1
|
Yao H, Liu T, Chen Y, She L, Wu T, Liu D, Deng Y, Han Y, Chen K, Deng J, Zhang J, Chen J, Liu F. Dysregulated gastric microbial communities and functional shifts in chronic atrophic versus non-atrophic gastritis: a Helicobacter pylori-Negative observational study. BMC Gastroenterol 2025; 25:304. [PMID: 40301773 PMCID: PMC12039101 DOI: 10.1186/s12876-025-03900-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Accepted: 04/15/2025] [Indexed: 05/01/2025] Open
Abstract
BACKGROUND Helicobacter pylori (H. pylori) infection was identified as a substantial risk factor for gastric cancer development, but the eradication of H. pylori did not necessarily lead to a reduction in the incidence of gastric cancer. Non-Helicobacter pylori (non-H. pylori) bacteria in the stomach are involved in the transformation of gastritis carcinoma. The aim of this study was to characterize the microbiome composition of the gastric mucosa and its functions in non-H. pylori (H. pylori-negative) patients with chronic atrophic gastritis (CAG) and chronic non-atrophic gastritis (CNAG). METHODS Fourteen CNAG samples and twenty-three CAG samples were collected. The composition of the gastric microbiome was analyzed using 16 S rDNA gene sequencing. The bioinformatic analysis was performed using alpha and beta diversity analyses, PICRUSt2, and linear discriminant analysis effect size (LEfSe). RESULTS The two groups shared the same most abundant bacterial phyla (Pseudomonadota, Bacillota, Actinomycetota, and Bacteroidota). The top 5 most abundant bacterial genera in the CAG group were Sphingomonas, Ralstonia, Brevundimonas, Methyloversatilis, and Pseudomonas. In the CNAG group, the top genera were Brevundimonas, Ralstonia, Sphingomonas, Methyloversatilis, and Acinetobacter. Differential analysis revealed distinct genera between groups: the CAG group showed enrichment in Sphingomonas, Ralstonia, Bradyrhizobium, Roseateles, and Acidithiobacillus, while the CNAG group was enriched in Brevundimonas, Rhodococcus, Hydrogenophaga, Bacteroides, and Leifsonia (p < 0.05). Sphingomonas exhibited a positive correlation with Acidithiobacillus but negative correlations with Brevundimonas, Hydrogenophaga, and Leifsonia. Pathways related to xenobiotic biodegradation, metabolism, signal transduction, cofactor/vitamin metabolism, cancer, infectious diseases, and digestive system were enriched in the CAG group. In contrast, the CNAG group showed enrichment in amino acid metabolism, translation, replication/repair, and terpenoid/polyketide metabolism. CONCLUSION Gastric mucosal microbiota dysbiosis and functional shifts are significantly associated with chronic atrophic gastritis. Taxa such as Sphingomonas and Ralstonia, enriched in CAG patients, may indicate microbial signatures associated with early atrophic transition and provide candidates for further mechanistic validation.
Collapse
Affiliation(s)
- Hong Yao
- Guangzhou University of Chinese Medicine, Guangzhou, 510405, Guangdong, People's Republic of China
- The Eighth Clinical Medical College of Guangzhou University of Chinese Medicine, Foshan, 528000, Guangdong, People's Republic of China
- Foshan Hospital of Traditional Chinese Medicine, Foshan, 528000, Guangdong, People's Republic of China
| | - Tingting Liu
- The Eighth Clinical Medical College of Guangzhou University of Chinese Medicine, Foshan, 528000, Guangdong, People's Republic of China
- Foshan Hospital of Traditional Chinese Medicine, Foshan, 528000, Guangdong, People's Republic of China
| | - Yiming Chen
- The Eighth Clinical Medical College of Guangzhou University of Chinese Medicine, Foshan, 528000, Guangdong, People's Republic of China
- Foshan Hospital of Traditional Chinese Medicine, Foshan, 528000, Guangdong, People's Republic of China
| | - Ling She
- The Eighth Clinical Medical College of Guangzhou University of Chinese Medicine, Foshan, 528000, Guangdong, People's Republic of China
- Foshan Hospital of Traditional Chinese Medicine, Foshan, 528000, Guangdong, People's Republic of China
| | - Tingfeng Wu
- The Eighth Clinical Medical College of Guangzhou University of Chinese Medicine, Foshan, 528000, Guangdong, People's Republic of China
- Foshan Hospital of Traditional Chinese Medicine, Foshan, 528000, Guangdong, People's Republic of China
| | - Dongsong Liu
- The Eighth Clinical Medical College of Guangzhou University of Chinese Medicine, Foshan, 528000, Guangdong, People's Republic of China
- Foshan Hospital of Traditional Chinese Medicine, Foshan, 528000, Guangdong, People's Republic of China
| | - Yuhong Deng
- The Eighth Clinical Medical College of Guangzhou University of Chinese Medicine, Foshan, 528000, Guangdong, People's Republic of China
- Foshan Hospital of Traditional Chinese Medicine, Foshan, 528000, Guangdong, People's Republic of China
| | - Yubin Han
- The Eighth Clinical Medical College of Guangzhou University of Chinese Medicine, Foshan, 528000, Guangdong, People's Republic of China
- Foshan Hospital of Traditional Chinese Medicine, Foshan, 528000, Guangdong, People's Republic of China
| | - Kai Chen
- The Eighth Clinical Medical College of Guangzhou University of Chinese Medicine, Foshan, 528000, Guangdong, People's Republic of China
- Foshan Hospital of Traditional Chinese Medicine, Foshan, 528000, Guangdong, People's Republic of China
| | - Jianmin Deng
- The Eighth Clinical Medical College of Guangzhou University of Chinese Medicine, Foshan, 528000, Guangdong, People's Republic of China
- Foshan Hospital of Traditional Chinese Medicine, Foshan, 528000, Guangdong, People's Republic of China
| | - Jue Zhang
- The Eighth Clinical Medical College of Guangzhou University of Chinese Medicine, Foshan, 528000, Guangdong, People's Republic of China
- Foshan Hospital of Traditional Chinese Medicine, Foshan, 528000, Guangdong, People's Republic of China
| | - Jinfeng Chen
- The Eighth Clinical Medical College of Guangzhou University of Chinese Medicine, Foshan, 528000, Guangdong, People's Republic of China.
