51
|
Lee JWJ, Zhu F, Srivastava S, Tsao SKK, Khor C, Ho KY, Fock KM, Lim WC, Ang TL, Chow WC, So JBY, Koh CJ, Chua SJ, Wong ASY, Rao J, Lim LG, Ling KL, Chia CK, Ooi CJ, Rajnakova A, Yap WM, Salto-Tellez M, Ho B, Soong R, Chia KS, Teo YY, Teh M, Yeoh KG. Severity of gastric intestinal metaplasia predicts the risk of gastric cancer: a prospective multicentre cohort study (GCEP). Gut 2022; 71:854-863. [PMID: 33975867 PMCID: PMC8995828 DOI: 10.1136/gutjnl-2021-324057] [Citation(s) in RCA: 54] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Revised: 04/15/2021] [Accepted: 04/30/2021] [Indexed: 12/24/2022]
Abstract
OBJECTIVE To investigate the incidence of gastric cancer (GC) attributed to gastric intestinal metaplasia (IM), and validate the Operative Link on Gastric Intestinal Metaplasia (OLGIM) for targeted endoscopic surveillance in regions with low-intermediate incidence of GC. METHODS A prospective, longitudinal and multicentre study was carried out in Singapore. The study participants comprised 2980 patients undergoing screening gastroscopy with standardised gastric mucosal sampling, from January 2004 and December 2010, with scheduled surveillance endoscopies at year 3 and 5. Participants were also matched against the National Registry of Diseases Office for missed diagnoses of early gastric neoplasia (EGN). RESULTS There were 21 participants diagnosed with EGN. IM was a significant risk factor for EGN (adjusted-HR 5.36; 95% CI 1.51 to 19.0; p<0.01). The age-adjusted EGN incidence rates for patients with and without IM were 133.9 and 12.5 per 100 000 person-years. Participants with OLGIM stages III-IV were at greatest risk (adjusted-HR 20.7; 95% CI 5.04 to 85.6; p<0.01). More than half of the EGNs (n=4/7) attributed to baseline OLGIM III-IV developed within 2 years (range: 12.7-44.8 months). Serum trefoil factor 3 distinguishes (Area Under the Receiver Operating Characteristics 0.749) patients with OLGIM III-IV if they are negative for H. pylori. Participants with OLGIM II were also at significant risk of EGN (adjusted-HR 7.34; 95% CI 1.60 to 33.7; p=0.02). A significant smoking history further increases the risk of EGN among patients with OLGIM stages II-IV. CONCLUSIONS We suggest a risk-stratified approach and recommend that high-risk patients (OLGIM III-IV) have endoscopic surveillance in 2 years, intermediate-risk patients (OLGIM II) in 5 years.
Collapse
Affiliation(s)
- Jonathan W J Lee
- Division of Gastroenterology and Hepatology, National University Hospital, Singapore,Department of Medicine, National University of Singapore, Singapore,Singapore Gastric Cancer Consortium, Singapore
| | - Feng Zhu
- Department of Medicine, National University of Singapore, Singapore,Singapore Gastric Cancer Consortium, Singapore
| | | | - Stephen KK Tsao
- Department of Gastroenterology & Hepatology, Tan Tock Seng Hospital, Singapore
| | - Christopher Khor
- Department of Gastroenterology & Hepatology, Singapore General Hospital, Singapore
| | - Khek Yu Ho
- Division of Gastroenterology and Hepatology, National University Hospital, Singapore,Department of Medicine, National University of Singapore, Singapore
| | - Kwong Ming Fock
- Department of Gastroenterology & Hepatology, Changi General Hospital, Singapore
| | - Wee Chian Lim
- Department of Gastroenterology & Hepatology, Tan Tock Seng Hospital, Singapore
| | - Tiing Leong Ang
- Department of Gastroenterology & Hepatology, Changi General Hospital, Singapore
| | - Wan Cheng Chow
- Department of Gastroenterology & Hepatology, Singapore General Hospital, Singapore
| | - Jimmy Bok Yan So
- Singapore Gastric Cancer Consortium, Singapore,Department of Surgery, National University of Singapore, Singapore
| | - Calvin J Koh
- Division of Gastroenterology and Hepatology, National University Hospital, Singapore,Department of Medicine, National University of Singapore, Singapore,Singapore Gastric Cancer Consortium, Singapore
| | - Shijia Joy Chua
- Department of Medicine, National University of Singapore, Singapore
| | | | - Jaideepraj Rao
- Department of Surgery, Tan Tock Seng Hospital, Singapore
| | | | | | | | | | - Andrea Rajnakova
- Andrea's Digestive, Colon, Liver and Gallbladder Clinic Pte Ltd, Singapore
| | - Wai Ming Yap
- Department of Pathology, Tan Tock Seng Hospital, Singapore
| | - Manuel Salto-Tellez
- Precision Medicine Centre of Excellence, Queen's University Belfast, Belfast, UK,Integrated Pathology Unit, Institute of Cancer Research, London, UK
| | - Bow Ho
- Department of Microbiology, National University of Singapore, Singapore
| | - Richie Soong
- Cancer Science Institute of Singapore, National University of Singapore, Singapore,Department of Pathology, National University of Singapore, Singapore,Pascific Laboratories, Singapore
| | - Kee Seng Chia
- Saw Swee Hock School of Public Health, National University of Singapore, Singapore
| | - Yik Ying Teo
- Saw Swee Hock School of Public Health, National University of Singapore, Singapore
| | - Ming Teh
- Singapore Gastric Cancer Consortium, Singapore,Department of Pathology, National University of Singapore, Singapore
| | - Khay-Guan Yeoh
- Division of Gastroenterology and Hepatology, National University Hospital, Singapore .,Department of Medicine, National University of Singapore, Singapore.,Singapore Gastric Cancer Consortium, Singapore
| |
Collapse
|
52
|
Serum Pepsinogens Combined with New Biomarkers Testing Using Chemiluminescent Enzyme Immunoassay for Non-Invasive Diagnosis of Atrophic Gastritis: A Prospective, Multicenter Study. Diagnostics (Basel) 2022; 12:diagnostics12030695. [PMID: 35328248 PMCID: PMC8947400 DOI: 10.3390/diagnostics12030695] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 03/05/2022] [Accepted: 03/05/2022] [Indexed: 12/24/2022] Open
Abstract
Background: Analysis of serum biomarkers for the assessment of atrophic gastritis (AG), a gastric precancerous lesion, is of growing interest for identification of patients at increased risk of gastric cancer. The aim was to analyze the diagnostic performance of serum pepsinogen testing using another method, chemiluminescent enzyme immunoassay (CLEIA), as well as of other new potential biomarkers. Material and Methods: The sera of patients considered at increased risk of gastric cancer and undergoing upper endoscopy collected in our previous prospective, multicenter study were tested for pepsinogen I (PGI) and II (PGII), interleukin-6 (IL-6), human epididymal protein 4 (HE-4), adiponectin, ferritin and Krebs von den Lungen (KL-6) using the CLEIA. The diagnostic performance for the detection of AG was calculated by taking histology as the reference. Results: In total, 356 patients (162 men (46%); mean age 58.6 (±14.2) years), including 152 with AG, were included. For the detection of moderate to severe corpus AG, sensitivity and specificity of the pepsinogen I/II ratio were of 75.0% (95%CI 57.8–87.9) and 92.6% (88.2–95.8), respectively. For the detection of moderate to severe antrum AG, sensitivity of IL-6 was of 72.2% (95%CI 46.5–90.3). Combination of pepsinogen I/II ratio or HE-4 showed a sensitivity of 85.2% (95%CI 72.9–93.4) for the detection of moderate to severe AG at any location. Conclusion: This study shows that PG testing by CLEIA represents an accurate assay for the detection of corpus AG. Additionally, IL-6 and HE-4 may be of interest for the detection of antrum AG. Mini-abstract: Pepsinogens testing by chemiluminescent enzyme immunoassay is accurate for the detection of corpus atrophic gastritis. IL-6 and HE-4 maybe of interest for the detection of antrum atrophic gastritis.
Collapse
|
53
|
Mommersteeg M, Simovic I, Yu B, van Nieuwenburg S, Bruno IM, Doukas M, Kuipers E, Spaander M, Peppelenbosch M, Castaño-Rodríguez N, Fuhler G. Autophagy mediates ER stress and inflammation in Helicobacter pylori-related gastric cancer. Gut Microbes 2022; 14:2015238. [PMID: 34965181 PMCID: PMC8726742 DOI: 10.1080/19490976.2021.2015238] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Autophagy is a cellular degradation mechanism, which is triggered by the bacterium Helicobacter pylori. A single nucleotide polymorphism (SNP) in the autophagy gene ATG16L1 (rs2241880, G-allele) has been shown to dysregulate autophagy and increase intestinal endoplasmic reticulum (ER) stress. Here, we investigate the role of this SNP in H.pylori-mediated gastric carcinogenesis and its molecular pathways. ATG16L1 rs2241880 was genotyped in subjects from different ethnic cohorts (Dutch and Australian) presenting with gastric (pre)malignant lesions of various severity. Expression of GRP78 (a marker for ER stress) was assessed in gastric tissues. The effect of ATG16L1 rs2241880 on H.pylori-mediated ER stress and pro-inflammatory cytokine induction was investigated in organoids and CRISPR/Cas9 modified cell lines. Development of gastric cancer was associated with the ATG16L1 rs2241880 G-allele. Intestinal metaplastic cells in gastric tissue of patients showed increased levels of ER-stress. In vitro models showed that H.pylori increases autophagy while reducing ER stress, which appeared partly mediated by the ATG16L1 rs2241880 genotype. H.pylori-induced IL-8 production was increased while TNF-α production was decreased, in cells homozygous for the G-allele. The ATG16L1 rs2241880 G-allele is associated with progression of gastric premalignant lesions and cancer. Modulation of H.pylori-induced ER stress pathways and pro-inflammatory mediators by ATG16L1 rs2441880 may underlie this increased risk.
Collapse
Affiliation(s)
- M.C. Mommersteeg
- Department of Gastroenterology and Hepatology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - I. Simovic
- School of Biotechnology and Biomolecular Sciences, Unsw, Sydney, Australia
| | - B. Yu
- Department of Gastroenterology and Hepatology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - S.A.V. van Nieuwenburg
- Department of Gastroenterology and Hepatology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - I, M.J. Bruno
- Department of Gastroenterology and Hepatology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - M. Doukas
- Department of Pathology, Erasmus University Medical Center, Rotterdam, Netherlands
| | - E.J. Kuipers
- Department of Gastroenterology and Hepatology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - M.C.W. Spaander
- Department of Gastroenterology and Hepatology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - M.P. Peppelenbosch
- Department of Gastroenterology and Hepatology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - N. Castaño-Rodríguez
- School of Biotechnology and Biomolecular Sciences, Unsw, Sydney, Australia,CONTACT N. Castaño-Rodríguez School of Biotechnology and Biomolecular Sciences, UNSW Sydney, NSW 2052, Australia
| | - G.M. Fuhler
- Department of Gastroenterology and Hepatology, Erasmus University Medical Center, Rotterdam, The Netherlands,G.M. Fuhler PhD Department of Gastroenterology and Hepatology, Erasmus University Medical Center, Unsw, Rotterdam, The Netherlands
| |
Collapse
|
54
|
Abstract
Gastric cancer (GC) is a leading contributor to global cancer incidence and mortality. Pioneering genomic studies, focusing largely on primary GCs, revealed driver alterations in genes such as ERBB2, FGFR2, TP53 and ARID1A as well as multiple molecular subtypes. However, clinical efforts targeting these alterations have produced variable results, hampered by complex co-alteration patterns in molecular profiles and intra-patient genomic heterogeneity. In this Review, we highlight foundational and translational advances in dissecting the genomic cartography of GC, including non-coding variants, epigenomic aberrations and transcriptomic alterations, and describe how these alterations interplay with environmental influences, germline factors and the tumour microenvironment. Mapping of these alterations over the GC life cycle in normal gastric tissues, metaplasia, primary carcinoma and distant metastasis will improve our understanding of biological mechanisms driving GC development and promoting cancer hallmarks. On the translational front, integrative genomic approaches are identifying diverse mechanisms of GC therapy resistance and emerging preclinical targets, enabled by technologies such as single-cell sequencing and liquid biopsies. Validating these insights will require specifically designed GC cohorts, converging multi-modal genomic data with longitudinal data on therapeutic challenges and patient outcomes. Genomic findings from these studies will facilitate 'next-generation' clinical initiatives in GC precision oncology and prevention.
Collapse
Affiliation(s)
- Khay Guan Yeoh
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Department of Gastroenterology and Hepatology, National University Health System, Singapore, Singapore
- Singapore Gastric Cancer Consortium, Singapore, Singapore
| | - Patrick Tan
- Singapore Gastric Cancer Consortium, Singapore, Singapore.
- Cancer and Stem Cell Biology, Duke-NUS Medical School Singapore, Singapore, Singapore.
- Genome Institute of Singapore, Singapore, Singapore.
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore.
| |
Collapse
|
55
|
Wang JE, Kim SE, Lee BE, Park S, Hwang JH, Huang RJ. The risk of diffuse-type gastric cancer following diagnosis with gastric precancerous lesions: a systematic review and meta-analysis. Cancer Causes Control 2022; 33:183-191. [PMID: 34797436 PMCID: PMC8776597 DOI: 10.1007/s10552-021-01522-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Accepted: 11/11/2021] [Indexed: 02/03/2023]
Abstract
PURPOSE Gastric cancers are classified as diffuse-type (DTGC) or intestinal-type (ITGC). DTGCs have distinct clinical and histopathologic features, and carry a worse overall prognosis compared to ITGCs. Atrophic gastritis (AG) and intestinal metaplasia (IM) are known precursors to ITGC. It is unknown if AG and IM increase risk for DTGC. METHODS We performed a systematic review to identify studies reporting on the association of AG/IM and DTGC. We extracted the odds ratio (OR) of the association from studies, and performed pool analysis. Subgroup analysis was performed on studies reporting histologic severity (using operative link systems) to assess if histologic severity of AG/IM was associated with higher risk. RESULTS We identified six case-control and eight cohort studies for inclusion. Both AG (pooled OR = 1.9, 95% CI 1.5 to 2.4, p < 0.001) and IM (pooled OR = 2.3, 95% CI 1.9 to 2.9, p < 0.001) demonstrated an association with DTGC. High AG severity was associated with increased risk for DTGC compared to low AG severity (OR = 1.7, 95% CI 1.2 to 2.3, p = 0.002). Similarly, high IM severity was associated with increased risk compared to low IM severity (OR = 1.9, 95% CI 1.3 to 2.7, p = 0.001). CONCLUSION Both AG and IM are associated with DTGC. Increasing histologic severity of both AG and IM increases risk for DTGC. There may exist a common pathway between ITGC and some DTGCs mediated through mucosal precursor lesions. These data may inform future strategies of cancer risk attenuation and control.
