potential to progress to dysplasia and cancer. Field effects have been well-characterized in ulcerative colitis, an inflammatory bowel disease that increases the risk of colorectal cancer. The study of the molecular alterations that define these fields is informative of mechanisms of tumor initiation and progression and has provided potential targets for early cancer detection. Herein, we summarize the current knowledge about the molecular alterations that comprise the field effect in ulcerative colitis and the clinical utility of these fields for cancer screening and prevention.
Collapse
Affiliation(s)
- Kathryn T Baker
- Department of Pathology, University of Washington, Seattle, WA, USA
| | - Jesse J Salk
- Division of Hematology and Oncology, Department of Medicine, University of
Washington, Seattle, WA, USA
- TwinStrand Biosciences Seattle, WA, USA
| | - Teresa A Brentnall
- Division of Gasteroenterology, Department of Medicine, University of
Washington, Seattle, WA, USA
| | - Rosa Ana Risques
- To whom correspondence should be addressed. Tel: +206-616-4976; Fax:
+206-543-1140;
| |
Collapse
|
48
|
Peppelenbosch MP. Preface to "Gut microbiome in health and disease". Best Pract Res Clin Gastroenterol 2017; 31:605-606. [PMID: 29566902 DOI: 10.1016/j.bpg.2017.10.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2017] [Accepted: 10/29/2017] [Indexed: 01/31/2023]
Affiliation(s)
- Maikel P Peppelenbosch
- Department of Gastroenterology & Hepatology, Erasmus MC, University Medical Center Rotterdam, PO Box 2040, NL-3000 CA Rotterdam, The Netherlands.
| |
Collapse
|
49
|
Rugge M, Genta RM, Di Mario F, El-Omar EM, El-Serag HB, Fassan M, Hunt RH, Kuipers EJ, Malfertheiner P, Sugano K, Graham DY. Gastric Cancer as Preventable Disease. Clin Gastroenterol Hepatol 2017; 15:1833-1843. [PMID: 28532700 DOI: 10.1016/j.cgh.2017.05.023] [Citation(s) in RCA: 135] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2017] [Revised: 04/26/2017] [Accepted: 05/16/2017] [Indexed: 02/07/2023]
Abstract
Gastric cancer, 1 of the 5 most common causes of cancer death, is associated with a 5-year overall survival rate less than 30%. A minority of cancers occurs as part of syndromic diseases; more than 90% of adenocarcinomas are considered as the ultimate consequence of a longstanding mucosal inflammation. Helicobacter pylori infection is the leading etiology of non-self-limiting gastritis, which may result in atrophy of the gastric mucosa and impaired acid secretion. Gastric atrophy establishes a field of cancerization prone to further molecular and phenotypic changes, possibly resulting in cancer growth. This well-understood natural history provides the clinicopathologic rationale for primary and secondary cancer prevention strategies. A large body of evidence demonstrates that combined primary (H pylori eradication) and secondary (mainly endoscopy) prevention efforts may prevent or limit the progression of gastric oncogenesis. This approach, which is tailored to different country-specific gastric cancer incidence, socioeconomic, and cultural factors, requires that the complementary competences of gastroenterologists, oncologists, and pathologists be amalgamated into a common strategy of health policy.
Collapse
Affiliation(s)
- Massimo Rugge
- Department of Medicine (DIMED), University of Padua, Padua, Italy; Veneto Tumor Registry, Veneto Region, Padua, Italy.