- Foshan Hospital of Traditional Chinese Medicine, Foshan, 528000, Guangdong, People's Republic of China.
| | - Fengbin Liu
- Lingnan Institute of Spleen and Stomach Diseases, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510405, Guangdong, People's Republic of China.
- Baiyun Hospital of the First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510080, Guangdong, People's Republic of China.
| |
Collapse
|
2
|
Yamada H, Abe S, Charvat H, Ando T, Maeda M, Murakami K, Oka S, Maekita T, Sugimoto M, Furuta T, Kaise M, Yamamichi N, Takamaru H, Sasaki A, Oda I, Nanjo S, Suzuki N, Sugiyama T, Kodama M, Mizukami K, Ito M, Kotachi T, Shimazu T, Yamamoto S, Ushijima T. Precision risk stratification of primary gastric cancer after eradication of H. pylori by a DNA methylation marker: a multicentre prospective study. Gut 2025:gutjnl-2025-335039. [PMID: 40240063 DOI: 10.1136/gutjnl-2025-335039] [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: 02/07/2025] [Accepted: 03/14/2025] [Indexed: 04/18/2025]
Abstract
BACKGROUND Precision cancer risk stratification for gastric cancer is urgently needed for the growing number of healthy people after Helicobacter pylori eradication. The epimutation burden in non-malignant tissues has been associated with cancer risk in multiple cross-sectional studies. OBJECTIVE To confirm the clinical usefulness of a DNA methylation marker for epimutation burden, and to identify a cut-off methylation level for a super-high-risk population. DESIGN Healthy people after H. pylori eradication with open-type atrophy were prospectively recruited. DNA methylation levels of a marker gene, RIMS1, were measured in biopsy specimens from gastric antrum and body. The primary endpoint was the incidence rate of gastric cancer in quartiles of the methylation levels. RESULTS 1624 participants had at least one endoscopic follow-up with a median follow-up of 4.05 years, and a primary gastric cancer developed in 27 participants. The highest quartile of RIMS1 methylation levels had a higher incidence rate (972.8 per 100 000 person-years) than the lowest quartile (127.1). Cox regression analysis revealed a univariate HR of 7.7 (95% CI 1.8-33.7) and an age- and sex-adjusted HR of 5.7 (95% CI 1.3-25.5). As a secondary objective, a cut-off methylation level of 25.7% (95% CI 1.7-7.7) was obtained to identify a population with a super-high risk based on the number needed to screen of 1000. CONCLUSION A DNA methylation marker can risk-stratify healthy people after H. pylori eradication even though all of them have clinically high risk. Individuals with super-high risk will need more frequent gastric cancer screening than currently recommended. TRIAL REGISTRATION NUMBER UMIN-CTR000016894.
Collapse
Affiliation(s)
- Harumi Yamada
- Department of Epigenomics, Institute for Advanced Life Sciences, Hoshi University, Tokyo, Japan
- Department of Gastrointestinal Surgery, Kyoto University, Kyoto, Japan
- Division of Epigenomics, National Cancer Center Research Institute, Tokyo, Japan
| | - Seiichiro Abe
- Endoscopy Division, National Cancer Center Hospital, Tokyo, Japan
| | - Hadrien Charvat
- Faculty of International Liberal Arts, Juntendo University, Tokyo, Japan
- Division of International Health Policy Research, Institute for Cancer Control, National Cancer Center, Tokyo, Japan
| | - Takayuki Ando
- Third Department of Internal Medicine, University of Toyama, Toyama, Japan
| | - Masahiro Maeda
- Department of Gastrointestinal Surgery, Kyoto University, Kyoto, Japan
- Division of Epigenomics, National Cancer Center Research Institute, Tokyo, Japan
| | - Kazunari Murakami
- Department of Gastroenterology, Faculty of Medicine, Oita University, Oita, Japan
| | - Shiro Oka
- Department of Gastroenterology, Hiroshima University Hospital, Hiroshima, Japan
| | - Takao Maekita
- Second Department of Internal Medicine, Wakayama Medical University, Wakayama, Japan
| | - Mitsushige Sugimoto
- Division of Digestive Endoscopy, Shiga University of Medical Science Hospital, Shiga, Japan
- Division of Genome-Wide Infectious Microbiology, Research Center for GLOBAL and LOCAL Infectious Disease, Oita University, Oita, Japan
| | - Takahisa Furuta
- Center for Clinical Research, Hamamatsu University School of Medicine, Shizuoka, Japan
| | - Mitsuru Kaise
- Department of Gastroenterology, Nippon Medical School, Graduate School of Medicine, Tokyo, Japan
| | - Nobutake Yamamichi
- Department of Gastroenterology, The University of Tokyo, Graduate School of Medicine, Tokyo, Japan
| | | | - Akiko Sasaki
- Gastroenterology Medicine Center, Shonan Kamakura General Hospital, Kanagawa, Japan
| | - Ichiro Oda
- Endoscopy Division, National Cancer Center Hospital, Tokyo, Japan
| | - Sohachi Nanjo
- Third Department of Internal Medicine, University of Toyama, Toyama, Japan