Collapse
Affiliation(s)
- John E Wang
- Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Sung Eun Kim
- Division of Gastroenterology and Hepatology, Stanford University School of Medicine, 300 Pasteur Drive, Alway Building, M211, Stanford, CA, 94305, USA
| | - Bong Eun Lee
- Division of Gastroenterology and Hepatology, Stanford University School of Medicine, 300 Pasteur Drive, Alway Building, M211, Stanford, CA, 94305, USA
| | - Sungho Park
- Division of Gastroenterology and Hepatology, Stanford University School of Medicine, 300 Pasteur Drive, Alway Building, M211, Stanford, CA, 94305, USA
| | - Joo Ha Hwang
- Division of Gastroenterology and Hepatology, Stanford University School of Medicine, 300 Pasteur Drive, Alway Building, M211, Stanford, CA, 94305, USA
| | - Robert J Huang
- Division of Gastroenterology and Hepatology, Stanford University School of Medicine, 300 Pasteur Drive, Alway Building, M211, Stanford, CA, 94305, USA.
| |
Collapse
|
56
|
Advances in the Aetiology & Endoscopic Detection and Management of Early Gastric Cancer. Cancers (Basel) 2021; 13:cancers13246242. [PMID: 34944861 PMCID: PMC8699285 DOI: 10.3390/cancers13246242] [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: 11/22/2021] [Revised: 12/06/2021] [Accepted: 12/10/2021] [Indexed: 12/24/2022] Open
Abstract
Simple Summary Gastric adenocarcinoma has remained a highly lethal disease. Awareness and recognition of preneoplastic conditions (including gastric atrophy and intestinal metaplasia) using high-resolution white-light endoscopy as well as chromoendoscopy is therefore essential. Helicobacter pylori, a class I carcinogen, remains the main contributor to the development of sporadic distal gastric neoplasia. Management of early gastric neoplasia with endoscopic resections should be in line with standard indications. A multidisciplinary approach to any case of an early gastric neoplasia is imperative. Hereditary forms of gastric cancer require a tailored approach and individua-lized surveillance. Abstract The mortality rates of gastric carcinoma remain high, despite the progress in research and development in disease mechanisms and treatment. Therefore, recognition of gastric precancerous lesions and early neoplasia is crucial. Two subtypes of sporadic gastric cancer have been recognized: cardia subtype and non-cardia (distal) subtype, the latter being more frequent and largely associated with infection of Helicobacter pylori, a class I carcinogen. Helicobacter pylori initiates the widely accepted Correa cascade, describing a stepwise progression through precursor lesions from chronic inflammation to gastric atrophy, gastric intestinal metaplasia and neoplasia. Our knowledge on He-licobacter pylori is still limited, and multiple questions in the context of its contribution to the pathogenesis of gastric neoplasia are yet to be answered. Awareness and recognition of gastric atrophy and intestinal metaplasia on high-definition white-light endoscopy, image-enhanced endoscopy and magnification endoscopy, in combination with histology from the biopsies taken accurately according to the protocol, are crucial to guiding the management. Standard indications for endoscopic resections (endoscopic mucosal resection and endoscopic submucosal dissection) of gastric dysplasia and intestinal type of gastric carcinoma have been recommended by multiple societies. Endoscopic evaluation and surveillance should be offered to individuals with an inherited predisposition to gastric carcinoma.
Collapse
|
57
|
De Re V, Vettori R, Cannizzaro R. Biomarkers and OLGIM Stage for Prospective Preneoplastic Risk Stratification. Clin Gastroenterol Hepatol 2021; 19:2677. [PMID: 33571714 DOI: 10.1016/j.cgh.2021.02.012] [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: 01/18/2021] [Revised: 02/01/2021] [Accepted: 02/05/2021] [Indexed: 02/07/2023]
Affiliation(s)
- Valli De Re
- 1-Unit of Immunopathologia e Biomarcatori Oncologici/Bio-Proteomics Facility, Department of Research and Advanced Tumor Diagnostics, Centro di Riferimento Oncologico di Aviano, IRCCS, Aviano, Italy
| | - Roberto Vettori
- 1-Unit of Immunopathologia e Biomarcatori Oncologici/Bio-Proteomics Facility, Department of Research and Advanced Tumor Diagnostics, Centro di Riferimento Oncologico di Aviano, IRCCS, Aviano, Italy
| | - Renato Cannizzaro
- 2-Unit of Oncological Gastroenterology, Department of Medical Oncology, Centro di Riferimento Oncologico di Aviano, IRCCS, Aviano, Italy
| |
Collapse
|
58
|
Huang RJ, Hwang JH. Reply. Clin Gastroenterol Hepatol 2021; 19:2677-2678. [PMID: 33716138 DOI: 10.1016/j.cgh.2021.03.010] [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: 02/28/2021] [Accepted: 03/09/2021] [Indexed: 02/07/2023]
Affiliation(s)
- Robert J Huang
- Division of Gastroenterology and Hepatology, Department of Medicine, Stanford University, Stanford, California
| | - Joo Ha Hwang
- Division of Gastroenterology and Hepatology, Department of Medicine, Stanford University, Stanford, California
| |
Collapse
|
59
|
Gu J, Xie S, Wang S, Xue L, Zhou J, Li M, Fan Z, Chen R, Middleton DRS, Hao C, Wang J, Li B, Li X, Wei W. Surveillance of premalignant gastric cardia lesions: A population-based prospective cohort study in China. Int J Cancer 2021; 149:1639-1648. [PMID: 34181269 DOI: 10.1002/ijc.33720] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2021] [Revised: 05/20/2021] [Accepted: 06/14/2021] [Indexed: 12/24/2022]
Abstract
In our study, we aimed to assess the long-term risk of gastric cardia adenocarcinoma (GCA) for patients with different histological cardia lesions to inform future guidelines for GCA screening in China. We conducted a population-based prospective study among 9740 subjects who underwent upper endoscopy screening during 2005 to 2009 and followed until December 2017. Cumulative incidence and mortality rates of GCA were calculated by the baseline histological diagnoses, and the hazard ratios (HRs), overall and by age and sex, were analyzed by Cox proportional hazards models. During a median follow-up of 10 years, we identified 123 new GCA cases (1.26%) and 31 GCA deaths (0.32%). The age-standardized incidence and mortality rates of GCA were 128.71/100 000 and 35.69/100 000 person-years, and cumulative incidence rate in patients with cardia high-grade dysplasia (CHGD), cardia low-grade dysplasia (CLGD) and atrophic carditis (AC)/cardia intestinal metaplasia (CIM) was 25%, 3.05% and 1.58%, respectively. The progression rate and cancer risk of GCA increased monotonically with each step in Correa's cascade. Individuals aged 50 to 69 years had 4.4 times higher GCA incidence than those aged 40 to 49 years. Patients with CLGD had a significantly higher 3-year GCA incidence than the normal group, while patients with AC/CIM had a comparable GCA risk during 3-year follow-up but a higher risk at 5-year intervals. Our results suggested a postponed starting age of 50 years for GCA screening, immediate treatment for patients with CHGD, a 3-year surveillance interval for patients with CLGD, and a lengthened surveillance interval of 5 years for patients with AC/CIM.
Collapse
Affiliation(s)
- Jianhua Gu
- Office of National Central Cancer Registry, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Shuanghua Xie
- Office of National Central Cancer Registry, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Shaoming Wang
- Office of National Central Cancer Registry, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Liyan Xue
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jiachen Zhou
- Department of Epidemiology and Biostatistics, School of Public Health, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, China
| | - Minjuan Li
- Office of National Central Cancer Registry, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zhiyuan Fan
- Office of National Central Cancer Registry, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Ru Chen
- Office of National Central Cancer Registry, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Daniel R S Middleton
- Section of Environment and Radiation, International Agency for Research on Cancer, Lyon, France
| | - Changqing Hao
- Department of Endoscopy, Cancer Institute/Hospital of Linzhou, Linzhou, China
| | - Jinwu Wang
- Department of Pathology, Cancer Institute/Hospital of Linzhou, Linzhou, China
| | - Bianyun Li
- Office of National Central Cancer Registry, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Department of Epidemiology, Cancer Institute/Hospital of Linzhou, Linzhou, China
| | - Xinqing Li
- Office of National Central Cancer Registry, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Wenqiang Wei
- Office of National Central Cancer Registry, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
60
|
Abstract
PURPOSE OF REVIEW Gastric intestinal metaplasia (GIM) is an attractive target for surveillance and treatment as it can progress to gastric adenocarcinoma (GAC). Yet, GIM remains a challenging area for clinicians as most patients do not progress to cancer, and there are conflicting data regarding the benefits of surveillance and therapy. This review aims to summarize recently published GIM surveillance guidelines, to discuss, which patients with GIM may benefit from treatment, and to review pivotal and recent literature on GIM therapy. RECENT FINDINGS Guidelines published by American, British, and European gastroenterology societies do not recommend universal surveillance, but do suggest endoscopic surveillance in patients with risk factors for progression to GAC. Although light examination for at least 7 min and mapping biopsies may increase yield for dysplasia and GAC. In randomized trials, Helicobacter pylori eradication reduced risk of dysplasia and cancer. In GIM with visible dysplasia and early-stage GAC, endoscopic resection improves quality of life without reducing survival compared with surgery. Endoscopic ablation therapies have shown promise for invisible or extensive dysplasia. SUMMARY Endoscopic resection is appropriate for visible dysplasia and early-stage GAC without high-risk features that persists despite H. pylori eradication therapy. Prospective studies are needed to assess the utility of endoscopic ablation in GIM.
Collapse
Affiliation(s)
- Thaer Abdelfattah
- Division of Gastroenterology and Hepatology, Virginia Commonwealth University
- Division of Gastroenterology and Hepatology, McGuire VA Medical Center, Richmond, Virginia, USA
| | - Omer Shahab
- Division of Gastroenterology and Hepatology, Virginia Commonwealth University
- Division of Gastroenterology and Hepatology, McGuire VA Medical Center, Richmond, Virginia, USA
| | - Tilak U Shah
- Division of Gastroenterology and Hepatology, Virginia Commonwealth University
- Division of Gastroenterology and Hepatology, McGuire VA Medical Center, Richmond, Virginia, USA
| |
Collapse
|
61
|
Grad C, Pop A, Gaborean E, Grad S, Dumitrascu D. Value of GastroPanel in the diagnosis of atrophic gastritis. Exp Ther Med 2021; 22:1347. [PMID: 34630701 PMCID: PMC8495588 DOI: 10.3892/etm.2021.10782] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Accepted: 06/15/2021] [Indexed: 01/10/2023] Open
Abstract
Analysis of serum biomarkers for the assessment of atrophic gastritis (AG), considered as precursor of the intestinal type of gastric cancer, is of growing interest. The combination of pepsinogen (PG), gastrin-17 (G17) and anti-Helicobacter pylori (H. pylori) antibody serological assays (panel test) is a non-invasive tool for the diagnosis of atrophic gastritis. However, the diagnostic reliability of this test remains uncertain. The aim of our study was to assess the diagnostic performance of the serum panel test (GastroPanel) for the diagnosis of atrophic gastritis. From dyspeptic patients, endoscopic biopsy samples (two from the gastric corpus and two from the antrum) and blood samples were collected. The determination of sPGI, sPGII, sG17 and IgG antibodies to H. pylori (H.p IgG) was performed using an enzyme-linked immunosorbent assay (GastroPanel; Biohit Oyj). Histopathology results were compared with GastroPanel values. Sixty patients were included: 35 (58.3%) females and 25 (41.66%) males; mean age 67.63±9.36 years; 45% H. pylori-positive. A total of 65% of patients had atrophic gastritis. There were no significant differences between the levels of biomarkers and localization of atrophy. The ratio PG1/PG2 was lower in patients with multifocal atrophy; the difference being close to the threshold of statistical significance. In cases of intestinal metaplasia the values of G17, PG1, PG2, H.p IgG were not statistically altered compared to those without intestinal metaplasia; only the ratio PG1/PG2 was lower in intestinal metaplasia; the difference being almost of statistical significance. Our results revealed that, GastroPanel values did not differ depending on the severity of the atrophy. Biomarkers used by GastroPanel do not have enough accuracy for use in the diagnosis of atrophy in the population studied. A low accuracy only for the ratio PG1/PG2 in patients with multifocal atrophy was found. However, our data revealed a correlation in detecting intestinal metaplasia.
Collapse
Affiliation(s)
- Cosmin Grad
- Second Medical Department, 'Iuliu Hatieganu' University of Medicine and Pharmacy, 400006 Cluj-Napoca, Romania
| | - Andrei Pop
- Second Medical Department, 'Iuliu Hatieganu' University of Medicine and Pharmacy, 400006 Cluj-Napoca, Romania
| | - Emil Gaborean
- Second Medical Department, 'Iuliu Hatieganu' University of Medicine and Pharmacy, 400006 Cluj-Napoca, Romania
| | - Simona Grad
- Second Medical Department, 'Iuliu Hatieganu' University of Medicine and Pharmacy, 400006 Cluj-Napoca, Romania
| | - Dan Dumitrascu
- Second Medical Department, 'Iuliu Hatieganu' University of Medicine and Pharmacy, 400006 Cluj-Napoca, Romania
| |
Collapse
|
62
|
Molecular Landscapes of Gastric Pre-Neoplastic and Pre-Invasive Lesions. Int J Mol Sci 2021; 22:ijms22189950. [PMID: 34576114 PMCID: PMC8468646 DOI: 10.3390/ijms22189950] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 09/03/2021] [Accepted: 09/11/2021] [Indexed: 12/24/2022] Open
Abstract
Gastric carcinoma (GC) represents one of the most common and most lethal malignancies worldwide. The histopathological characterization of GC precursor lesions has provided great knowledge about gastric carcinogenesis, with the consequent introduction of effective strategies of primary and secondary prevention. In recent years, a large amount of data about the molecular events in GC development is emerging, flanking the histomorphological descriptions. In this review, we describe the landscape of molecular alterations in gastric pre-invasive lesions with a glance at their potential use in the diagnostic and therapeutic decision-making process.
Collapse
|
63
|
Aumpan N, Vilaichone RK, Pornthisarn B, Chonprasertsuk S, Siramolpiwat S, Bhanthumkomol P, Nunanan P, Issariyakulkarn N, Ratana-Amornpin S, Miftahussurur M, Mahachai V, Yamaoka Y. Predictors for regression and progression of intestinal metaplasia (IM): A large population-based study from low prevalence area of gastric cancer (IM-predictor trial). PLoS One 2021; 16:e0255601. [PMID: 34379655 PMCID: PMC8357097 DOI: 10.1371/journal.pone.0255601] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Accepted: 07/19/2021] [Indexed: 12/12/2022] Open
Abstract
Background Gastric intestinal metaplasia (IM) can lead to gastric cancer. Until now, there have been limited studies of predictors for regression and progression of IM. This study aimed to determine risk factors associated with regression or progression of IM for guiding proper management and prevention of gastric cancer. Methods 2,025 patients undergoing gastroscopy in Thammasat University Hospital, Thailand were enrolled during September 2017-August 2019. Patients’ data including baseline characteristics, laboratory results, and histopathology of gastric biopsies from University medical database were extensively reviewed. Results 2,025 patients had mean age of 61.3 years and 44.2% were males. Overall H. pylori prevalence was 47.5%. There were 1,551(76.6%) patients with chronic gastritis and 361(17.8%) with IM. Of 400 patients with chronic gastritis having follow-up endoscopy and repeated gastric biopsies, 104(26%) had persistent H. pylori infection and 27(26%) developed IM during mean follow-up time of 24 months. Persistent H. pylori infection was significantly associated with development of IM (OR 3.16, 95%CI 1.56–6.39, p = 0.001). Regression, persistence, and progression of IM were demonstrated in 57.3%, 39.2%, and 3.5% of patients, respectively. Age >65 years, persistent H. pylori infection, and diabetes mellitus were significantly associated with persistent IM or progression to dysplasia with OR 2.47(95%CI 1.33–4.61, p = 0.004), OR 2.64(95%CI 1.13–6.18, p = 0.025), and OR 2.54(95%CI 1.16–5.54, p = 0.019), respectively. Patients without H. pylori infection had more IM regression than patients with persistent infection (60.4%vs.39.4%, p = 0.035). Patients with persistent H. pylori infection significantly had higher IM progression to dysplasia (15.2%vs.2.1%; OR 11.15, 95%CI 1.18–105.24, p = 0.035) than noninfected. During 24 months of study, 30 patients (1.5%) were diagnosed with gastric cancer. Conclusion Regression of IM could be achieved by successful H. pylori eradication. Persistent H. pylori infection was significantly associated with development and progression of IM to dysplasia. Age >65 years and diabetes mellitus were also significant predictors for IM progression.