| | - Robert M Genta
- Miraca Life Sciences Research Institute, Irving, and Departments of Pathology and Medicine, Baylor College of Medicine, Houston, Texas
| | - Francesco Di Mario
- Department of Clinical and Experimental Medicine, University of Parma, Parma, Italy
| | - Emad M El-Omar
- St George and Sutherland Clinical School, University of New South Wales, Sydney, Australia
| | - Hashem B El-Serag
- Department of Medicine, Michael E. DeBakey VA Medical Center, Baylor College of Medicine, Houston, Texas
| | - Matteo Fassan
- Department of Medicine (DIMED), University of Padua, Padua, Italy
| | - Richard H Hunt
- Division of Gastroenterology, Department of Medicine and Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, Canada
| | - Ernst J Kuipers
- Department of Gastroenterology and Hepatology, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | | | - Kentaro Sugano
- Department of Medicine, Jichi Medical University, Tochigi, Japan
| | - David Y Graham
- Department of Medicine, Michael E. DeBakey VA Medical Center, Baylor College of Medicine, Houston, Texas
| |
Collapse
|
50
|
Tong Y, Wu Y, Song Z, Yu Y, Yu X. The potential value of serum pepsinogen for the diagnosis of atrophic gastritis among the health check-up populations in China: a diagnostic clinical research. BMC Gastroenterol 2017; 17:88. [PMID: 28728545 PMCID: PMC5520218 DOI: 10.1186/s12876-017-0641-6] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2016] [Accepted: 06/29/2017] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND The aim of this study is to assess the validity of the measurement of pepsinogen as a screening test for chronic atrophic gastritis (AG) in health check-up populations in China. METHODS Patients from consecutive regular health check-up were enrolled from January 2014 to June 2015. Endoscopy, combined with monitoring the Helicobacter pylori (Hp) infections, and measuring the serum pepsinogen (PG) were used to determine the diagnostic accuracy of PG for the screening of atrophic gastritis. Histopathology was assessed by the Operative Link on Gastritis Assessment (OLGA) system. Statistical analysis was performed using SPSS statistical software. RESULTS The total Hp infection rate was 40%. Based on pathology, the 996 participants were divided into three groups: non-atrophic (NAG), mild-moderate atrophic (MAG): stage I and II of the OLGA classification, and severe atrophic (SAG): stage III and IV of the OLGA classification. Compared with NAG and MAG groups, PGR decreased significantly in SAG group (p < 0.05). PGI and PGII levels were significantly elevated in Hp-positive group, while the PGR was markedly decreased (p < 0.01). When MAG and SAG groups were combined and compared with NAG group, the best cutoff value for atrophy diagnosis was PGI ≤50.3 ng/ml; the cutoff value in Hp-negative group was absolutely higher than in Hp-positive group. When NAG and MAG groups were combined and compared with the SAG group, the best cutoff value for diagnosis of severe atrophy was at PGR ≤4.28. The cutoff values in Hp-negative and Hp-positive groups were calculated at PGR ≤6.28 and ≤4.28, respectively. CONCLUSIONS Pepsinogens play an important role in the identification of patients with atrophic gastritis and severe AG. Use of different cutoff values of PG for Hp-negative and Hp-positive groups may offer greater efficacy in the diagnosis of AG.
Collapse
Affiliation(s)
- Yuling Tong
- International HealthCare Center, The Second Affiliated Hospital of Zhejiang University, School of Medicine, NO.88 Jiefang Road, Hangzhou, Zhejiang Province, People's Republic of China
| | - Yulian Wu
- General Surgery Department, The Second Affiliated Hospital of Zhejiang University, School of Medicine, NO.88 Jiefang Road, Hangzhou, Zhejiang Province, People's Republic of China.
| | - Zhenya Song
- International HealthCare Center, The Second Affiliated Hospital of Zhejiang University, School of Medicine, NO.88 Jiefang Road, Hangzhou, Zhejiang Province, People's Republic of China.
| | - Yingying Yu
- International HealthCare Center, The Second Affiliated Hospital of Zhejiang University, School of Medicine, NO.88 Jiefang Road, Hangzhou, Zhejiang Province, People's Republic of China
| | - Xinyan Yu
- International HealthCare Center, The Second Affiliated Hospital of Zhejiang University, School of Medicine, NO.88 Jiefang Road, Hangzhou, Zhejiang Province, People's Republic of China
| |
Collapse
|