| | - Nobuhiro Suzuki
- Department of Internal Medicine, Joetsu General Hospital, Niigata, Japan
| | - Toshiro Sugiyama
- Third Department of Internal Medicine, University of Toyama, Toyama, Japan
- Health Sciences University of Hokkaido, Hokkaido, Japan (Present adrress)
| | - Masaaki Kodama
- Department of Gastroenterology, Faculty of Medicine, Oita University, Oita, Japan
- Department of Advanced Medical Sciences, Faculty of Medicine, Oita University, Oita, Japan
| | - Kazuhiro Mizukami
- Department of Gastroenterology, Faculty of Medicine, Oita University, Oita, Japan
| | - Masanori Ito
- Department of Gastroenterology, Hiroshima University Hospital, Hiroshima, Japan
| | - Takahiro Kotachi
- Department of Gastroenterology, Hiroshima University Hospital, Hiroshima, Japan
| | - Taichi Shimazu
- Epidemiology and Prevention Group, Research Center for Cancer Prevention andScreening, National Cancer Center, Tokyo, Japan
| | | | - Toshikazu Ushijima
- Department of Epigenomics, Institute for Advanced Life Sciences, Hoshi University, Tokyo, Japan
- Division of Epigenomics, National Cancer Center Research Institute, Tokyo, Japan
| |
Collapse
|
3
|
Yang P, Liang G, Ni Y, Chu X, Zhang X, Wang Z, Khan A, Jin F, Shen H, Li M, Xu Z. Investigating the role of intratumoral Streptococcus mitis in gastric cancer progression: insights into tumor microenvironment. J Transl Med 2025; 23:126. [PMID: 39875915 PMCID: PMC11773703 DOI: 10.1186/s12967-025-06142-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Accepted: 12/18/2024] [Indexed: 01/30/2025] Open
Abstract
Growing evidence implicates that intratumoral microbiota are closely linked to cancer progression; however, research on the role of these microbiota in the development of gastric cancer remains limited. Here, using 16 S rRNA sequencing, tumor tissue proteomics and serum cytokines analysis, we identified enrichment of specific microbial communities within tumors of gastric cancer patients, possibly affecting the tumor microenvironment by immune modulation, metabolic processes, and inflammatory responses. Based on the results of in vivo experiments and intratumoral microbiota analysis, we found that Streptococcus mitis can inhibit gastric cancer progression via suppressing M2 macrophage polarization and infiltration, as well as altering the intratumoral microbial community. In summary, our findings suggest that the intratumoral microbiota, exemplified by Streptococcus mites, may be involved in regulating the progression of gastric cancer, thereby emerging as potential therapeutic targets for this disease.
Collapse
Affiliation(s)
- Ping Yang
- Department of Clinical Laboratory, Nanjing Drum Tower Hospital Clinical College of Xuzhou Medical University, Nanjing, 210008, China
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Physiology, Jiangsu Engineering Research Center for MicroRNA Biology and Biotechnology, School of Life Sciences, NJU Advanced Institute of Life Sciences (NAILS), Nanjing University, Nanjing, 210008, Jiangsu, P. R. China
| | - Gaoli Liang
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Physiology, Jiangsu Engineering Research Center for MicroRNA Biology and Biotechnology, School of Life Sciences, NJU Advanced Institute of Life Sciences (NAILS), Nanjing University, Nanjing, 210008, Jiangsu, P. R. China
| | - Yangyue Ni
- Department of Pathogen Biology, Key Laboratory for Pathogen Infection and Control of Jiangsu Province, Nanjing Medical University, Nanjing, 211166, Jiangsu, P.R. China
| | - Xiaojie Chu
- Department of Clinical Laboratory, Nanjing Drum Tower Hospital Clinical College of Xuzhou Medical University, Nanjing, 210008, China
| | - Xiaoshan Zhang
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Physiology, Jiangsu Engineering Research Center for MicroRNA Biology and Biotechnology, School of Life Sciences, NJU Advanced Institute of Life Sciences (NAILS), Nanjing University, Nanjing, 210008, Jiangsu, P. R. China
| | - Zhongyu Wang
- Department of Clinical Laboratory, Nanjing Drum Tower Hospital Clinical College of Xuzhou Medical University, Nanjing, 210008, China
| | - Adeel Khan
- Department of Biotechnology, University of Science and Technology Bannu, Bannu, 28100, KP, Pakistan
| | - Fangfang Jin
- School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, P. R. China.
| | - Han Shen
- Department of Clinical Laboratory, Nanjing Drum Tower Hospital Clinical College of Xuzhou Medical University, Nanjing, 210008, China.
| | - Miao Li
- Department of Clinical Laboratory, Nanjing Drum Tower Hospital Clinical College of Xuzhou Medical University, Nanjing, 210008, China.
| | - Zhipeng Xu
- Department of Pathogen Biology, Key Laboratory for Pathogen Infection and Control of Jiangsu Province, Nanjing Medical University, Nanjing, 211166, Jiangsu, P.R. China.