Collapse
Affiliation(s)
- Natsuda Aumpan
- Center of Excellence in Digestive Diseases and Gastroenterology, Faculty of Medicine, Thammasat University, Pathumthani, Thailand
| | - Ratha-korn Vilaichone
- Center of Excellence in Digestive Diseases and Gastroenterology, Faculty of Medicine, Thammasat University, Pathumthani, Thailand
- Chulabhorn International College of Medicine (CICM), Thammasat University, Pathumthani, Thailand
- Division of Gastroentero-Hepatology, Department of Internal Medicine, Faculty of Medicine, Universitas Airlangga, Surabaya, Indonesia
- * E-mail:
| | - Bubpha Pornthisarn
- Center of Excellence in Digestive Diseases and Gastroenterology, Faculty of Medicine, Thammasat University, Pathumthani, Thailand
| | - Soonthorn Chonprasertsuk
- Center of Excellence in Digestive Diseases and Gastroenterology, Faculty of Medicine, Thammasat University, Pathumthani, Thailand
| | - Sith Siramolpiwat
- Center of Excellence in Digestive Diseases and Gastroenterology, Faculty of Medicine, Thammasat University, Pathumthani, Thailand
- Chulabhorn International College of Medicine (CICM), Thammasat University, Pathumthani, Thailand
| | - Patommatat Bhanthumkomol
- Center of Excellence in Digestive Diseases and Gastroenterology, Faculty of Medicine, Thammasat University, Pathumthani, Thailand
| | - Pongjarat Nunanan
- Center of Excellence in Digestive Diseases and Gastroenterology, Faculty of Medicine, Thammasat University, Pathumthani, Thailand
| | - Navapan Issariyakulkarn
- Center of Excellence in Digestive Diseases and Gastroenterology, Faculty of Medicine, Thammasat University, Pathumthani, Thailand
| | - Sarita Ratana-Amornpin
- Center of Excellence in Digestive Diseases and Gastroenterology, Faculty of Medicine, Thammasat University, Pathumthani, Thailand
| | - Muhammad Miftahussurur
- Division of Gastroentero-Hepatology, Department of Internal Medicine, Faculty of Medicine, Universitas Airlangga, Surabaya, Indonesia
| | - Varocha Mahachai
- Chulabhorn International College of Medicine (CICM), Thammasat University, Pathumthani, Thailand
| | - Yoshio Yamaoka
- Department of Environmental and Preventive Medicine, Faculty of Medicine, Oita University, Yufu, Japan
| |
Collapse
|
64
|
Abstract
Data on the evolution of gastric precancerous lesions (GPL), especially in countries of a Low gastric cancer incidence area are limited. Our objective was to study a long-term evolution of GPL in France.
Collapse
|
65
|
Chen Z, Li Y, Tan B, Li F, Zhao Q, Fan L, Zhang Z, Zhao X, Liu Y, Wang D. Long Non-coding RNA ASNR Targeting miR-519e-5p Promotes Gastric Cancer Development by Regulating FGFR2. Front Cell Dev Biol 2021; 9:679176. [PMID: 34307360 PMCID: PMC8299726 DOI: 10.3389/fcell.2021.679176] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Accepted: 06/10/2021] [Indexed: 12/24/2022] Open
Abstract
Gastric cancer (GC), as a common gastrointestinal tumor, is an important cause of death from cancer all around the world. Long non-coding RNAs (lncRNAs), a novel class of transcripts, have attracted great attention of researchers. However, the mechanisms of the clinical significance of most lncRNAs in human cancer are mainly undocumented. This research desires to explore the clinical significance, biological function, and mechanism of Lnc_ASNR (apoptosis suppressing-non-coding RNA) in GC. Cell proliferation, cell cycle, cell migration, and invasion abilities were respectively determined by 3-(4,5)-dimethylthiahiazo (-z-y1)-3,5-di-phenytetrazoliumromide (MTT), flow cytometry, wound healing, and Transwell assay (Sigma-Aldrich, St. Louis, MO, United States). The association of Lnc_ASNR, miR-519e-5p, and fibroblast growth factor receptor 2 (FGFR2) was evaluated via luciferase reporter experiments. The tumor xenograft assay was conducted to confirm the results of cell experiments. High expressed Lnc_ASNR was detected in both GC cells and tissues using qRT-PCR. Downregulated Lnc_ASNR could reduce proliferation, migration, and invasion in GC cells, while upregulated Lnc_ASNR could promote the cell proliferation, migration, and invasion. Moreover, the effect of Lnc_ASNR on migration and invasion ability is closely related to epithelial-mesenchymal transition (EMT). The bioinformatics analysis, luciferase assay, and Western blot demonstrated that Lnc_ASNR inhibited miR-519e-5p expression but increased FGFR2 expression. Lnc_ASNR and FGFR2 were both targeted to miR-519e-5p, and they were negatively correlated with the expression of miR-519e-5p. All investigations indicated that Lnc_ASNR functioned as a ceRNA targeting miR-519e-5p and facilitated GC development by regulating the pathway of miR-519e-5p/FGFR2.
Collapse
Affiliation(s)
- Zihao Chen
- Graduate School of Hebei Medical University, Shijiazhuang, China.,The Third Department of Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Yong Li
- The Third Department of Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Bibo Tan
- The Third Department of Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Fang Li
- Department of Pathology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Qun Zhao
- The Third Department of Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Liqiao Fan
- The Third Department of Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Zhidong Zhang
- The Third Department of Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Xuefeng Zhao
- The Third Department of Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Yu Liu
- The Third Department of Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Dong Wang
- The Third Department of Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| |
Collapse
|
66
|
Abstract
Gastric cancer (GC) remains a leading cause of cancer morbidity and mortality worldwide. Outcomes from GC remain poor, especially in Western nations where cancer diagnosis is usually at advanced stages where curative resection is not possible. By contrast, nations of East Asia have adopted methods of population-level screening with improvements in stage of diagnosis and survival. In this review, the authors discuss the epidemiology of GC in Western populations, highlight at-risk populations who may benefit from screening, overview screening modalities, and discuss promising approaches to early GC detection.
Collapse
Affiliation(s)
- Robert J Huang
- Division of Gastroenterology and Hepatology, Stanford University, 300 Pasteur Drive, Alway Building M211, Stanford, CA 94305, USA.
| | - Joo Ha Hwang
- Division of Gastroenterology and Hepatology, Stanford University, 300 Pasteur Drive, Alway Building M211, Stanford, CA 94305, USA
| |
Collapse
|
67
|
Sánchez-Alonzo K, Silva-Mieres F, Arellano-Arriagada L, Parra-Sepúlveda C, Bernasconi H, Smith CT, Campos VL, García-Cancino A. Nutrient Deficiency Promotes the Entry of Helicobacter pylori Cells into Candida Yeast Cells. BIOLOGY 2021; 10:426. [PMID: 34065788 PMCID: PMC8151769 DOI: 10.3390/biology10050426] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 05/02/2021] [Accepted: 05/04/2021] [Indexed: 12/14/2022]
Abstract
Helicobacter pylori, a Gram-negative bacterium, has as a natural niche the human gastric epithelium. This pathogen has been reported to enter into Candida yeast cells; however, factors triggering this endosymbiotic relationship remain unknown. The aim of this work was to evaluate in vitro if variations in nutrient concentration in the cultured medium trigger the internalization of H. pylori within Candida cells. We used H. pylori-Candida co-cultures in Brucella broth supplemented with 1%, 5% or 20% fetal bovine serum or in saline solution. Intra-yeast bacteria-like bodies (BLBs) were observed using optical microscopy, while intra-yeast BLBs were identified as H. pylori using FISH and PCR techniques. Intra-yeast H. pylori (BLBs) viability was confirmed using the LIVE/DEAD BacLight Bacterial Viability kit. Intra-yeast H. pylori was present in all combinations of bacteria-yeast strains co-cultured. However, the percentages of yeast cells harboring bacteria (Y-BLBs) varied according to nutrient concentrations and also were strain-dependent. In conclusion, reduced nutrients stresses H. pylori, promoting its entry into Candida cells. The starvation of both H. pylori and Candida strains reduced the percentages of Y-BLBs, suggesting that starving yeast cells may be less capable of harboring stressed H. pylori cells. Moreover, the endosymbiotic relationship between H. pylori and Candida is dependent on the strains co-cultured.
Collapse
Affiliation(s)
- Kimberly Sánchez-Alonzo
- Laboratory of Bacterial Pathogenicity, Department of Microbiology, Faculty of Biological Sciences, Universidad de Concepcion, Concepción 4070386, Chile; (K.S.-A.); (F.S.-M.); (L.A.-A.); (C.P.-S.); (C.T.S.)
| | - Fabiola Silva-Mieres
- Laboratory of Bacterial Pathogenicity, Department of Microbiology, Faculty of Biological Sciences, Universidad de Concepcion, Concepción 4070386, Chile; (K.S.-A.); (F.S.-M.); (L.A.-A.); (C.P.-S.); (C.T.S.)
| | - Luciano Arellano-Arriagada
- Laboratory of Bacterial Pathogenicity, Department of Microbiology, Faculty of Biological Sciences, Universidad de Concepcion, Concepción 4070386, Chile; (K.S.-A.); (F.S.-M.); (L.A.-A.); (C.P.-S.); (C.T.S.)
| | - Cristian Parra-Sepúlveda
- Laboratory of Bacterial Pathogenicity, Department of Microbiology, Faculty of Biological Sciences, Universidad de Concepcion, Concepción 4070386, Chile; (K.S.-A.); (F.S.-M.); (L.A.-A.); (C.P.-S.); (C.T.S.)
| | | | - Carlos T. Smith
- Laboratory of Bacterial Pathogenicity, Department of Microbiology, Faculty of Biological Sciences, Universidad de Concepcion, Concepción 4070386, Chile; (K.S.-A.); (F.S.-M.); (L.A.-A.); (C.P.-S.); (C.T.S.)
| | - Víctor L. Campos
- Laboratory of Environmental Microbiology, Department of Microbiology, Faculty of Biological Sciences, Universidad de Concepción, Concepción 4070386, Chile;
| | - Apolinaria García-Cancino
- Laboratory of Bacterial Pathogenicity, Department of Microbiology, Faculty of Biological Sciences, Universidad de Concepcion, Concepción 4070386, Chile; (K.S.-A.); (F.S.-M.); (L.A.-A.); (C.P.-S.); (C.T.S.)
| |
Collapse
|
68
|
Li J, Ren M, Yang J, Zhao Y, Li Y, Zhang D, Wu F, Zhang Z, Lu X, Ren L, He S, Lu G. Screening value for gastrointestinal lesions of magnetic-controlled capsule endoscopy in asymptomatic individuals. J Gastroenterol Hepatol 2021; 36:1267-1275. [PMID: 33000488 DOI: 10.1111/jgh.15282] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2020] [Revised: 08/16/2020] [Accepted: 09/25/2020] [Indexed: 02/05/2023]
Abstract
BACKGROUND AND AIM Most patients with gastric tumors and precancerous lesions are asymptomatic, which often results in delayed diagnosis and treatment. Compared with conventional gastroscopy and capsule endoscopy, magnetic-controlled capsule endoscopy is a non-invasive, effective, and cost-efficient diagnostic modality for gastric examination. We retrospectively investigated magnetic-controlled capsule endoscopy as a screening tool for gastrointestinal lesions (particularly gastric tumors and precancerous lesions) in asymptomatic individuals. METHODS In this retrospective study, 1757 patients who voluntarily underwent magnetic-controlled capsule endoscopy between January and December 2019 at nine medical centers across Shaanxi province based on strict inclusion and exclusion criteria were enrolled. The primary outcomes were gastric tumor and precancerous lesion detection rates and procedural safety. RESULTS The upper and lower gastrointestinal lesion detection rates were 98.35% (1728/1757) and 21.61% (78/361), respectively; 2.28% of patients were diagnosed with gastric tumors including gastric cancer (4/1757) and submucosal tumors (36/1757). Three types of precancerous lesions were found in 591 patients (33.64%), including chronic atrophic gastritis (23.16%), gastric polyp (10.98%), and gastric ulcer (2.96%). For patients aged over 40 years, the detection rate of precancerous lesions was higher (14.36% vs 42.58%, P < 0.001). No patient was diagnosed with small intestinal cancer. No adverse events occurred. CONCLUSIONS Magnetic-controlled capsule endoscopy could be used as a promising novel screening modality for diagnosis of gastrointestinal lesions in asymptomatic individuals, specifically gastric tumors and precancerous lesions, with the advantages of safety, non-invasiveness, effectiveness, and cost-efficiency.
Collapse
Affiliation(s)
- Jing Li
- Department of Gastroenterology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Mudan Ren
- Department of Gastroenterology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Jiahui Yang
- Department of Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Yan Zhao
- Department of Gastroenterology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Yarui Li
- Department of Gastroenterology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Dan Zhang
- Department of Gastroenterology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Fangli Wu
- Department of Gastroenterology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Zhiyong Zhang
- Department of Gastroenterology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Xinlan Lu
- Department of Gastroenterology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Li Ren
- Department of Gastroenterology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Shuixiang He
- Department of Gastroenterology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Guifang Lu
- Department of Gastroenterology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
69
|
Coelho MCF, Ribeiro HG, Gomes CGDO, Marinho FP, Barbosa AJA, Coelho LGV. HELICOBACTER PYLORI CHRONIC GASTRITIS ON PATIENTS WITH PREMALIGNANT CONDITIONS: OLGA AND OLGIM EVALUATION AND SERUM BIOMARKERS PERFORMANCE. ARQUIVOS DE GASTROENTEROLOGIA 2021; 58:39-47. [PMID: 33909795 DOI: 10.1590/s0004-2803.202100000-08] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Accepted: 10/15/2020] [Indexed: 02/07/2023]
Abstract
BACKGROUND H. pylori chronic atrophic gastritis is a premalignant lesion, and its staging, according to OLGA and OLGIM systems aims to identify patients at increased risk of developing gastric cancer and optimize their follow-up. GastroPanel®, serum biomarkers panel including pepsinogen I (PGI), pepsinogen II (PGII), Gastrin 17 (G17) and anti- H. pylori antibodies is a noninvasive test for adenocarcinoma risk assessment in chronic H. pylori gastritis patients. OBJECTIVE Prospective study to evaluate the concordance between OLGA and OLGIM grading systems, as well as to evaluate GastroPanel´s performance in patients with premalignant lesions secondary to H. pylori chronic gastritis in Brazil. METHODS Patients with H. pylori chronic gastritis with premalignant lesions confirmed by histology were recruited from the gastrointestinal clinic of a University Hospital. All participants underwent endoscopic examination with biopsies which were reported according to updated Sydney system and premalignant lesions grading systems (OLGA and OLGIM). Blood samples were collected for biomarkers serological analysis (GastroPanel®, Biohit, Helsinki, Finland). The cut off values used to define high risk patients were those recommended by the manufacturer: PGI ≤30 µm/L and PGI/PGII ≤3. RESULTS 41 patients were recruited: 28 women, 13 men, mean age 67.3 (47-89, SD: 9.6) years. By OLGA system, were obtained: OLGA 0 (n=1), OLGA I (n=7), OLGA II (n=17), OLGA III (n=9), and OLGA IV (n=7). By OLGIM system, were obtained: OLGIM 0 (n=14), OLGIM I (n=5), OLGIM II (n=10), OLGIM III (n=10), and OLGIM IV (n=2). Regarding histological staging among patients staged as low risk (OLGA/OLGIM 0, I and II) and high risk (OLGA/OLGIM III and IV) for gastric cancer development, the concordance rate found between both classifications was 85.4%. Considering high risk patients, those patients thus included in at least one of the systems the final distribution of our sample considered 24 low-risk and 17 high-risk patients for the development of gastric cancer. To determine by GastroPanel® whether the patient would be at low or high risk of developing gastric cancer, PGI showed a sensitivity, specificity and accuracy of 0.47 (95%CI: 0.26-0.69), 0.67 (95%CI: 0.47-0.82), and 0.58 (95%CI: 0.43-0.72), respectively, while PGI/PGII showed sensitivity, specificity and accuracy of 0.06 (95%CI: 0.01-0.27), 0.83 (95%CI: 0.64-0.93) and 0.51 (95%CI: 0.36-0.66), respectively. CONCLUSION The histological classifications OLGA and OLGIM presented a substantial concordance rate among themselves. Simultaneous use of both histological classification systems increased the identification's rate of high-risk patients. Biomarker analysis was not effective to distinguish low to high risk patients in the studied population. Further studies are needed to validate its use in clinical practice in Brazil.