| |
Collapse
|
4
|
Heidary M, Akrami S, Madanipour T, Shakib NH, Mahdizade Ari M, Beig M, Khoshnood S, Ghanavati R, Bazdar M. Effect of Helicobacter pylori-induced gastric cancer on gastrointestinal microbiota: a narrative review. Front Oncol 2025; 14:1495596. [PMID: 39868371 PMCID: PMC11757270 DOI: 10.3389/fonc.2024.1495596] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Accepted: 12/12/2024] [Indexed: 01/28/2025] Open
Abstract
Helicobacter pylori (H. pylori) infection is a typical microbial agent that interferes with the complex mechanisms of gastric homeostasis by disrupting the balance between the host gastric microbiota and mucosa-related factors, ultimately leading to inflammatory changes, dysbiosis, and gastric cancer (GC). We searched this field on the basis of PubMed, Google Scholar, Web of Science, and Scopus databases. Most studies show that H. pylori inhibits the colonization of other bacteria, resulting in a less variety of bacteria in the gastrointestinal (GI) tract. When comparing the patients with H. pylori-positive and H. pylori-negative GC, the composition of the gastric microbiome changes with increasing abundance of H. pylori (where present) in the gastritis stage, whereas, as the gastric carcinogenesis cascade progresses to GC, oral and intestinal-type pathogenic microbial strains predominate. H. pylori infection induces a premalignant milieu of atrophy and intestinal metaplasia, and the resulting change in gastric microbiota appears to play an important role in gastric carcinogenesis. The effect of H. pylori-induced GC on GI microbiota is discussed in this review.
Collapse
Affiliation(s)
- Mohsen Heidary
- Leishmaniasis Research Center, Sabzevar University of Medical Sciences, Sabzevar, Iran
| | - Sousan Akrami
- Department of Microbiology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Tohid Madanipour
- Department of Microbiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Nafiseh Hosseinzadeh Shakib
- Department of Bacteriology and Virology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Marzie Mahdizade Ari
- Department of Microbiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Masoumeh Beig
- Department of Bacteriology, Pasteur Institute of Iran, Tehran, Iran
| | - Saeed Khoshnood
- Clinical Microbiology Research Center, Ilam University of Medical Sciences, Ilam, Iran
| | - Roya Ghanavati
- School of Medicine, Behbahan Faculty of Medical Sciences, Behbahan, Iran
| | - Monireh Bazdar
- School of Medicine, Razi Hospital, Ilam University of Medical Sciences, Ilam, Iran
| |
Collapse
|
5
|
Zou PY, Zhu JR, Zhao Z, Mei H, Zhao JT, Sun WJ, Wang GH, Chen DF, Fan LL, Lan CH. Development and application of an artificial intelligence-assisted endoscopy system for diagnosis of Helicobacter pylori infection: a multicenter randomized controlled study. BMC Gastroenterol 2024; 24:335. [PMID: 39350033 PMCID: PMC11440712 DOI: 10.1186/s12876-024-03389-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Accepted: 08/27/2024] [Indexed: 10/04/2024] Open
Abstract
BACKGROUND The early diagnosis and treatment of Heliobacter pylori (H.pylori) gastrointestinal infection provide significant benefits to patients. We constructed a convolutional neural network (CNN) model based on an endoscopic system to diagnose H. pylori infection, and then examined the potential benefit of this model to endoscopists in their diagnosis of H. pylori infection. MATERIALS AND METHODS A CNN neural network system for endoscopic diagnosis of H.pylori infection was established by collecting 7377 endoscopic images from 639 patients. The accuracy, sensitivity, and specificity were determined. Then, a randomized controlled study was used to compare the accuracy of diagnosis of H. pylori infection by endoscopists who were assisted or unassisted by this CNN model. RESULTS The deep CNN model for diagnosis of H. pylori infection had an accuracy of 89.6%, a sensitivity of 90.9%, and a specificity of 88.9%. Relative to the group of endoscopists unassisted by AI, the AI-assisted group had better accuracy (92.8% [194/209; 95%CI: 89.3%, 96.4%] vs. 75.6% [158/209; 95%CI: 69.7%, 81.5%]), sensitivity (91.8% [67/73; 95%CI: 85.3%, 98.2%] vs. 78.6% [44/56; 95%CI: 67.5%, 89.7%]), and specificity (93.4% [127/136; 95%CI: 89.2%, 97.6%] vs. 74.5% [114/153; 95%CI: 67.5%, 81.5%]). All of these differences were statistically significant (P < 0.05). CONCLUSION Our AI-assisted system for diagnosis of H. pylori infection has significant ability for diagnostic, and can improve the accuracy of endoscopists in gastroscopic diagnosis. TRIAL REGISTRATION This study was approved by the Ethics Committee of Daping Hospital (10/07/2020) (No.89,2020) and was registered with the Chinese Clinical Trial Registration Center (02/09/2020) ( www.chictr.org.cn ; registration number: ChiCTR2000037801).