Collapse
Affiliation(s)
- Maria Clara Freitas Coelho
- Universidade Federal de Minas Gerais, Faculdade de Medicina, Programa de Pós-Graduação em Ciências Aplicadas à Saúde do Adulto, Belo Horizonte, MG, Brasil.,Faculdade de Ciências Médicas de Minas Gerais, Belo Horizonte, MG, Brasil
| | - Henrique Gomes Ribeiro
- Universidade Federal de Minas Gerais, Instituto Alfa de Gastroenterologia, Hospital das Clínicas, Belo Horizonte, MG, Brasil
| | - Celio Geraldo de Oliveira Gomes
- Universidade Federal de Minas Gerais, Instituto Alfa de Gastroenterologia, Hospital das Clínicas, Belo Horizonte, MG, Brasil
| | - Frederico Passos Marinho
- Universidade Federal de Minas Gerais, Instituto Alfa de Gastroenterologia, Hospital das Clínicas, Belo Horizonte, MG, Brasil
| | - Alfredo J A Barbosa
- Universidade Federal de Minas Gerais, Faculdade de Medicina, Programa de Pós-Graduação em Ciências Aplicadas à Saúde do Adulto, Belo Horizonte, MG, Brasil.,Universidade Federal de Minas Gerais, Instituto Alfa de Gastroenterologia, Hospital das Clínicas, Belo Horizonte, MG, Brasil
| | - Luiz Gonzaga Vaz Coelho
- Universidade Federal de Minas Gerais, Faculdade de Medicina, Programa de Pós-Graduação em Ciências Aplicadas à Saúde do Adulto, Belo Horizonte, MG, Brasil.,Universidade Federal de Minas Gerais, Instituto Alfa de Gastroenterologia, Hospital das Clínicas, Belo Horizonte, MG, Brasil
| |
Collapse
|
70
|
Lau JWL, Khoo MJW, Leong XH, Lim TZ, Shabbir A, Yeoh KG, Koh CJ, So JBY. Opportunistic upper endoscopy during colonoscopy as a screening strategy for countries with intermediate gastric cancer risk. J Gastroenterol Hepatol 2021; 36:1081-1087. [PMID: 33037826 PMCID: PMC8247052 DOI: 10.1111/jgh.15290] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2020] [Revised: 09/19/2020] [Accepted: 09/28/2020] [Indexed: 12/13/2022]
Abstract
BACKGROUND AND AIM Screening upper endoscopy can detect esophagogastric (OG) cancers early with improved outcomes. Recent cost-utility studies suggest that opportunistic upper endoscopy at the same setting of colonoscopy might be a useful strategy for screening of OG cancers, and it may be more acceptable to the patients due to cost-saving and convenience. We aim to study the diagnostic performance of this screening strategy in a country with intermediate gastric cancer risk. METHODS A retrospective cohort study using a prospective endoscopy database from 2015 to 2017 was performed. Patients included were individuals age > 40 who underwent opportunistic upper endoscopy at the same setting of colonoscopy without any OG symptoms. Neoplastic OG lesions are defined as cancer and high-grade dysplasia. Pre-neoplastic lesions include Barrett's esophagus (BE), intestinal metaplasia (IM), and atrophic gastritis (AG). RESULTS The study population involved 1414 patients. Neoplastic OG lesions were detected in five patients (0.35%). Pre-neoplastic lesions were identified in 174 (12.3%) patients. IM was found in 146 (10.3%) patients with 21 (1.4%) having extensive IM. The number needed to scope to detect a neoplastic OG lesion is 282.8 with an estimated cost of USD$141 400 per lesion detected. On multivariate regression, age ≥ 60 (RR: 1.84, 95% CI: 1.29-2.63) and first-degree relatives with gastric cancer (RR: 1.64, 95% CI: 1.06-2.55) were independent risk factors for neoplastic or pre-neoplastic OG lesion. CONCLUSION For countries with intermediate gastric cancer risk, opportunistic upper endoscopy may be an alternative screening strategy in a selected patient population. Prospective trials are warranted to validate its performance.
Collapse
Affiliation(s)
| | - Mark Junn Wei Khoo
- Yong Loo Lin School of MedicineNational University of SingaporeSingapore
| | - Xue Hao Leong
- Yong Loo Lin School of MedicineNational University of SingaporeSingapore
| | - Tian Zhi Lim
- Department of SurgeryNational University HospitalSingapore
| | - Asim Shabbir
- Division of General Surgery (Upper Gastrointestinal Surgery), Department of SurgeryNational University HospitalSingapore
| | - Khay Guan Yeoh
- Yong Loo Lin School of MedicineNational University of SingaporeSingapore,Division of Gastroenterology and Hepatology, Department of MedicineNational University HospitalSingapore
| | - Calvin Jianyi Koh
- Division of Gastroenterology and Hepatology, Department of MedicineNational University HospitalSingapore
| | - Jimmy Bok Yan So
- Yong Loo Lin School of MedicineNational University of SingaporeSingapore,Division of General Surgery (Upper Gastrointestinal Surgery), Department of SurgeryNational University HospitalSingapore
| |
Collapse
|
71
|
Nieuwenburg SAV, Mommersteeg MC, Eikenboom EL, Yu B, den Hollander WJ, Holster IL, den Hoed CM, Capelle LG, Tang TJ, Anten MP, Prytz-Berset I, Witteman EM, ter Borg F, Burger JPW, Bruno MJ, Fuhler GM, Peppelenbosch MP, Doukas M, Kuipers EJ, Spaander MC. Factors associated with the progression of gastric intestinal metaplasia: a multicenter, prospective cohort study. Endosc Int Open 2021; 9:E297-E305. [PMID: 33655025 PMCID: PMC7892268 DOI: 10.1055/a-1314-6626] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Indexed: 12/13/2022] Open
Abstract
Background and study aims Gastric cancer (GC) is usually preceded by premalignant gastric lesions (GPLs) such as gastric intestinal metaplasia (GIM). Information on risk factors associated with neoplastic progression of GIM are scarce. This study aimed to identify predictors for progression of GIM in areas with low GC incidence. Patients and methods The Progression and Regression of Precancerous Gastric Lesions (PROREGAL) study includes patients with GPL. Patients underwent at least two upper endoscopies with random biopsy sampling. Progression of GIM means an increase in severity according to OLGIM (operative link on gastric intestinal metaplasia) during follow-up (FU). Family history and lifestyle factors were determined through questionnaires. Serum Helicobacter pylori infection, pepsinogens (PG), gastrin-17 and GC-associated single nucleotide polymorphisms (SNPs) were determined. Cox regression was performed for risk analysis and a chi-squared test for analysis of single nucleotide polymorphisms. Results Three hundred and eight patients (median age at inclusion 61 years, interquartile range (IQR: 17; male 48.4 %; median FU 48 months, IQR: 24) were included. During FU, 116 patients (37.7 %) showed progression of IM and six patients (1.9 %) developed high-grade dysplasia or GC. The minor allele (C) on TLR4 (rs11536889) was inversely associated with progression of GIM (OR 0.6; 95 %CI 0.4-1.0). Family history (HR 1.5; 95 %CI 0.9-2.4) and smoking (HR 1.6; 95 %CI 0.9-2.7) showed trends towards progression of GIM. Alcohol use, body mass index, history of H. pylori infection, and serological markers were not associated with progression. Conclusions Family history and smoking appear to be related to an increased risk of GIM progression in low GC incidence countries. TLR4 (rs11536889) showed a significant inverse association, suggesting that genetic information may play a role in GIM progression.
Collapse
Affiliation(s)
- S. A. V. Nieuwenburg
- Departments of Gastroenterology and Hepatology, Erasmus University Medical Centre, Rotterdam, The Netherlands
| | - M. C. Mommersteeg
- Departments of Gastroenterology and Hepatology, Erasmus University Medical Centre, Rotterdam, The Netherlands
| | - E. L. Eikenboom
- Departments of Gastroenterology and Hepatology, Erasmus University Medical Centre, Rotterdam, The Netherlands
| | - B. Yu
- Departments of Gastroenterology and Hepatology, Erasmus University Medical Centre, Rotterdam, The Netherlands
| | | | - I. Lisanne Holster
- Departments of Gastroenterology and Hepatology, Erasmus University Medical Centre, Rotterdam, The Netherlands
| | - Caroline M. den Hoed
- Departments of Gastroenterology and Hepatology, Erasmus University Medical Centre, Rotterdam, The Netherlands
| | - L. G Capelle
- Meander Medical Centre, Amersfoort, the Netherlands
| | - Thjon J. Tang
- IJsselland Hospital, Capelle aan den IJssel, The Netherlands
| | | | | | | | - F. ter Borg
- Deventer Hospital, Deventer, The Netherlands
| | - Jordy P. W. Burger
- Department of Gastroenterology and Hepatology, Rijnstate, Arnhem, The Netherlands
| | - Marco J. Bruno
- Departments of Gastroenterology and Hepatology, Erasmus University Medical Centre, Rotterdam, The Netherlands
| | - G. M. Fuhler
- Departments of Gastroenterology and Hepatology, Erasmus University Medical Centre, Rotterdam, The Netherlands
| | - Maikel P. Peppelenbosch
- Departments of Gastroenterology and Hepatology, Erasmus University Medical Centre, Rotterdam, The Netherlands
| | - Michael Doukas
- Department of Pathology, Erasmus University Medical Centre, Rotterdam, The Netherlands
| | - Ernst J. Kuipers
- Departments of Gastroenterology and Hepatology, Erasmus University Medical Centre, Rotterdam, The Netherlands
| | - Manon C.W. Spaander
- Departments of Gastroenterology and Hepatology, Erasmus University Medical Centre, Rotterdam, The Netherlands
| |
Collapse
|
72
|
Piazuelo MB, Bravo LE, Mera RM, Camargo MC, Bravo JC, Delgado AG, Washington MK, Rosero A, Garcia LS, Realpe JL, Cifuentes SP, Morgan DR, Peek RM, Correa P, Wilson KT. The Colombian Chemoprevention Trial: 20-Year Follow-Up of a Cohort of Patients With Gastric Precancerous Lesions. Gastroenterology 2021; 160:1106-1117.e3. [PMID: 33220252 PMCID: PMC7956231 DOI: 10.1053/j.gastro.2020.11.017] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Revised: 11/10/2020] [Accepted: 11/12/2020] [Indexed: 02/08/2023]
Abstract
BACKGROUND & AIMS Helicobacter pylori eradication and endoscopic surveillance of gastric precancerous lesions are strategies to reduce gastric cancer (GC) risk. To our knowledge, this study is the longest prospective cohort of an H pylori eradication trial in a Hispanic population. METHODS A total of 800 adults with precancerous lesions were randomized to anti-H pylori treatment or placebo. Gastric biopsy samples taken at baseline and 3, 6, 12, 16, and 20 years were assessed by our Correa histopathology score. A generalized linear mixed model with a participant-level random intercept was used to estimate the effect of H pylori status on the score over time. Logistic regression models were used to estimate progression by baseline diagnosis and to estimate GC risk by intestinal metaplasia (IM) subtype and anatomic location. RESULTS Overall, 356 individuals completed 20 years of follow-up. Anti-H pylori therapy (intention-to-treat) reduced progression of the Correa score (odds ratio [OR], 0.59; 95% confidence interval [CI], 0.38-0.93). H pylori-negative status had a beneficial effect on the score over time (P = .036). Among individuals with IM (including indefinite for dysplasia) at baseline, incidence rates per 100 person-years were 1.09 (95% CI, 0.85-1.33) for low-grade/high-grade dysplasia and 0.14 (95% CI, 0.06-0.22) for GC. Incomplete-type (vs complete-type) IM at baseline presented higher GC risk (OR, 13.4; 95% CI, 1.8-103.8). Individuals with corpus (vs antrum-restricted) IM showed an OR of 2.1 (95% CI, 0.7-6.6) for GC. CONCLUSIONS In a high-GC-risk Hispanic population, anti-H pylori therapy had a long-term beneficial effect against histologic progression. Incomplete IM is a strong predictor of GC risk.
Collapse
Affiliation(s)
- M. Blanca Piazuelo
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA,Center for Mucosal Inflammation and Cancer, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Luis E. Bravo
- Department of Pathology, Universidad del Valle School of Medicine, Cali, Valle del Cauca, Colombia
| | - Robertino M. Mera
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - M. Constanza Camargo
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD, USA
| | - Juan C. Bravo
- Department of Pathology, Universidad del Valle School of Medicine, Cali, Valle del Cauca, Colombia
| | - Alberto G. Delgado
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - M. Kay Washington
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| | | | - Luz S. Garcia
- Department of Pathology, Universidad del Valle School of Medicine, Cali, Valle del Cauca, Colombia
| | | | | | - Douglas R. Morgan
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA,Division of Gastroenterology, Department of Medicine, University of Alabama, Birmingham, AL, USA
| | - Richard M. Peek
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA,Center for Mucosal Inflammation and Cancer, Vanderbilt University Medical Center, Nashville, TN, USA,Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Pelayo Correa
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Keith T. Wilson
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA,Center for Mucosal Inflammation and Cancer, Vanderbilt University Medical Center, Nashville, TN, USA,Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA,Veterans Affairs Tennessee Valley Healthcare System, Nashville, TN, USA
| |
Collapse
|
73
|
Polymorphisms in Pepsinogen C and miRNA Genes Associate with High Serum Pepsinogen II in Gastric Cancer Patients. Microorganisms 2021; 9:microorganisms9010126. [PMID: 33430456 PMCID: PMC7827830 DOI: 10.3390/microorganisms9010126] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Accepted: 01/03/2021] [Indexed: 12/18/2022] Open
Abstract
Background: Pepsinogen (PG) II (PGII) is a serological marker used to estimate the risk of gastric cancer but how PGII expression is regulated is largely unknown. It has been suggested that PGII expression, from the PGC (Progastricsin) gene, is regulated by microRNAs (miRNA), but how PGII levels vary with Helicobacter pylori (H. pylori) infection and miRNAs genotype remains unclear. Methods: Serum levels of PGI and PGII were determined in 80 patients with gastric cancer and persons at risk for gastric cancer (74 first-degree relatives of patients, 62 patients with autoimmune chronic atrophic gastritis, and 2 patients with dysplasia), with and without H. pylori infection. As control from the general population, 52 blood donors were added to the analyses. Associations between PGII levels and genetic variants in PGC and miRNA genes in these groups were explored based on H. pylori seropositivity and the risk for gastric cancer. The two-dimensional difference in gel electrophoresis (2D-DIGE) and the NanoString analysis of messenger RNA (mRNAs) from gastric cancer tissue were used to determine the pathways associated with increased PGII levels. Results: PGII levels were significantly higher in patients with gastric cancer, and in those with H. pylori infection, than in other patients or controls. A PGI/PGII ratio ≤ 3 was found better than PGI < 25 ng/mL to identify patients with gastric cancer (15.0% vs. 8.8%). For two genetic variants, namely rs8111742 in miR-Let-7e and rs121224 in miR-365b, there were significant differences in PGII levels between genotype groups among patients with gastric cancer (p = 0.02 and p = 0.01, respectively), but not among other study subjects. Moreover, a strict relation between rs9471643 C-allele with H. pylori infection and gastric cancer was underlined. Fold change in gene expression of mRNA isolated from gastric cancer tissue correlated well with polymorphism, H. pylori infection, increased PGII level, and pathway for bacteria cell entry into the host. Conclusions: Serum PGII levels depend in part on an interaction between H. pylori and host miRNA genotypes, which may interfere with the cut-off of PGI/PGII ratio used to identify persons at risk of gastric cancer. Results reported new findings regarding the relation among H. pylori, PGII-related host polymorphism, and genes involved in this interaction in the gastric cancer setting.
Collapse
|
74
|
Zhang G, Zheng J, Zheng L, Yu S, Jiang C, Lin W, Li D, Qu L, Wang W. Gastric intestinal metaplasia assessment between linked color imaging based on endoscopy and pathology. Scand J Gastroenterol 2021; 56:103-110. [PMID: 33232631 DOI: 10.1080/00365521.2020.1849385] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
OBJECTIVE Cumulative evidence suggests that linked color imaging (LCI) can be used to identify gastric intestinal metaplasia (GIM). We aimed to develop endoscopic grading for GIM (EGGIM) with LCI. METHODS Two hundred and seventy-seven patients who underwent high-resolution white-light gastroscopy followed by LCI for EGGIM estimation were included. LCI was performed for the entire mucosa, and images of five areas each were recorded from the lesser and greater curvatures of the antrum and corpus, and for the incisura. For each area, scores of 0 (no GIM), 1 (focal GIM, ≤30% of the area), and 2 (extensive GIM, >30% of the area) were attributed for 10 points. If GIM was suspected based on endoscopy findings, targeted biopsies were performed; if GIM was not evident, random biopsies were performed according to the Sydney system to estimate the operative link on GIM (OLGIM). RESULTS GIM was staged as OLGIM 0, I, II, III, and IV in 136, 70, 37, 28, and 6 patients, respectively. For OLGIM III/IV diagnosis, the area under the receiver operating curve was 0.949 (95% CI 0.916-0.972). EGGIM of 4, with sensitivity and specificity of 94.12% (95% CI 80.3%-99.3%) and 86.42% (95% CI 81.5%-90.5%), respectively, was determined the best cut-off value for identifying OLGIM III/IV patients. CONCLUSIONS Our findings demonstrated the ability of EGGIM for diagnosing the extent of intestinal metaplasia and showed that EGGIM is related to OLGIM staging. EGGIM of 4 was the best cut-off value for identifying OLGIM III/IV patients.