Collapse
Affiliation(s)
- Pei-Ying Zou
- Department of Gastroenterology, Chongqing Key Laboratory of Digestive, Malignancies, Daping Hospital, Army Medical University, Third Military Medical University), 10 Changjiang Branch Road, Chongqing, 400000, China
| | - Jian-Ru Zhu
- Department of Gastroenterology, Chongqing Key Laboratory of Digestive, Malignancies, Daping Hospital, Army Medical University, Third Military Medical University), 10 Changjiang Branch Road, Chongqing, 400000, China
| | - Zhe Zhao
- Department of Gastroenterology, Chongqing Key Laboratory of Digestive, Malignancies, Daping Hospital, Army Medical University, Third Military Medical University), 10 Changjiang Branch Road, Chongqing, 400000, China
| | - Hao Mei
- Department of Gastroenterology, Chongqing Key Laboratory of Digestive, Malignancies, Daping Hospital, Army Medical University, Third Military Medical University), 10 Changjiang Branch Road, Chongqing, 400000, China
| | - Jing-Tao Zhao
- Department of Gastroenterology, Chongqing Key Laboratory of Digestive, Malignancies, Daping Hospital, Army Medical University, Third Military Medical University), 10 Changjiang Branch Road, Chongqing, 400000, China
| | - Wen-Jing Sun
- Chongqing 13, People's Hospital, Chongqing, China
| | | | - Dong-Feng Chen
- Department of Gastroenterology, Chongqing Key Laboratory of Digestive, Malignancies, Daping Hospital, Army Medical University, Third Military Medical University), 10 Changjiang Branch Road, Chongqing, 400000, China
| | - Li-Lin Fan
- Chongqing Jiulongpo District Second People's Hospital, Chongqing, China
| | - Chun-Hui Lan
- Department of Gastroenterology, Chongqing Key Laboratory of Digestive, Malignancies, Daping Hospital, Army Medical University, Third Military Medical University), 10 Changjiang Branch Road, Chongqing, 400000, China.
| |
Collapse
|
6
|
Drnovsek J, Homan M, Zidar N, Smid LM. Pathogenesis and potential reversibility of intestinal metaplasia - a milestone in gastric carcinogenesis. Radiol Oncol 2024; 58:186-195. [PMID: 38643513 PMCID: PMC11165985 DOI: 10.2478/raon-2024-0028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Accepted: 03/19/2024] [Indexed: 04/23/2024] Open
Abstract
BACKGROUND Non-cardia gastric cancer remains a major cause of cancer-related mortality worldwide, despite declining incidence rates in many industrialized countries. The development of intestinal-type gastric cancer occurs through a multistep process in which normal mucosa is sequentially transformed into hyperproliferative epithelium, followed by metaplastic processes leading to carcinogenesis. Chronic infection with Helicobacter pylori is the primary etiological agent that causes chronic inflammation of the gastric mucosa, induces atrophic gastritis, and can lead to intestinal metaplasia and dysplasia. Both intestinal metaplasia and dysplasia are precancerous lesions, in which gastric cancer is more likely to occur. Atrophic gastritis often improves after eradication of Helicobacter pylori; however, the occurrence of intestinal metaplasia has been traditionally regarded as "the point of no return" in the carcinogenesis sequence. Helicobacter pylori eradication heals non-atrophic chronic gastritis, may lead to regression of atrophic gastritis, and reduces the risk of gastric cancer in patients with these conditions. In this article, we discuss the pathogenesis, epigenomics, and reversibility of intestinal metaplasia and briefly touch upon potential treatment strategy. CONCLUSIONS Gastric intestinal metaplasia no longer appears to be an irreversible precancerous lesion. However, there are still many controversies regarding the improvement of intestinal metaplasia after Helicobacter pylori eradication.
Collapse
Affiliation(s)
- Jan Drnovsek
- Department of Gastroenterology, University Medical Centre Ljubljana, Ljubljana, Slovenia
- Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Matjaz Homan
- Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
- Department of Gastroenterology, Hepatology and Nutrition, University Children’s Hospital, Ljubljana, Slovenia
| | - Nina Zidar
- Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
- Institute of Pathology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Lojze M Smid
- Department of Gastroenterology, University Medical Centre Ljubljana, Ljubljana, Slovenia
- Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| |
Collapse
|
7
|
Liang Y, Yang Y, Nong R, Huang H, Chen X, Deng Y, Huang Z, Huang J, Cheng C, Ji M, Chen Y, Hu F. Do atrophic gastritis and intestinal metaplasia reverse after Helicobacter pylori eradication? Helicobacter 2024; 29:e13042. [PMID: 38018403 DOI: 10.1111/hel.13042] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Revised: 11/15/2023] [Accepted: 11/17/2023] [Indexed: 11/30/2023]
Abstract
BACKGROUND It's still controversial whether Helicobacter pylori (H. pylori) eradication can reverse atrophic gastritis (AG) and intestinal metaplasia (IM). Therefore, we performed a meta-analysis to estimate the effect of H. pylori eradication on AG and IM. METHODS We searched the PubMed, Web of Science and EMBASE datasets through April 2023 for epidemiological studies, which provided mean glandular atrophy (GA) or IM score before and after H. pylori eradication, or provided ORs, RRs or HRs and 95% CIs for the association of AG or IM with H. pylori eradication. Weighted mean difference (WMD) and pooled ORs and 95%CIs were used to estimate the effect of H. pylori eradication on AG and IM. RESULTS Twenty articles with a total of 5242 participants were included in this meta-analysis. H. pylori eradication significantly decreased GA score in the antrum (WMD -0.36; 95% CI: -0.52, -0.19, p < 0.01), GA score in the corpus (WMD -0.35; 95% CI: -0.52, -0.19, p < 0.01), IM score in the antrum (WMD -0.16; 95% CI: -0.26, -0.07, p < 0.01) and IM score in the corpus (WMD -0.20; 95% CI: -0.37, -0.04, p = 0.01). H. pylori eradication significantly improved AG (pooled OR 2.96; 95% CI: 1.70, 5.14, p < 0.01) and IM (pooled OR 2.41; 95% CI: 1.24, 4.70, p < 0.01). The association remained significant in the subgroup analyses by study design, sites of lesions, regions and follow-up time. Although Publication bias was observed for AG, the association remained significant after trim-and-fill adjustment. CONCLUSIONS H. pylori eradication could significantly improve AG and IM at early stage.
Collapse
Affiliation(s)
- Yongqiang Liang
- Department of Gastroenterology, Shenzhen Qianhai Shekou Free Trade Zone Hospital, Shenzhen, Guangdong, People's Republic of China
- Department of Epidemiology and Health Statistics, School of Public Health, Shenzhen University Health Science Centre, Shenzhen, Guangdong, People's Republic of China
- 2019 Preventive Medicine, School of Public Health, Shenzhen University Health Science Centre, Shenzhen, Guangdong, People's Republic of China
| | - Yuanhai Yang
- Department of Epidemiology and Health Statistics, School of Public Health, Shenzhen University Health Science Centre, Shenzhen, Guangdong, People's Republic of China
- 2020 Preventive Medicine, School of Public Health, Shenzhen University Health Science Centre, Shenzhen, Guangdong, People's Republic of China
| | - Ruiheng Nong
- Department of Epidemiology and Health Statistics, School of Public Health, Shenzhen University Health Science Centre, Shenzhen, Guangdong, People's Republic of China
- 2020 Preventive Medicine, School of Public Health, Shenzhen University Health Science Centre, Shenzhen, Guangdong, People's Republic of China
| | - Hao Huang
- Department of Epidemiology and Health Statistics, School of Public Health, Shenzhen University Health Science Centre, Shenzhen, Guangdong, People's Republic of China
| | - Xiuyun Chen
- Department of Gastroenterology, Shenzhen Qianhai Shekou Free Trade Zone Hospital, Shenzhen, Guangdong, People's Republic of China
| | - Ying Deng
- Department of Epidemiology and Health Statistics, Fujian Provincial Key Laboratory of Environment Factors and Cancer, School of Public Health, Fujian Medical University, Fujian, China
| | - Zhicong Huang
- Department of Epidemiology and Health Statistics, Fujian Provincial Key Laboratory of Environment Factors and Cancer, School of Public Health, Fujian Medical University, Fujian, China
| | - Jingyao Huang
- Department of Epidemiology and Health Statistics, Fujian Provincial Key Laboratory of Environment Factors and Cancer, School of Public Health, Fujian Medical University, Fujian, China
| | - Chunsheng Cheng
- Department of Gastroenterology and Endoscopy Centre, Huazhong University of Science and Technology Union Shenzhen Hospital (Nanshan Hospital) and The 6th Affiliated Hospital of Shenzhen University School of Medicine, Shenzhen, Guangdong, China
| | - Mingzhu Ji
- Department of Gastroenterology, Shenzhen Qianhai Shekou Free Trade Zone Hospital, Shenzhen, Guangdong, People's Republic of China
| | - Yinggang Chen
- National Cancer Centre/National Clinical Research Centre for Cancer/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, China
| | - Fulan Hu
- Department of Epidemiology and Health Statistics, School of Public Health, Shenzhen University Health Science Centre, Shenzhen, Guangdong, People's Republic of China
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Shenzhen University Health Science Centre, Shenzhen, Guangdong, People's Republic of China
| |
Collapse
|
8
|
Wang S, Qian M, Wu M, Feng S, Zhang K. The prediction model of operative link on gastric intestinal metaplasia stage III-IV: A multicenter study. Heliyon 2023; 9:e21905. [PMID: 38027917 PMCID: PMC10665748 DOI: 10.1016/j.heliyon.2023.e21905] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2023] [Revised: 10/31/2023] [Accepted: 10/31/2023] [Indexed: 12/01/2023] Open
Abstract
Purpose Intestinal metaplasia plays a crucial role in the risk stratification of gastric cancer development. The objective of the study was to develop a prediction model for Operative Link on Gastric Intestinal Metaplasia (OLGIM) Stage III-IV. Methods We analyzed 7945 high-risk gastric cancer individuals from 115 hospitals who underwent questionnaires and gastroscope. The participants were assigned to either the development or validation cohort randomly. Demographics and clinical characteristics were obtained. The outcome measurement was OLGIM III-IV. Univariate logistic regression was used for feature selection and multivariate logistic analysis was performed to develop the nomogram. Area under the curves, calibration plots, decision curve and clinical impact analysis were used to assess the performance of the nomogram. Results 4600 individuals and 3345 individuals were included in the development and validation cohort, of which 124 and 86 individuals were diagnosed with OLGIM III-IV, respectively. Parameters in the training validation cohort matched well and there was no significant difference between two cohorts. A nomogram model for predicting OLGIM Stage III-IV consisted of 4 significantly associated variables, including age, gender, PG I and G-17 (AUC 0.723 and 0.700 for the 2 cohorts). The nomogram demonstrated excellent performance in the calibration curve. Decision curve and clinical impact analysis suggested clinical benefit of the prediction model. Conclusions This reliable individualized nomogram might contribute to more accurate management for patients with OLGIM III-IV. Therefore, we suggest that this study be used as an incentive to promote the application.