Collapse
Affiliation(s)
- Guanpo Zhang
- Department of Gastroenterology, 900th Hospital of Joint Logistics Support Force, Fujian Medical University, Fuzhou, China.,Department of Gastroenterology, 900th Hospital of Joint Logistics Support Force, People's Liberation Army, Fuzhou, China
| | - Jin Zheng
- Department of Gastroenterology, 900th Hospital of Joint Logistics Support Force, People's Liberation Army, Fuzhou, China.,Department of Gastroenterology, Oriental Hospital Affiliated to Xiamen University, Fuzhou, China
| | - Linfu Zheng
- Department of Gastroenterology, 900th Hospital of Joint Logistics Support Force, Fujian Medical University, Fuzhou, China.,Department of Gastroenterology, 900th Hospital of Joint Logistics Support Force, People's Liberation Army, Fuzhou, China
| | - Shentong Yu
- Department of Pathology, 900th Hospital of Joint Logistics Support Force, People's Liberation Army, Fuzhou, China
| | - Chuanshen Jiang
- Department of Gastroenterology, 900th Hospital of Joint Logistics Support Force, Fujian Medical University, Fuzhou, China.,Department of Gastroenterology, 900th Hospital of Joint Logistics Support Force, People's Liberation Army, Fuzhou, China
| | - Wulian Lin
- Department of Gastroenterology, 900th Hospital of Joint Logistics Support Force, Fujian Medical University, Fuzhou, China.,Department of Gastroenterology, 900th Hospital of Joint Logistics Support Force, People's Liberation Army, Fuzhou, China
| | - Dazhou Li
- Department of Gastroenterology, 900th Hospital of Joint Logistics Support Force, Fujian Medical University, Fuzhou, China.,Department of Gastroenterology, 900th Hospital of Joint Logistics Support Force, People's Liberation Army, Fuzhou, China
| | - Lijuan Qu
- Department of Pathology, 900th Hospital of Joint Logistics Support Force, People's Liberation Army, Fuzhou, China
| | - Wen Wang
- Department of Gastroenterology, 900th Hospital of Joint Logistics Support Force, Fujian Medical University, Fuzhou, China.,Department of Gastroenterology, 900th Hospital of Joint Logistics Support Force, People's Liberation Army, Fuzhou, China
| |
Collapse
|
75
|
Accuracy of upper endoscopies with random biopsies to identify patients with gastric premalignant lesions who can safely be exempt from surveillance. Gastric Cancer 2021; 24:680-690. [PMID: 33616776 PMCID: PMC8065002 DOI: 10.1007/s10120-020-01149-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Accepted: 12/09/2020] [Indexed: 02/07/2023]
Abstract
INTRODUCTION Guidelines recommend endoscopy with biopsies to stratify patients with gastric premalignant lesions (GPL) to high and low progression risk. High-risk patients are recommended to undergo surveillance. We aimed to assess the accuracy of guideline recommendations to identify low-risk patients, who can safely be discharged from surveillance. METHODS This study includes patients with GPL. Patients underwent at least two endoscopies with an interval of 1-6 years. Patients were defined 'low risk' if they fulfilled requirements for discharge, and 'high risk' if they fulfilled requirements for surveillance, according to European guidelines (MAPS-2012, updated MAPS-2019, BSG). Patients defined 'low risk' with progression of disease during follow-up (FU) were considered 'misclassified' as low risk. RESULTS 334 patients (median age 60 years IQR11; 48.7% male) were included and followed for a median of 48 months. At baseline, 181/334 (54%) patients were defined low risk. Of these, 32.6% were 'misclassified', showing progression of disease during FU. If MAPS-2019 were followed, 169/334 (51%) patients were defined low risk, of which 32.5% were 'misclassified'. If BSG were followed, 174/334 (51%) patients were defined low risk, of which 32.2% were 'misclassified'. Seven patients developed gastric cancer (GC) or dysplasia, four patients were 'misclassified' based on MAPS-2012 and three on MAPS-2019 and BSG. By performing one additional endoscopy 72.9% (95% CI 62.4-83.3) of high-risk patients and all patients who developed GC or dysplasia were identified. CONCLUSION One-third of patients that would have been discharged from GC surveillance, appeared to be 'misclassified' as low risk. One additional endoscopy will reduce this risk by 70%.
Collapse
|
76
|
Liou JM, Malfertheiner P, Lee YC, Sheu BS, Sugano K, Cheng HC, Yeoh KG, Hsu PI, Goh KL, Mahachai V, Gotoda T, Chang WL, Chen MJ, Chiang TH, Chen CC, Wu CY, Leow AHR, Wu JY, Wu DC, Hong TC, Lu H, Yamaoka Y, Megraud F, Chan FKL, Sung JJ, Lin JT, Graham DY, Wu MS, El-Omar EM. Screening and eradication of Helicobacter pylori for gastric cancer prevention: the Taipei global consensus. Gut 2020; 69:2093-2112. [PMID: 33004546 DOI: 10.1136/gutjnl-2020-322368] [Citation(s) in RCA: 262] [Impact Index Per Article: 52.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2020] [Revised: 07/27/2020] [Accepted: 08/12/2020] [Indexed: 02/06/2023]
Abstract
OBJECTIVE A global consensus meeting was held to review current evidence and knowledge gaps and propose collaborative studies on population-wide screening and eradication of Helicobacter pylori for prevention of gastric cancer (GC). METHODS 28 experts from 11 countries reviewed the evidence and modified the statements using the Delphi method, with consensus level predefined as ≥80% of agreement on each statement. The Grading of Recommendation Assessment, Development and Evaluation (GRADE) approach was followed. RESULTS Consensus was reached in 26 statements. At an individual level, eradication of H. pylori reduces the risk of GC in asymptomatic subjects and is recommended unless there are competing considerations. In cohorts of vulnerable subjects (eg, first-degree relatives of patients with GC), a screen-and-treat strategy is also beneficial. H. pylori eradication in patients with early GC after curative endoscopic resection reduces the risk of metachronous cancer and calls for a re-examination on the hypothesis of 'the point of no return'. At the general population level, the strategy of screen-and-treat for H. pylori infection is most cost-effective in young adults in regions with a high incidence of GC and is recommended preferably before the development of atrophic gastritis and intestinal metaplasia. However, such a strategy may still be effective in people aged over 50, and may be integrated or included into national healthcare priorities, such as colorectal cancer screening programmes, to optimise the resources. Reliable locally effective regimens based on the principles of antibiotic stewardship are recommended. Subjects at higher risk of GC, such as those with advanced gastric atrophy or intestinal metaplasia, should receive surveillance endoscopy after eradication of H. pylori. CONCLUSION Evidence supports the proposal that eradication therapy should be offered to all individuals infected with H. pylori. Vulnerable subjects should be tested, and treated if the test is positive. Mass screening and eradication of H. pylori should be considered in populations at higher risk of GC.
Collapse
Affiliation(s)
- Jyh-Ming Liou
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan.,Department of Internal Medicine, National Taiwan University College of Medicine, Taipei, Taiwan.,Department of Medicine, National Taiwan University Cancer Center, Taipei, Taiwan
| | - Peter Malfertheiner
- Department of Gastroenterology, Hepatology and Infectious Diseases, Otto-von-Guericke University Magdeburg, Magdeburg, Germany.,Department of Medicine II, University Hospital, LMU Munich, Munich, Germany
| | - Yi-Chia Lee
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan.,Department of Internal Medicine, National Taiwan University College of Medicine, Taipei, Taiwan.,Department of Medical Research, National Taiwan University Hospital, Taipei, Taiwan
| | - Bor-Shyang Sheu
- Department of Internal Medicine and Institute of Clinical Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan.,Division of Gastroenterology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
| | - Kentaro Sugano
- Department of Medicine, Jichi Medical School, Tochigi, Japan
| | - Hsiu-Chi Cheng
- Department of Internal Medicine and Institute of Clinical Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan.,Department of Internal Medicine, Tainan Hospital, Ministry of Health and Welfare, Tainan, Taiwan
| | - Khay-Guan Yeoh
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Ping-I Hsu
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, An Nan Hospital, China Medical University, Tainan, Taiwan
| | - Khean-Lee Goh
- Department of Gastroenterology and Hepatology, University of Malaya, Kuala Lumpur, Malaysia
| | - Varocha Mahachai
- Department of Medicine, Faculty of Medicine, Chulalongkorn University and King Chulalongkorn Memorial Hospital, Bangkok, Thailand
| | - Takuji Gotoda
- Division of Gastroenterology and Hepatology, Department of Medicine, Nihon University School of Medicine, Tokyo, Japan
| | - Wei-Lun Chang
- Department of Internal Medicine and Institute of Clinical Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Mei-Jyh Chen
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan.,Department of Internal Medicine, National Taiwan University College of Medicine, Taipei, Taiwan.,Integrated Diagnostics and Therapeutics, National Taiwan University Hospital, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Tsung-Hsien Chiang
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan.,Department of Internal Medicine, National Taiwan University College of Medicine, Taipei, Taiwan.,Integrated Diagnostics and Therapeutics, National Taiwan University Hospital, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Chieh-Chang Chen
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan.,Department of Internal Medicine, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Chun-Ying Wu
- Institute of Biomedical Informatics, School of Medicine, National Yang-Ming University, Taipei, Taiwan.,Division of Translational Research, Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Alex Hwong-Ruey Leow
- Department of Gastroenterology and Hepatology, University of Malaya, Kuala Lumpur, Malaysia
| | - Jeng-Yih Wu
- Division of Gastroenterology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
| | - Deng-Chyang Wu
- Division of Gastroenterology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
| | - Tzu-Chan Hong
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan.,Department of Internal Medicine, National Taiwan University College of Medicine, Taipei, Taiwan.,Department of Internal Medicine, National Taiwan University Hospital Bei-Hu Branch, Taipei, Taiwan
| | - Hong Lu
- Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Shanghai Institute of Digestive Disease, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yoshio Yamaoka
- Oita University Faculty of Medicine, Yufu, Oita, Japan.,Department of Medicine, Michael E DeBakey VA Medical Center and Baylor College of Medicine, Houston, Texas, USA
| | - Francis Megraud
- French National Reference Centre for Helicobacters, Bacteriology laboratory, Pellegrin Hospital, Bordeaux, & INSERM U1053, University of Bordeaux, Bordeaux, France
| | - Francis K L Chan
- Institute of Digestive Disease, Chinese University of Hong Kong, Shatin, Hong Kong, China.,Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Shatin, Hong Kong, China
| | - Joseph Jy Sung
- Institute of Digestive Disease, Chinese University of Hong Kong, Shatin, Hong Kong, China.,Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Shatin, Hong Kong, China
| | - Jaw-Town Lin
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan.,Digestive Medicine Center, China Medical University Hospital, Taichung, Taiwan
| | - David Y Graham
- Department of Medicine, Michael E DeBakey VA Medical Center and Baylor College of Medicine, Houston, Texas, USA
| | - Ming-Shiang Wu
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan .,Department of Internal Medicine, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Emad M El-Omar
- Department of Medicine, University of New South Wales, Sydney, New South Wales, Australia.,Microbiome Research Centre, St George & Sutherland Clinical School, University of New South Wales, Sydney, NSW, Australia
| | | |
Collapse
|
77
|
Chapelle N, Jirka I, Péron M, Quénéhervé L, Cauchin E, Touchefeu Y, Coron E, Mosnier JF, Matysiak-Budnik T. Evaluation of a Phone Call Reminder Strategy in the Surveillance of Patients with Gastric Precancerous Lesions Lost to Follow-Up. Gastrointest Tumors 2020; 7:110-116. [PMID: 33173774 DOI: 10.1159/000508873] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Accepted: 05/22/2020] [Indexed: 12/24/2022] Open
Abstract
Background Surveillance of gastric precancerous lesions (GPL) may reduce gastric cancer (GC)-related mortality, but some patients with GPL are lost to follow-up. Objective The aim of this study was to evaluate the feasibility and efficacy of a "phone-call" strategy in surveillance of the lost to follow-up patients. Patients and Methods Among all the patients diagnosed with GPL (atrophic gastritis, intestinal metaplasia, low-grade dysplasia) between 2000 and 2015, we identified those who should undergo surveillance endoscopy according to the current guidelines. They were contacted by telephone and invited to undergo endoscopy with gastric biopsies for histological analysis. Results Among 535 patients with GPL, 134 were contacted. Sixty-two (46%) could not be joined, 36 did not have endoscopy for other reasons, and finally, 36 patients (22 males, median age 65 years) were included. After the median time interval of 57 months between 2 endoscopies, 18 patients showed stability, 11 regression, and 7 progression of GPL, including 1 patient who developed GC. Conclusion Despite several telephone calls, only one-third of the contacted patients could be brought to surveillance endoscopy. Most of the patients showed stability of GPL, but 1 progressed to GC and could be successfully treated.
Collapse
Affiliation(s)
- Nicolas Chapelle
- CHU de Nantes, Hôtel Dieu, Institut des Maladies de l'Appareil Digestif, Nantes, France.,Université de Nantes, Nantes, France.,INSERM, U1235, Nantes, France
| | - Iva Jirka
- CHU de Nantes, Hôtel Dieu, Institut des Maladies de l'Appareil Digestif, Nantes, France
| | - Matthieu Péron
- CHU de Nantes, Hôtel Dieu, Institut des Maladies de l'Appareil Digestif, Nantes, France.,Université de Nantes, Nantes, France
| | - Lucille Quénéhervé
- CHU de Nantes, Hôtel Dieu, Institut des Maladies de l'Appareil Digestif, Nantes, France.,Université de Nantes, Nantes, France.,INSERM, U1235, Nantes, France
| | - Estelle Cauchin
- CHU de Nantes, Hôtel Dieu, Institut des Maladies de l'Appareil Digestif, Nantes, France.,INSERM, U1235, Nantes, France
| | - Yann Touchefeu
- CHU de Nantes, Hôtel Dieu, Institut des Maladies de l'Appareil Digestif, Nantes, France.,INSERM, U1235, Nantes, France
| | - Emmanuel Coron
- CHU de Nantes, Hôtel Dieu, Institut des Maladies de l'Appareil Digestif, Nantes, France.,Université de Nantes, Nantes, France.,INSERM, U1235, Nantes, France
| | - Jean-François Mosnier
- Université de Nantes, Nantes, France.,Service d'Anatomie Pathologique, CHU de Nantes, Nantes, France
| | - Tamara Matysiak-Budnik
- CHU de Nantes, Hôtel Dieu, Institut des Maladies de l'Appareil Digestif, Nantes, France.,Université de Nantes, Nantes, France.,INSERM, U1235, Nantes, France
| |
Collapse
|
78
|
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: 21] [Impact Index Per Article: 4.2] [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
|
79
|
Bornschein J, Tran-Nguyen T, Fernandez-Esparrach G, Ash S, Balaguer F, Bird-Lieberman EL, Córdova H, Dzerve Z, Fassan M, Leja M, Lyutakov I, Middelburg T, Moreira L, Nakov R, Nieuwenburg SAV, O'Connor A, Realdon S, De Schepper H, Smet A, Spaander MCW, Tolmanis I, Urbonas T, Weigt J, Hold GL, Link A, Kupcinskas J. Biopsy Sampling in Upper Gastrointestinal Endoscopy: A Survey from 10 Tertiary Referral Centres Across Europe. Dig Dis 2020; 39:179-189. [PMID: 33002891 PMCID: PMC8220928 DOI: 10.1159/000511867] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Accepted: 09/26/2020] [Indexed: 02/02/2023]
Abstract
BACKGROUND Guidelines give robust recommendations on which biopsies should be taken when there is endoscopic suggestion of gastric inflammation. Adherence to these guidelines often seems arbitrary. This study aimed to give an overview on current practice in tertiary referral centres across Europe. METHODS Data were collected at 10 tertiary referral centres. Demographic data, the indication for each procedure, endoscopic findings, and the number and sampling site of biopsies were recorded. Findings were compared between centres, and factors influencing the decision to take biopsies were explored. RESULTS Biopsies were taken in 56.6% of 9,425 procedures, with significant variation between centres (p < 0.001). Gastric biopsies were taken in 43.8% of all procedures. Sampling location varied with the procedure indication (p < 0.001) without consistent pattern across the centres. Fewer biopsies were taken in centres which routinely applied the updated Sydney classification for gastritis assessment (46.0%), compared to centres where this was done only upon request (75.3%, p < 0.001). This was the same for centres stratifying patients according to the OLGA system (51.8 vs. 73.0%, p < 0.001). More biopsies were taken in centres following the MAPS guidelines on stomach surveillance (68.1 vs. 37.1%, p < 0.001). Biopsy sampling was more likely in younger patients in 8 centres (p < 0.05), but this was not true for the whole cohort (p = 0.537). The percentage of procedures with biopsies correlated directly with additional costs charged in case of biopsies (r = 0.709, p = 0.022). CONCLUSION Adherence to guideline recommendations for biopsy sampling at gastroscopy was inconsistent across the participating centres. Our data suggest that centre-specific policies are applied instead.