Collapse
Affiliation(s)
- Song Wang
- Digestive Endoscopic Center, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Gastroenterology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Meng Qian
- Department of Gastroenterology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Min Wu
- Department of Gastroenterology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Shuo Feng
- Department of Gastroenterology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Kaiguang Zhang
- Department of Gastroenterology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| |
Collapse
|
9
|
Kang SJ, Kim JG, Moon HS, Kook MC, Lee JY, Bang CS, Tae CH, Gong EJ, Nam SY, Kim HJ. Clinical Practice Guideline for Gastritis in Korea. J Korean Med Sci 2023; 38:e115. [PMID: 37012690 PMCID: PMC10070048 DOI: 10.3346/jkms.2023.38.e115] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Accepted: 03/09/2023] [Indexed: 04/05/2023] Open
Abstract
Gastritis is a disease characterized by inflammation of the gastric mucosa. It is very common and has various classification systems such as the updated Sydney system. As there is a lot of evidence that Helicobacter pylori infection is associated with the development of gastric cancer and that gastric cancer can be prevented by eradication, H. pylori gastritis has been emphasized recently. The incidence rate of gastric cancer in Korea is the highest in the world, and due to the spread of screening endoscopy, atrophic gastritis and intestinal metaplasia are commonly diagnosed in the general population. However, there have been no clinical guidelines developed in Korea for these lesions. Therefore, this clinical guideline has been developed by the Korean College of Helicobacter and Upper Gastrointestinal Research for important topics that are frequently encountered in clinical situations related to gastritis. Evidence-based guidelines were developed through systematic review and de novo processes, and eight recommendations were made for eight key questions. This guideline needs to be periodically revised according to the needs of clinical practice or as important evidence about this issue is published in the future.
Collapse
Affiliation(s)
- Seung Joo Kang
- Deparment of Internal Medicine, Seoul National University Hospital Healthcare System Gangnam Center, Seoul, Korea
| | - Jae Gyu Kim
- Department of Internal Medicine, Chung-Ang University College of Medicine, Seoul, Korea.
| | - Hee Seok Moon
- Department of Internal Medicine, Chungnam National University College of Medicine, Daejeon, Korea
| | | | - Jong Yeul Lee
- Center for Gastric Cancer, National Cancer Center, Goyang, Korea
| | - Chang Seok Bang
- Department of Internal Medicine, Hallym University College of Medicine, Chuncheon, Korea
| | - Chung Hyun Tae
- Department of Internal Medicine, Ewha Womans University College of Medicine, Seoul, Korea
| | - Eun Jeong Gong
- Department of Internal Medicine, Hallym University College of Medicine, Chuncheon, Korea
| | - Su Youn Nam
- Department of Internal Medicine, School of Medicine, Kyungpook National University, Daegu, Korea
- Division of Gastroenterology, Department of Internal Medicine, Kyungpook National University Chilgok Hospital, Daegu, Korea
| | - Hyun Jung Kim
- Department of Preventive Medicine, Korea University College of Medicine, Seoul, Korea
| |
Collapse
|
10
|
Noh CK, Lee E, Park B, Lim SG, Shin SJ, Lee KM, Lee GH. Effect of Helicobacter pylori Eradication Treatment on Metachronous Gastric Neoplasm Prevention Following Endoscopic Submucosal Dissection for Gastric Adenoma. J Clin Med 2023; 12:1512. [PMID: 36836045 PMCID: PMC9962017 DOI: 10.3390/jcm12041512] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2023] [Revised: 02/11/2023] [Accepted: 02/13/2023] [Indexed: 02/17/2023] Open
Abstract
The long-term effect of Helicobacter pylori eradication on metachronous gastric neoplasm prevention after endoscopic submucosal dissection (ESD) of gastric adenoma is unclear. This study included patients with confirmed H. pylori infection after ESD with curative resection for gastric adenoma. Patients were divided based on the success of H. pylori eradication treatment into two groups: eradication and non-eradication. Patients with any newly detected lesion within 1 year after ESD and recurrence at the ESD site were excluded from the analysis. Further, 1:1 propensity score matching was also performed to eliminate baseline differences between the two groups. H. pylori eradication treatment was administered to 673 patients after ESD (163 in the successful eradication group and 510 in the non-eradication group). During the median follow-up periods of 25 and 39 months in the eradication and non-eradication groups, metachronous gastric neoplasm was identified in 6 (3.7%) and 22 patients (4.3%), respectively. Adjusted Cox analysis revealed that H. pylori eradication was not associated with increased risk of metachronous gastric neoplasm after ESD. Kaplan-Meier analysis in the matched population yielded similar findings (p = 0.546). H. pylori eradication treatment was not associated with metachronous gastric neoplasm after ESD with curative resection for gastric adenoma.