Collapse
Affiliation(s)
- Jan Bornschein
- Translational Gastroenterology Unit, John Radcliffe Hospital, Oxford University Hospitals, Headington, Oxford, United Kingdom
- NIHR Oxford Biomedical Research Centre, Oxford, United Kingdom
- Department of Gastroenterology, Hepatology and Infectious Diseases, Otto-von-Guericke University, Magdeburg, Germany
| | - Terry Tran-Nguyen
- Translational Gastroenterology Unit, John Radcliffe Hospital, Oxford University Hospitals, Headington, Oxford, United Kingdom
- NIHR Oxford Biomedical Research Centre, Oxford, United Kingdom
| | - Gloria Fernandez-Esparrach
- Gastroenterology Department, Hospital Clinic of Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of Barcelona, Centro de Investigación Biomédica en Red en Enfermedades Hepáticas y Digestivas (CIBEREHD), Barcelona, Spain
| | - Stephen Ash
- Nuffield Department of Medicine, Ludwig Institute for Cancer Research, University of Oxford, Oxford, United Kingdom
| | - Francesc Balaguer
- Gastroenterology Department, Hospital Clinic of Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of Barcelona, Centro de Investigación Biomédica en Red en Enfermedades Hepáticas y Digestivas (CIBEREHD), Barcelona, Spain
| | - Elisabeth L. Bird-Lieberman
- Translational Gastroenterology Unit, John Radcliffe Hospital, Oxford University Hospitals, Headington, Oxford, United Kingdom
- NIHR Oxford Biomedical Research Centre, Oxford, United Kingdom
| | - Henry Córdova
- Gastroenterology Department, Hospital Clinic of Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of Barcelona, Centro de Investigación Biomédica en Red en Enfermedades Hepáticas y Digestivas (CIBEREHD), Barcelona, Spain
| | - Zane Dzerve
- Digestive Diseases Centre GASTRO, Institute of Clinical and Preventive Medicine, University of Latvia, Riga, Latvia
| | - Matteo Fassan
- Department of Medicine (DIMED), Surgical Pathology Unit, University of Padua, Padua, Italy
| | - Marcis Leja
- Digestive Diseases Centre GASTRO, Institute of Clinical and Preventive Medicine, University of Latvia, Riga, Latvia
| | - Ivan Lyutakov
- Department of Gastroenterology, Tsaritsa Yoanna University Hospital, Medical University of Sofia, Sofia, Bulgaria
| | - Tim Middelburg
- Department of Gastroenterology & Hepatology, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Leticia Moreira
- Gastroenterology Department, Hospital Clinic of Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of Barcelona, Centro de Investigación Biomédica en Red en Enfermedades Hepáticas y Digestivas (CIBEREHD), Barcelona, Spain
| | - Radislav Nakov
- Department of Gastroenterology, Tsaritsa Yoanna University Hospital, Medical University of Sofia, Sofia, Bulgaria
| | - Stella A. V. Nieuwenburg
- Department of Gastroenterology & Hepatology, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Anthony O'Connor
- Department of Gastroenterology, Tallaght University Hospital, University of Dublin, Trinity College, Dublin, Ireland
| | - Stefano Realdon
- Endoscopy Unit, Veneto Institute of Oncology IOV-IRCCS, Padua, Italy
| | - Heiko De Schepper
- Department of Gastroenterology & Hepatology, University Hospital Antwerp, Edegem, Belgium
| | - Annemieke Smet
- Laboratory of Experimental Medicine and Pediatrics, Faculty of Medicine and Health Sciences, University of Antwerp, Wilrijk, Belgium
| | - M. C. W. Spaander
- Department of Gastroenterology & Hepatology, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Ivars Tolmanis
- Digestive Diseases Centre GASTRO, Institute of Clinical and Preventive Medicine, University of Latvia, Riga, Latvia
| | - Tadas Urbonas
- Department of Gastroenterology and Institute for Digestive Research, Lithuanian University of Health Sciences, Kaunas, Lithuania
| | - Jochen Weigt
- Department of Gastroenterology, Hepatology and Infectious Diseases, Otto-von-Guericke University, Magdeburg, Germany
| | - Georgina L. Hold
- Microbiome Research Centre, St George & Sutherland Clinical School, St George Hospital, University of New South Wales, Sydney, New South Wales, Australia
| | - Alexander Link
- Department of Gastroenterology, Hepatology and Infectious Diseases, Otto-von-Guericke University, Magdeburg, Germany
| | - Juozas Kupcinskas
- Department of Gastroenterology and Institute for Digestive Research, Lithuanian University of Health Sciences, Kaunas, Lithuania
| |
Collapse
|
80
|
Marcos P, Brito-Gonçalves G, Libânio D, Pita I, Castro R, Sá I, Dinis-Ribeiro M, Pimentel-Nunes P. Endoscopic grading of gastric intestinal metaplasia on risk assessment for early gastric neoplasia: can we replace histology assessment also in the West? Gut 2020; 69:1762-1768. [PMID: 32051208 DOI: 10.1136/gutjnl-2019-320091] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Revised: 01/09/2020] [Accepted: 01/26/2020] [Indexed: 12/24/2022]
Abstract
OBJECTIVES To assess the value of endoscopic grading of gastric intestinal metaplasia (EGGIM), operative link on gastritis assessment (OLGA) and operative link on gastric intestinal metaplasia (OLGIM) on risk stratification for early gastric neoplasia (EGN) and to investigate other factors possibly associated with its development. DESIGN Single centre, case-control study including 187 patients with EGN treated endoscopically and 187 age-matched and sex-matched control subjects. Individuals were classified according to EGGIM, OLGA and OLGIM systems. EGN risk according to gastritis stages and other clinical parameters was further evaluated. RESULTS More patients with EGN had EGGIM of ≥5 than control subjects (68.6% vs 13.3%, p<0.001). OLGA and OLGIM stages III/IV were more prevalent in patients with EGN than in control subjects (68% vs 11%, p<0.001, and 61% vs 3%, p<0.001, respectively). The three systems were the only parameters significantly related to the risk of EGN in multivariate analysis: for EGGIM 1-4 (adjusted OR (AOR) 12.9, 95% CI 1.4 to 118.6) and EGGIM 5-10 (AOR 21.2, 95% CI 5.0 to 90.2); for OLGA I/II (AOR 5.0, 95% CI 0.56 to 44.5) and OLGA III/IV (AOR 11.1, 95% CI 3.7 to 33.1); for OLGIM I/II (AOR 11.5, 95% CI 4.1 to 32.3) and OLGIM III/IV (AOR 16.0, 95% CI 7.6 to 33.4). CONCLUSION This study confirms the role of histological assessment as an independent risk factor for gastric cancer (GC), but it is the first study to show that an endoscopic classification of gastric intestinal metaplasia is highly associated with that outcome. After further prospective validation, this classification may be appropriate for GC risk stratification and may simplify every day practice by reducing the need for biopsies.
Collapse
Affiliation(s)
- Pedro Marcos
- Gastroenterology Department, Portuguese Oncology Institute of Porto, Porto, Portugal .,Gastroenterology Department, Centro Hospitalar de Leiria, Leiria, Portugal
| | | | - Diogo Libânio
- Gastroenterology Department, Portuguese Oncology Institute of Porto, Porto, Portugal.,MEDCIS, Faculty of Medicine, University of Porto, Porto, Portugal
| | - Inês Pita
- Gastroenterology Department, Portuguese Oncology Institute of Porto, Porto, Portugal
| | - Rui Castro
- Gastroenterology Department, Portuguese Oncology Institute of Porto, Porto, Portugal
| | - Inês Sá
- Gastroenterology Department, Portuguese Oncology Institute of Porto, Porto, Portugal
| | - Mário Dinis-Ribeiro
- Gastroenterology Department, Portuguese Oncology Institute of Porto, Porto, Portugal.,MEDCIS, Faculty of Medicine, University of Porto, Porto, Portugal
| | - Pedro Pimentel-Nunes
- Gastroenterology Department, Portuguese Oncology Institute of Porto, Porto, Portugal.,MEDCIS, Faculty of Medicine, University of Porto, Porto, Portugal.,Surgery and Physiology Department, Faculty of Medicine, University of Porto, Porto, Portugal
| |
Collapse
|
81
|
Chapelle N, Petryszyn P, Blin J, Leroy M, Le Berre-Scoul C, Jirka I, Neunlist M, Moussata D, Lamarque D, Olivier R, Tougeron D, Mosnier JF, Matysiak-Budnik T. A panel of stomach-specific biomarkers (GastroPanel®) for the diagnosis of atrophic gastritis: A prospective, multicenter study in a low gastric cancer incidence area. Helicobacter 2020; 25:e12727. [PMID: 32700438 DOI: 10.1111/hel.12727] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Revised: 06/15/2020] [Accepted: 06/17/2020] [Indexed: 12/12/2022]
Abstract
BACKGROUND Analysis of serum biomarkers for the assessment of atrophic gastritis (AG), considered as gastric precancerous lesion, is of growing interest and recommended by current guidelines. Our aim was to evaluate the diagnostic performance of a panel of biomarkers (GastroPanel®) for the detection of AG in France, a country of a low gastric cancer (GC) incidence. MATERIAL AND METHODS In this prospective, multicenter, cross-sectional study, consecutive patients considered at increased risk of GC and undergoing upper endoscopy with gastric biopsies were included. Blood samples were collected for the analysis of GastroPanel® (association of Pepsinogens I and II, Gastrin-17, and Helicobacter pylori serology) using ELISA. The results of GastroPanel® were compared to the results of histology considered as the reference. RESULTS Between 2016 and 2019, 344 patients (148 cases with AG, 196 controls without AG) were included. Sensitivity, specificity, positive, and negative predictive values for the detection of AG by GastroPanel® were of 39.9% (95% CI 31.9; 48.2), 93.4% (95% CI 88.9; 96.4), 81.9 (95% CI 71.1; 90.0), and 67.3 (95% CI 61.4; 72.8), respectively. The sensitivity was significantly higher for the detection of severe AG [60.8% (95% CI 46.1; 74.6) P = .015] and corpus AG [61.0% (95% CI 49.2; 72.0), P = .004]. Diagnostic performances of GastroPanel® tended to be better than those of Pepsinogen I alone, but the difference did not reach statistical significance (P = .068). CONCLUSION Serum pepsinogen and GastroPanel® tests show promising results for the detection of AG, especially of corpus AG and severe AG, in patients at high risk of GC in France.
Collapse
Affiliation(s)
- Nicolas Chapelle
- IMAD, Hepato-Gastroenterology & Digestive Oncology Unit, University Hospital of Nantes, Nantes, France.,University of Nantes, Nantes, France.,INSERM UMR1235, The Enteric Nervous System in Gut and Brain Disorders, Nantes, France
| | - Pawel Petryszyn
- Department of Clinical Pharmacology, Wroclaw Medical University, Wroclaw, Poland
| | - Justine Blin
- University of Nantes, Nantes, France.,INSERM UMR1235, The Enteric Nervous System in Gut and Brain Disorders, Nantes, France.,Department of Biochemistry, University Hospital of Nantes, Nantes, France
| | - Maxime Leroy
- Department of Biostatistics, University Hospital of Nantes, Nantes, France
| | - Catherine Le Berre-Scoul
- University of Nantes, Nantes, France.,INSERM UMR1235, The Enteric Nervous System in Gut and Brain Disorders, Nantes, France
| | - Iva Jirka
- IMAD, Hepato-Gastroenterology & Digestive Oncology Unit, University Hospital of Nantes, Nantes, France
| | - Michel Neunlist
- University of Nantes, Nantes, France.,INSERM UMR1235, The Enteric Nervous System in Gut and Brain Disorders, Nantes, France
| | - Driffa Moussata
- Department of Hepato-Gastroenterology, University Hospital of Tours, Tours, France
| | - Dominique Lamarque
- Department of Hepato-Gastroenterology, Ambroise-Paré Hospital, AP-HP, Paris Saclay University, UVSQ, INSERM, Infection and Inflammation, Paris, France
| | - Raphael Olivier
- IMAD, Hepato-Gastroenterology & Digestive Oncology Unit, University Hospital of Nantes, Nantes, France.,Department of Hepato-Gastroenterology, Poitiers University Hospital and University of Poitiers, Poitiers, France
| | - David Tougeron
- Department of Hepato-Gastroenterology, Poitiers University Hospital and University of Poitiers, Poitiers, France
| | - Jean-François Mosnier
- University of Nantes, Nantes, France.,Department of Pathology, University Hospital of Nantes, Nantes, France
| | - Tamara Matysiak-Budnik
- IMAD, Hepato-Gastroenterology & Digestive Oncology Unit, University Hospital of Nantes, Nantes, France.,University of Nantes, Nantes, France.,INSERM UMR1235, The Enteric Nervous System in Gut and Brain Disorders, Nantes, France
| |
Collapse
|
82
|
Waddingham W, Nieuwenburg SAV, Carlson S, Rodriguez-Justo M, Spaander M, Kuipers EJ, Jansen M, Graham DG, Banks M. Recent advances in the detection and management of early gastric cancer and its precursors. Frontline Gastroenterol 2020; 12:322-331. [PMID: 34249318 PMCID: PMC8223672 DOI: 10.1136/flgastro-2018-101089] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Revised: 04/02/2020] [Accepted: 04/13/2020] [Indexed: 02/06/2023] Open
Abstract
Despite declines in incidence, gastric cancer remains a disease with a poor prognosis and limited treatment options due to its often late stage of diagnosis. In contrast, early gastric cancer has a good to excellent prognosis, with 5-year survival rates as high as 92.6% after endoscopic resection. There remains an East-West divide for this disease, with high incidence countries such as Japan seeing earlier diagnoses and reduced mortality, in part thanks to the success of a national screening programme. With missed cancers still prevalent at upper endoscopy in the West, and variable approaches to assessment of the high-risk stomach, the quality of endoscopy we provide must be a focus for improvement, with particular attention paid to the minority of patients at increased cancer risk. High-definition endoscopy with virtual chromoendoscopy is superior to white light endoscopy alone. These enhanced imaging modalities allow the experienced endoscopist to accurately and robustly detect high-risk lesions in the stomach. An endoscopy-led staging strategy would mean biopsies could be targeted to histologically confirm the endoscopic impression of premalignant lesions including atrophic gastritis, gastric intestinal metaplasia, dysplasia and early cancer. This approach to quality improvement will reduce missed diagnoses and, combined with the latest endoscopic resection techniques performed at expert centres, will improve early detection and ultimately patient outcomes. In this review, we outline the latest evidence relating to diagnosis, staging and treatment of early gastric cancer and its precursor lesions.
Collapse
Affiliation(s)
- William Waddingham
- Gastroenterology, University College London Hospitals NHS Foundation Trust, London, UK,Research Department of Pathology, UCL Cancer Institute, London, UK
| | - Stella A V Nieuwenburg
- Gastroenterology and Hepatology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Sean Carlson
- Gastroenterology, University College London Hospitals NHS Foundation Trust, London, UK
| | | | - Manon Spaander
- Gastroenterology and Hepatology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Ernst J Kuipers
- Gastroenterology and Hepatology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Marnix Jansen
- Research Department of Pathology, UCL Cancer Institute, London, UK,Pathology, University College London Hospitals NHS Foundation Trust, London, UK
| | - David G Graham
- Gastroenterology, University College London Hospitals NHS Foundation Trust, London, UK
| | - Matthew Banks
- Gastroenterology, University College London Hospitals NHS Foundation Trust, London, UK
| |
Collapse
|
83
|
Gastritis Stages in Monozygotic and Dizygotic Dyspeptic Twins. Gastroenterol Res Pract 2020; 2020:9510717. [PMID: 32714389 PMCID: PMC7352144 DOI: 10.1155/2020/9510717] [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: 02/22/2020] [Revised: 05/30/2020] [Accepted: 06/01/2020] [Indexed: 02/08/2023] Open
Abstract
Background The progression of Helicobacter pylori-associated gastritis towards atrophic gastritis is modulated by host-related and environmental factors. Studies that explore the possible involvement of host-related versus environmental factors in the development of gastritis phenotype induced by H. pylori are highly needed. Aims Our study was aimed at investigating the phenotype of H. pylori-associated gastritis in two cohorts of monozygotic and dizygotic twins, using the OLGA/OLGIM gastritis staging system. Methods Two cohorts of monozygotic (14 pairs) and dizygotic (15 pairs) dyspeptic twins prospectively underwent endoscopy with biopsy sampling based on Sydney protocol. H. pylori status and OLGA/OLGIM stages were assessed and compared. Results The mean age of monozygotic and dizygotic twins was 40.4 and 38.6 years, respectively (p = 0.623). The overall prevalence of H. pylori infection was 51.7%. Among the 14 monozygotic twin pairs, five pairs were H. pylori-positive, four were H. pylori-negative, and five were H. pylori-discordant. Among the 15 dizygotic twin pairs, five pairs were H. pylori-positive, five were H. pylori-negative, and five were H. pylori-discordant. Concordance for antrum atrophy in monozygotic twins was 78.6% (11/14 pairs) and in dizygotic twins 73.3% (11/15 pairs) (p = 0.742). Concordance for corpus atrophy in monozygotic versus dizygotic twins was 92.9% (13/14 pairs) and 86.7% (13/15 pairs), respectively (p = 0.584). Concordance for antrum intestinal metaplasia (IM) in monozygotic twins was 85.7% (12/14 pairs) and in dizygotic 73.3% (11/15 pairs) (p = 0.411). Concordance for corpus IM in monozygotic twins was 85.7% (12/14 pairs) and in dizygotic 86.7% (13/15 pairs) (p = 0.941). Among monozygotic and dizygotic subjects, the stage of gastritis was concordant in both H. pylori-positive and H. pylori-negative subjects. Conclusions In conclusion, histological gastric mucosa alterations in monozygotic and dizygotic twins showed high rates of concordance. Furthermore, OLGA/OLGIM gastritis stages were not modulated by the zygosity of the twins.
Collapse
|
84
|
Cho JH, Jin SY. Optimized diagnosis of Helicobacter pylori and tailored eradication therapy for preventing gastric cancer: a proposal for SHAKE strategy. Expert Rev Gastroenterol Hepatol 2020; 14:553-564. [PMID: 32410515 DOI: 10.1080/17474124.2020.1770594] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
INTRODUCTION To decrease gastric cancer-related mortality, the Korean National Cancer Screening Program provides biennial screening gastroscopy to all individuals aged >40 years. However, a test-and-treat strategy of Helicobacter pylori for preventing gastric cancer has not been established. AREAS COVERED In this review, we present up-to-date results of endoscopic findings of H. pylori gastritis, optimal sites for H. pylori detection, gastric cancer risk assessment using serum pepsinogen, tailored eradication based on the antimicrobial resistance against H. pylori, and post-eradication surveillance. EXPERT OPINION Here we propose approaches to H. pylori diagnosis and treatment for preventing gastric cancer, termed 'Screening for H. pylori in Korea and Eradication (SHAKE)' strategy. This strategy consists of the following: (1) optimized H. pylori diagnosis, (2) individualized management based on the H. pylori infection status, and (3) tailored eradication therapy. H. pylori gastritis can be diagnosed by endoscopic observation of the gastric mucosal pattern at the greater curvature of the corpus. Measurement of the serum pepsinogen I/II ratio is useful for assessing the risk of gastric cancer. As a first-line treatment, tailored eradication based on the results of molecular testing is effective in a country with a high rate of clarithromycin-resistant H. pylori.
Collapse
Affiliation(s)
- Jun-Hyung Cho
- Digestive Disease Center, Soonchunhyang University Hospital , Seoul, Korea
| | - So-Young Jin
- Department of Pathology, Soonchunhyang University Hospital , Seoul, Korea
| |
Collapse
|
85
|
Alberts CJ, Jeske R, de Martel C, den Hollander WJ, Michel A, Prins M, Snijder MB, Schim van der Loeff MF, Waterboer T. Helicobacter pylori seroprevalence in six different ethnic groups living in Amsterdam: The HELIUS study. Helicobacter 2020; 25:e12687. [PMID: 32147867 DOI: 10.1111/hel.12687] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 01/20/2020] [Accepted: 02/10/2020] [Indexed: 12/30/2022]
Abstract
BACKGROUND Helicobacter pylori prevalence varies greatly worldwide. We explored the prevalence of H. pylori and CagA seropositivity among adults aged 18-44 years living in the Netherlands by ethnicity and migration status (first vs second generation). MATERIALS AND METHODS Participants from six different ethnic groups were selected from the population-based multi-ethnic HELIUS study in Amsterdam, the Netherlands. Serum samples were tested for H. pylori antigens using a validated Luminex-based multiplex serology assay. Prevalence ratios were estimated using Poisson regression analysis. RESULTS A total of 4683 participants aged 18-44 years were randomly selected based on sex, ethnicity, and age. H. pylori seroprevalence was highest in the Ghanaian group (84%), followed by Moroccan (81%), Turkish (66%), African Surinamese (51%), South-Asian Surinamese (48%), and Dutch (17%) participants. All ethnic minority groups had a significantly higher risk of being H. pylori seropositive compared to the Dutch group. This association was strongest among participants born outside the Netherlands (first generation), but was still significant and apparent among second-generation participants. Among first-generation participants, all groups, except the Moroccans, had a significantly higher proportion of individuals with a cagA + H. pylori strain compared to the Dutch participants. CONCLUSION Helicobacter pylori seroprevalence among first-generation migrants is high in the Netherlands and remains elevated among second-generation migrants (ie, those born in the Netherlands). High exposure to H. pylori, and especially to the more virulent cagA+ strain, highlights the need for tailored prevention of gastric diseases (notably peptic ulcers and cancers) among migrants.
Collapse
Affiliation(s)
- Catharina J Alberts
- International Agency for Research on Cancer, Lyon, France
- Department of Infectious Diseases, Public Health Service (GGD) of Amsterdam, Amsterdam, the Netherlands
| | - Rima Jeske
- Infection, Inflammation and Cancer Program, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
| | | | - Wouter J den Hollander
- Department of Gastroenterology and Hepatology, Leiden University Medical Center (LUMC), Leiden, the Netherlands
| | - Angelika Michel
- Infection, Inflammation and Cancer Program, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Maria Prins
- Department of Infectious Diseases, Public Health Service (GGD) of Amsterdam, Amsterdam, the Netherlands
- Department of Internal Medicine, Amsterdam Infection & Immunity Institute (AIII), Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Marieke B Snijder
- Department of Public Health, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Maarten F Schim van der Loeff
- Department of Infectious Diseases, Public Health Service (GGD) of Amsterdam, Amsterdam, the Netherlands
- Department of Internal Medicine, Amsterdam Infection & Immunity Institute (AIII), Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Tim Waterboer
- Infection, Inflammation and Cancer Program, German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
86
|
Gawron AJ, Shah SC, Altayar O, Davitkov P, Douglas M, Kevin T, Mustafa RA. AGA Technical Review on Gastric Intestinal Metaplasia-Natural History and Clinical Outcomes. Gastroenterology 2020; 158:705-731.e5. [PMID: 31816300 PMCID: PMC7375032 DOI: 10.1053/j.gastro.2019.12.001] [Citation(s) in RCA: 94] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Andrew J. Gawron
- Salt Lake City Specialty Care Center of Innovation & Gastroenterology Section, VA Salt Lake City Health Care System, Salt Lake City, Utah, USA,Division of Gastroenterology and Hepatology, Department of Internal Medicine, University of Utah School of Medicine, Salt Lake City, Utah, USA
| | - Shailja C. Shah
- Division of Gastroenterology, Hepatology, and Nutrition, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Osama Altayar
- Division of Gastroenterology, Washington University School of Medicine, St Louis, MO
| | - Perica Davitkov
- VA Northeast Ohio Healthcare System,Case Western Reserve University, Cleveland, OH, USA
| | - Morgan Douglas
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Turner Kevin
- University of Texas Southwestern College of Medicine, Dallas, TX, USA.,Inform Diagnostics Research Institute, Irving, TX, USA
| | - Reem A. Mustafa
- Division of Nephrology and Hypertension, University of Kansas Medical Center, Kansas City, KS, USA
| |
Collapse
|
87
|
Altayar O, Davitkov P, Shah SC, Gawron AJ, Morgan D, Turner K, Mustafa RA. AGA Technical Review on Gastric Intestinal Metaplasia-Epidemiology and Risk Factors. Gastroenterology 2020; 158:732-744.e16. [PMID: 31816301 PMCID: PMC7425600 DOI: 10.1053/j.gastro.2019.12.002] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Osama Altayar
- Division of Gastroenterology, Washington University School of Medicine, St Louis, MO
| | - Perica Davitkov
- VA Northeast Ohio Healthcare System,Case Western Reserve University, Cleveland, OH, USA
| | - Shailja C. Shah
- Division of Gastroenterology, Hepatology, and Nutrition, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Andrew J. Gawron
- Salt Lake City Specialty Care Center of Innovation & Gastroenterology Section, VA Salt Lake City Health Care System, Salt Lake City, Utah, USA,Division of Gastroenterology and Hepatology, Department of Internal Medicine, University of Utah School of Medicine, Salt Lake City, Utah, USA
| | - Douglas Morgan
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Kevin Turner
- University of Texas Southwestern College of Medicine, Dallas, TX, USA.,Inform Diagnostics Research Institute, Irving, TX, USA
| | - Reem A. Mustafa
- Division of Nephrology and Hypertension, University of Kansas Medical Center, Kansas City, KS, USA
| |
Collapse
|
88
|
Nieuwenburg SA, Mommersteeg MC, Spaander MC, Kuipers EJ. Gastric Premalignant Lesions. ENCYCLOPEDIA OF GASTROENTEROLOGY 2020:620-628. [DOI: 10.1016/b978-0-12-801238-3.65674-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
89
|
Wang S, Kuang J, Li G, Huang G, Zheng L, Li J, Wang L. Gastric precancerous lesions present in Apc Min/+ mice. Biomed Pharmacother 2019; 121:109534. [PMID: 31810128 DOI: 10.1016/j.biopha.2019.109534] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Revised: 09/28/2019] [Accepted: 10/02/2019] [Indexed: 12/25/2022] Open
Abstract
The ApcMin/+ mouse is an animal model for familial adenomatous polyposis, and aged ApcMin/+ mice also spontaneously develop multiple tumors in their stomachs. However, gastric premalignant lesions in ApcMin/+ mice have not been well characterized. The stomachs of ApcMin/+ mice were compared with those of their wild type littermates at 24 weeks with hematoxylin and eosin (H&E) staining and alcian blue staining. Ki67, CD68 and CA199 expression was analyzed by immunohistochemistry. The results revealed the presence of epithelial proliferation and inflammatory infiltration in the forestomachs, glandular atrophy and intestinal metaplasia in the gastric bodies, and dysplasia in the gastric antra. The effect of mutations in the Apc gene on chronic gastritis and gastric precancerous lesions was characterized in ApcMin/+ mice. These results suggest that ApcMin/+ mice represent a genetic model for mechanistic studies and drug discovery in gastric precancerous lesions.
Collapse
Affiliation(s)
- Sheng Wang
- School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Jianbiao Kuang
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Guifeng Li
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Guilan Huang
- School of Nursing, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Lingyun Zheng
- School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Jiangchao Li
- School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Lijing Wang
- School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou 510006, China.
| |
Collapse
|
90
|
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
|
91
|
Incidence Rate of Gastric Cancer Adenocarcinoma in Patients With Gastric Dysplasia: A Systematic Review and Meta-Analysis. J Clin Gastroenterol 2019; 53:703-710. [PMID: 31415022 DOI: 10.1097/mcg.0000000000001257] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Several studies have reported the risk of progression to gastric adenocarcinoma (GAC) in patients with gastric dysplasia (GD); however, the findings are controversial. We performed a systematic review and meta-analysis to study the incidence rate of GAC among patients with GD. Using a comprehensive search strategy, we systematically searched online databases including PubMed, Scopus, EMBASE, Cochrane Library, and Web of Science databases for identifying all relevant original articles through inception until July 2018. Cochran Q and I tests were used to assess heterogeneities between included studies. The incidence rates of GAC and their corresponding 95% confidence intervals (CIs) were pooled using random-effect or fixed-effect models. Of the 1980 retrieved records, 30 eligible articles (61 studies) were included. The overall pooled incidence rate of GAC was 40.36 (95% CI, 27.08-55.71; I, 96.0%) cases per 1000 person-years in patients with GD. Subgroup analysis according to the type of GD indicated the highest incidence rate of GAC was 186.40 (95% CI, 106.63-285.60; I, 94.6%) per 1000 person-years among patients with high-grade dysplasia (HGD) lesions. Although the incidence rates of GAC in low-grade dysplasia (LGD) lesions and in nonclassified lesions were 11.25 (95% CI, 3.91-21.22; I, 89.3%), and 1.40 (95% CI, 0.00-9.71; I, 78.8%), respectively. Compared with patients with LGD lesions, progression rate from GD to GAC was roughly 16 times greater in patients with HGD lesions. As the majority of patients with GAC are diagnosed in an advanced stage our study suggests strict management of HGD lesions to prevent GAC.
Collapse
|
92
|
Nieuwenburg SAV, Waddingham WW, Graham D, Rodriguez-Justo M, Biermann K, Kuipers EJ, Banks M, Jansen M, Spaander MCW. Accuracy of endoscopic staging and targeted biopsies for routine gastric intestinal metaplasia and gastric atrophy evaluation study protocol of a prospective, cohort study: the estimate study. BMJ Open 2019; 9:e032013. [PMID: 31537576 PMCID: PMC6756576 DOI: 10.1136/bmjopen-2019-032013] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Revised: 08/26/2019] [Accepted: 08/28/2019] [Indexed: 12/27/2022] Open
Abstract
INTRODUCTION Patients with chronic atrophic gastritis (CAG) and intestinal metaplasia (IM) are at risk of developing gastric adenocarcinoma. Their diagnosis and management currently rely on histopathological guidance after random endoscopic biopsy sampling (Sydney biopsy strategy). This approach has significant flaws such as under-diagnosis, poor reproducibility and poor correlation between endoscopy and histology. This prospective, international multicentre study aims to establish whether endoscopy-led risk stratification accurately and reproducibly predicts CAG and IM extent and disease stage. METHODS AND ANALYSIS Patients with CAG and/or IM on standard white light endoscopy (WLE) will be prospectively identified and invited to undergo a second endoscopy performed by an expert endoscopist using enhanced endoscopic imaging techniques with virtual chromoendoscopy. Extent of CAG/IM will be endoscopically staged with enhanced imaging and compared with standard WLE. Histopathological risk stratification through targeted biopsies will be compared with endoscopic disease staging and to random biopsy staging on WLE as a reference. At least 234 patients are required to show a 10 % difference in sensitivity and accuracy between enhanced imaging endoscopy-led staging and the current biopsy-led staging protocol of gastric atrophy with a power (beta) of 80 % and a 0.05 probability of a type I error (alpha). ETHICS AND DISSEMINATION The study was approved by the respective Institutional Review Boards (Netherlands: MEC-2018-078; UK: 19/LO/0089). The findings will be published in peer-reviewed journals and presented at scientific meetings. TRIAL REGISTRATION NUMBER NTR7661; Pre-results.
Collapse
Affiliation(s)
- Stella A V Nieuwenburg
- Gastroenterology & Hepatology, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - William W Waddingham
- Endoscopy, University College London Hospital, London, UK
- UCL Cancer Institute, University College London, London, UK
| | - David Graham
- Endoscopy, University College London Hospital, London, UK
| | | | - Katharina Biermann
- Pathology, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Ernst J Kuipers
- Gastroenterology & Hepatology, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Matthew Banks
- Endoscopy, University College London Hospital, London, UK
| | - Marnix Jansen
- Endoscopy, University College London Hospital, London, UK
- Pathology, University College London Hospital, London, UK
| | - Manon C W Spaander
- Gastroenterology & Hepatology, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| |
Collapse
|
93
|
Koulis A, Buckle A, Boussioutas A. Premalignant lesions and gastric cancer: Current understanding. World J Gastrointest Oncol 2019; 11:665-678. [PMID: 31558972 PMCID: PMC6755108 DOI: 10.4251/wjgo.v11.i9.665] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2019] [Revised: 05/29/2019] [Accepted: 08/21/2019] [Indexed: 02/05/2023] Open
Abstract
Over the last two decades there has been a broad paradigm shift in our understanding of gastric cancer (GC) and its premalignant states from gross histological models to increasingly precise molecular descriptions. In this review we reflect upon the historic approaches to describing premalignant lesions and GC, highlight the current molecular landscape and how this could inform future risk assessment prevention strategies.
Collapse
Affiliation(s)
- Athanasios Koulis
- Upper Gastrointestinal Translational Laboratory, Peter MacCallum Cancer Centre, Melbourne 3000, Australia
- the Sir Peter MacCallum Department of Surgical Oncology, the University of Melbourne, Melbourne 3010, Australia
| | - Andrew Buckle
- Upper Gastrointestinal Translational Laboratory, Peter MacCallum Cancer Centre, Melbourne 3000, Australia
- the Sir Peter MacCallum Department of Surgical Oncology, the University of Melbourne, Melbourne 3010, Australia
| | - Alex Boussioutas
- Upper Gastrointestinal Translational Laboratory, Peter MacCallum Cancer Centre, Melbourne 3000, Australia
- the Sir Peter MacCallum Department of Surgical Oncology, the University of Melbourne, Melbourne 3010, Australia
- Department of Medicine, Royal Melbourne Hospital, University of Melbourne, Parkville, 3050, Australia
| |
Collapse
|
94
|
Banks M, Graham D, Jansen M, Gotoda T, Coda S, di Pietro M, Uedo N, Bhandari P, Pritchard DM, Kuipers EJ, Rodriguez-Justo M, Novelli MR, Ragunath K, Shepherd N, Dinis-Ribeiro M. British Society of Gastroenterology guidelines on the diagnosis and management of patients at risk of gastric adenocarcinoma. Gut 2019; 68:1545-1575. [PMID: 31278206 PMCID: PMC6709778 DOI: 10.1136/gutjnl-2018-318126] [Citation(s) in RCA: 362] [Impact Index Per Article: 60.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2018] [Revised: 05/06/2019] [Accepted: 05/17/2019] [Indexed: 12/11/2022]
Abstract
Gastric adenocarcinoma carries a poor prognosis, in part due to the late stage of diagnosis. Risk factors include Helicobacter pylori infection, family history of gastric cancer-in particular, hereditary diffuse gastric cancer and pernicious anaemia. The stages in the progression to cancer include chronic gastritis, gastric atrophy (GA), gastric intestinal metaplasia (GIM) and dysplasia. The key to early detection of cancer and improved survival is to non-invasively identify those at risk before endoscopy. However, although biomarkers may help in the detection of patients with chronic atrophic gastritis, there is insufficient evidence to support their use for population screening. High-quality endoscopy with full mucosal visualisation is an important part of improving early detection. Image-enhanced endoscopy combined with biopsy sampling for histopathology is the best approach to detect and accurately risk-stratify GA and GIM. Biopsies following the Sydney protocol from the antrum, incisura, lesser and greater curvature allow both diagnostic confirmation and risk stratification for progression to cancer. Ideally biopsies should be directed to areas of GA or GIM visualised by high-quality endoscopy. There is insufficient evidence to support screening in a low-risk population (undergoing routine diagnostic oesophagogastroduodenoscopy) such as the UK, but endoscopic surveillance every 3 years should be offered to patients with extensive GA or GIM. Endoscopic mucosal resection or endoscopic submucosal dissection of visible gastric dysplasia and early cancer has been shown to be efficacious with a high success rate and low rate of recurrence, providing that specific quality criteria are met.
Collapse
Affiliation(s)
- Matthew Banks
- University College London Hospital, University College London Hospitals NHS Foundation Trust, London, UK.,Research Department of Targeted Intervention, University College London, London, UK
| | - David Graham
- University College London Hospital, University College London Hospitals NHS Foundation Trust, London, UK.,Division of Surgery and Interventional Science, University College London Division of Biosciences, London, UK
| | - Marnix Jansen
- Department of Histopathology, University College London, London, UK
| | - Takuji Gotoda
- Gastroenterology, Nihon University School of Medicine Graduate School of Medicine, Itabashi-ku, Tokyo, Japan
| | | | - Massimiliano di Pietro
- MRC Cancer Unit, University of Cambridge, Cambridge, UK.,Gastroenterology, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Noriya Uedo
- Department of Gastrointestinal Oncology, Endoscopic Training and Learning Center, Osaka Medical Center for Cancer and Cardiovascular Diseases, Osaka, Japan
| | | | - D Mark Pritchard
- Institute of Translational Medicine, University of Liverpool, Liverpool, UK
| | | | | | - Marco R Novelli
- Department of Histopathology, University College London, London, UK
| | - Krish Ragunath
- Nottingham Digestive Diseases Centre, Nottingham University Hospital, Nottingham, UK
| | - Neil Shepherd
- Gloucestershire Cellular Pathology Laboratory, Cheltenham General Hospital, Cheltenham, Gloucestershire, UK
| | | |
Collapse
|
95
|
Akbari M, Tabrizi R, Kardeh S, Lankarani KB. Gastric cancer in patients with gastric atrophy and intestinal metaplasia: A systematic review and meta-analysis. PLoS One 2019; 14:e0219865. [PMID: 31348819 PMCID: PMC6660080 DOI: 10.1371/journal.pone.0219865] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Accepted: 07/02/2019] [Indexed: 02/07/2023] Open
Abstract
AIM Intestinal metaplasia (IM) and gastric atrophy (GA) are precancerous lesions in the stomach. There is a large debate on natural course of these lesions and surveillance strategy in these patients. This meta-analysis was aimed to find the most appropriate follow up and the rate of progression from IM and GA to GC. METHODS This meta-analysis is followed and reported according to the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines. Electronic databases including EMBASE, PubMed, Web of Science databases, Scopus, and the Cochrane Library were searched until July 2018. Cochran's Q test and I-square (I2) test were used to examine heterogeneity across included studies. We pooled data using random-effect or fixed effect models indicated as incidence rate or proportion with 95% confidence intervals (CI). The variables of study included demographic data, endoscopy interval, follow up interval and time, GA and IM type and GC stage. Moreover, incidence rate of GC and progress rate, regress and persistence proportion in both GA and IM patients were assessed. RESULTS Overall, 68 original articles out of 32981 citations were included in our meta-analysis. The pooled GC incidence rate in patients with GA was 1.24 (95% CI, 0.80, 1.76; I2: 83.6%) cases per 1,000 person-years. The rates of later diagnosis of IM and gastric dysplasia in patients with GA were estimated as 41.42 (95% CI, 3.11, 64.45; I2: 95.6%) and 6.23 (95% CI, 2.34, 11.46; I2: 83.0%) cases per 1,000 person-years, respectively. The pooled regressed proportion was 32.23 (95% CI, 18.07-48.02; I2: 94.0%) and the persistence proportion was 38.83 (95% CI, 20.20-59.13; I2: 97.0%) per 100 observations in GA patients. In IM studies, the pooled incidence rate of GC was 3.38 (95% CI, 2.13, 4.85; I2: 93.4%) cases per 1,000 person-years. The progressed rate to dysplasia in IM patient was estimated to be 12.51 (95% CI, 5.45, 22.03; I2: 95.1%) cases per 1,000 person-years. The pooled regressed proportion was 31.83 (95% CI, 25.48-38.51; I2: 91.0%) and the persistence proportion was 43.46 (95% CI, 32.52-54.71; I2: 96.0%) per 100 observations in IM patients. CONCLUSION Overall, the incidence of GC in patients with IM and GA are low but there is heterogeneity in data with the highest rate in Asian, males with those with incomplete IM. There is probability of regression or persistence without progression in patients with IM and GA who receive appropriate management.
Collapse
Affiliation(s)
- Maryam Akbari
- Health Policy Research Center, Institute of Health, Student Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Reza Tabrizi
- Health Policy Research Center, Institute of Health, Student Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Sina Kardeh
- Cellular and Molecular Medicine Student Research Group, Shiraz School of Medicine, Shiraz, Iran
| | - Kamran B. Lankarani
- Health Policy Research Center, Institute of Health, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
96
|
Trieu JA, Bilal M, Saraireh H, Wang AY. Update on the Diagnosis and Management of Gastric Intestinal Metaplasia in the USA. Dig Dis Sci 2019; 64:1079-1088. [PMID: 30771043 DOI: 10.1007/s10620-019-05526-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Gastric intestinal metaplasia (GIM) is a premalignant condition that can lead to intestinal-type gastric adenocarcinoma. It is characterized by a change in the gastric mucosa to a small-intestinal phenotype. Infection with Helicobacter pylori is the most common factor associated with GIM. Although GIM is typically a histologic diagnosis, various techniques have been developed to enable the endoscopic identification of GIM. There are presently no widely accepted guidelines on screening and surveillance strategies in patients with GIM in the USA. The aim of this review is to provide an update regarding the problem, diagnosis, and management of GIM in the USA.
Collapse
Affiliation(s)
- Judy A Trieu
- Department of Internal Medicine, The University of Texas Medical Branch, 301 University Blvd, Galveston, TX, 77555, USA
| | - Mohammad Bilal
- Division of Gastroenterology and Hepatology, The University of Texas Medical Branch, 301 University Blvd, Galveston, TX, 77555, USA.
| | - Hamzeh Saraireh
- Division of Gastroenterology, Hepatology and Nutrition, Virginia Commonwealth University, 1200 East Broad Street, P.O. Box 98034, Richmond, VA, 23298, USA
| | - Andrew Y Wang
- Division of Gastroenterology and Hepatology, University of Virginia, P.O. Box 800708, Charlottesville, VA, 22908, USA
| |
Collapse
|
97
|
Efficacy and Long-Term Safety of H. pylori Eradication for Gastric Cancer Prevention. Cancers (Basel) 2019; 11:cancers11050593. [PMID: 31035365 PMCID: PMC6562927 DOI: 10.3390/cancers11050593] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Revised: 04/12/2019] [Accepted: 04/25/2019] [Indexed: 02/07/2023] Open
Abstract
Helicobacter pylori (H. pylori) has been shown to be a causal factor of gastric cancer in cohort studies and animal models. Meta-analysis of case-control studies nested within prospective cohorts showed that H. pylori infection was associated with a 5.9-fold increased risk of non-cardia gastric cancer. Prospective cohort studies showed that gastric cancer developed in 1–4% of H. pylori-infected subjects. Gastric cancer was successfully induced in Mongolian gerbils and insulin-gastrin (INS-GAS) transgenic mice after inoculation of H. pylori. Meta-analysis of randomized control trials also showed that eradication of H. pylori may reduce the risk of gastric cancer. However, there are several concerns regarding the widespread use of antibiotics to prevent gastric cancer, including the emergence of antibiotic resistance and the perturbation of gut microbiota after H. pylori eradication. Recent studies showed that eradication of H. pylori resulted in an increase in the bacterial diversity and restoration of the relative abundance of other bacteria to levels similar to H. pylori non-infected subjects in the gastric microbiota. The administration of antibiotics may also alter the composition of intestinal microbiota. The α-diversity and β-diversity of fecal microbiota are significantly altered immediately after H. pylori eradication but are gradually restored to levels similar to those before therapy. Yet, the rate of recovery varies with regimens. The diversity was restored at week 8 after triple therapy but was not yet fully recovered at 1 year after concomitant and quadruple therapies. Some studies showed that supplementation of probiotics may reduce the dysbiosis during H. pylori eradication therapy. Although some earlier studies showed high levels of macrolide resistance after triple therapy, recent studies showed that the increased antibiotic resistance rate may be restored 2–12 months after eradication therapy. These results collectively provide evidence of the long-term safety of H. pylori eradication. Yet, more prospective cohort studies and randomized trials are warranted to assess the efficacy and long-term safety of H. pylori eradication for gastric cancer prevention.
Collapse
|
98
|
Januszewicz W, Fitzgerald RC. Early detection and therapeutics. Mol Oncol 2019; 13:599-613. [PMID: 30677217 PMCID: PMC6396365 DOI: 10.1002/1878-0261.12458] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2018] [Revised: 01/11/2019] [Accepted: 01/19/2019] [Indexed: 12/11/2022] Open
Abstract
Early detection, including cancer screening and surveillance, is emerging as one of the most important topics in modern oncology. Because symptomatic presentation remains the predominant route to cancer diagnosis, there is a growing interest in developing techniques to detect the disease at an early, curative stage. Moreover, growing understanding of cancer biology has paved the way for prevention studies with the focus on therapeutic interventions for premalignant conditions. Where there is a recognisable precursor stage, such as a colorectal adenoma or Barrett's metaplasia, the removal of abnormal tissue prevents the development of cancer and enables stratification of the patient to a high-risk group requiring further surveillance. Here, we provide a review of the available technologies for early diagnosis and minimally-invasive treatment.
Collapse
Affiliation(s)
- Wladyslaw Januszewicz
- MRC Cancer Unit, University of Cambridge, UK.,Department of Gastroenterology, Hepatology and Clinical Oncology, Medical Centre for Postgraduate Education, Warsaw, Poland
| | | |
Collapse
|
99
|
Song L, Liu D, Zhang X, Zhu X, Lu X, Huang J, Yang L, Wu Y. Low expression of PDHA1 predicts poor prognosis in gastric cancer. Pathol Res Pract 2018; 215:478-482. [PMID: 30611622 DOI: 10.1016/j.prp.2018.12.038] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Revised: 12/14/2018] [Accepted: 12/30/2018] [Indexed: 12/12/2022]
Abstract
PDH E1 component subunit alpha (PDHA1) has been reported to be biologically significant in several human tumors. The aim of this study was to investigate the expression of PDHA1 in gastric cancer (GC) and its relationship with clinicopathological characteristics and prognosis. Oncomine analysis of neoplastic vs. normal tissue showed that the mRNA levels of PDHA1 were significantly underexpressed in different types of GC across three analyses. Underexpression of PDHA1 was found in intestinal-type GC (P = 0.009), diffuse-type GC (P = 0.036), and mixed-type GC (P = 0.025). Immunohistochemical staining of the 174 GC tissue microarray showed that PDHA1 staining is much stronger in normal mucosa than in GC samples (P = 0.040). Furthermore, PDHA1 expression levels were found to be significantly lower in 69.05% (87/126) of poorly differentiated GCs as compared to the well or moderately differentiated ones (P = 0.037). Intriguingly, PDHA1 expression was significantly correlated with depth of invasion (P < 0.001), lymph node metastasis (P < 0.001), TNM stage (P < 0.001), and nerve invasion (P = 0.006). However, it was not correlated with gender, age, Lauren classification, and lymphovascular invasion (P > 0.05 for all). Kaplan-Meier analysis revealed that low tumor expression of PDHA1 was significantly correlated with a poorer overall survival in patients with GC (5-year overall survival rates for patients with low vs high PDHA1 expression = 49.8% vs 72.7%, hazard ratio of death from GC = 2.594, 95% CI = 1.527 to 4.408, P < 0.001). Multivariate analysis showed that PDHA1 (P = 0.025) was an independent predictor of overall survival. These findings are of potential clinical utility and merit further validation.
Collapse
Affiliation(s)
- Li Song
- Department of Oncology, Affiliated Tumor Hospital of Nantong University, Nantong 226361, Jiangsu, China
| | - Danyang Liu
- Department of Oncology, Affiliated Tumor Hospital of Nantong University, Nantong 226361, Jiangsu, China
| | - Xunlei Zhang
- Department of Oncology, Affiliated Tumor Hospital of Nantong University, Nantong 226361, Jiangsu, China
| | - Xinghua Zhu
- Department of Pathology, Affiliated Tumor Hospital of Nantong University, Nantong 226361, Jiangsu, China
| | - Xiaoyun Lu
- Department of Pathology, Affiliated Tumor Hospital of Nantong University, Nantong 226361, Jiangsu, China
| | - Jieyu Huang
- Department of Pathology, Affiliated Tumor Hospital of Nantong University, Nantong 226361, Jiangsu, China
| | - Lei Yang
- Department of Oncology, Affiliated Tumor Hospital of Nantong University, Nantong 226361, Jiangsu, China.
| | - Yaxun Wu
- Department of Pathology, Affiliated Tumor Hospital of Nantong University, Nantong 226361, Jiangsu, China.
| |
Collapse
|