Collapse
Affiliation(s)
- Choong-Kyun Noh
- Department of Gastroenterology, Ajou University School of Medicine, Suwon 16499, Republic of Korea
| | - Eunyoung Lee
- Department of Biomedical Informatics, Ajou University School of Medicine, Suwon 16499, Republic of Korea
- Office of Biostatistics, Ajou Research Institute for Innovative Medicine, Ajou University Medical Center, Suwon 16499, Republic of Korea
| | - Bumhee Park
- Department of Biomedical Informatics, Ajou University School of Medicine, Suwon 16499, Republic of Korea
- Office of Biostatistics, Ajou Research Institute for Innovative Medicine, Ajou University Medical Center, Suwon 16499, Republic of Korea
| | - Sun Gyo Lim
- Department of Gastroenterology, Ajou University School of Medicine, Suwon 16499, Republic of Korea
| | - Sung Jae Shin
- Department of Gastroenterology, Ajou University School of Medicine, Suwon 16499, Republic of Korea
| | - Kee Myung Lee
- Department of Gastroenterology, Ajou University School of Medicine, Suwon 16499, Republic of Korea
| | - Gil Ho Lee
- Department of Gastroenterology, Ajou University School of Medicine, Suwon 16499, Republic of Korea
| |
Collapse
|
11
|
Kim N, Yoon H. Atrophic Gastritis and Intestinal Metaplasia. HELICOBACTER PYLORI 2023:641-659. [DOI: 10.1007/978-981-97-0013-4_55] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
12
|
Bhat MH, Hajam YA, Neelam, Kumar R, Diksha. Microbial Diversity and Their Role in Human Health and Diseases. ROLE OF MICROBES IN SUSTAINABLE DEVELOPMENT 2023:1-33. [DOI: 10.1007/978-981-99-3126-2_1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
13
|
Wang P, Li P, Chen Y, Li L, Lu Y, Zhou W, Bian L, Zhang B, Yin X, Li J, Chen J, Zhang S, Shi Y, Tang X. Chinese integrated guideline on the management of gastric precancerous conditions and lesions. Chin Med 2022; 17:138. [PMID: 36517854 PMCID: PMC9749368 DOI: 10.1186/s13020-022-00677-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Accepted: 10/17/2022] [Indexed: 12/15/2022] Open
Abstract
The standardized diagnosis and management of gastric precancerous conditions and lesions are important to prevent gastric cancer. This guideline, created by 5 traditional Chinese medicine and Western medicine associations, based on the current morbidity and diagnosis and treatment of gastric precancerous conditions and lesions, provides specific key points and strategies for diagnosis and treatment in the following five aspects: definition and epidemiology, diagnosis and stage, surveillance, treatment and efficacy evaluation. It is hoped that these aspects, assessed by integrating Western medicine and traditional Chinese medicine and involving multidisciplinary participation, will play a guiding role in clinical diagnosis and treatment and achieve effective secondary prevention of gastric cancer.
Collapse
Affiliation(s)
- Ping Wang
- China Academy of Chinese Medical Sciences, Xiyuan Hospital, Beijing, China
| | - Peng Li
- Capital Medical University Affiliated Beijing Friendship Hospital, Beijing, China
| | - Yingxuan Chen
- Shanghai Jiao Tong University School of Medicine Affiliated Renji Hospital, Shanghai, China
| | - Li Li
- China Academy of Chinese Medical Sciences, Guanganmen Hospital, Beijing, China
| | - Yuanyuan Lu
- Air Force Medical University Xijing Hospital, Xi'an, China
| | - Weixun Zhou
- Peking Union Medical College Hospital, Beijing, China
| | - Liqun Bian
- China Academy of Chinese Medical Sciences, Xiyuan Hospital, Beijing, China
| | - Beihua Zhang
- China Academy of Chinese Medical Sciences, Xiyuan Hospital, Beijing, China
| | - Xiaolan Yin
- China Academy of Chinese Medical Sciences, Xiyuan Hospital, Beijing, China
| | - Junxiang Li
- Beijing University of Chinese Medicine School of Traditional Chinese Medicine, Beijing, China.
| | - Jie Chen
- Peking Union Medical College Hospital, Beijing, China.
| | - Shutian Zhang
- Capital Medical University Affiliated Beijing Friendship Hospital, Beijing, China.
| | - Yongquan Shi
- Air Force Medical University Xijing Hospital, Xi'an, China.
| | - Xudong Tang
- China Academy of Chinese Medical Sciences, Xiyuan Hospital, Beijing, China.
| |
Collapse
|
14
|
Kligman E, Ali H, Chen E, Peng F, Szafron D, Staggers K, Tan MC, Patel K, Othman MO. Ethnicity Is an Important Consideration in Screening for Gastric Intestinal Metaplasia. Dig Dis Sci 2022; 67:4509-4517. [PMID: 34981309 DOI: 10.1007/s10620-021-07326-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Accepted: 11/03/2021] [Indexed: 12/24/2022]
Abstract
BACKGROUND Gastric intestinal metaplasia (GIM) is a precursor to gastric adenocarcinoma, making it an attractive target for early detection by endoscopy. The aim of this study was to determine the prevalence, risk factors, and associated histologic findings of GIM among patients undergoing endoscopy in a diverse US population. METHODS We conducted a retrospective, cross-sectional study of patients undergoing elective endoscopy with gastric biopsies at 6 academic and community centers in Houston, Texas. GIM prevalence was estimated with a 95% confidence interval (CI), and patient demographic and clinical characteristics were summarized using mean with standard deviation, or frequency with percentage. Generalized estimating equations (GEE) were used to compare characteristics between those with and without GIM. RESULTS Our final cohort consisted of 2685 patients, including 216 cases with GIM and 2469 controls. The prevalence of GIM in our cohort was 8.04% (95% CI 7.07%, 9.14%). The mean age of GIM cases was higher than in the control group (59.8 vs 54.7 years, p < 0.0001). The prevalence of GIM in Asians, Hispanic, Black and Non-Hispanic Whites (NHW) was 14.7%, 11.7%, 9.8% and 5.8%, respectively. On multivariable analysis, factors associated with GIM include age (adj. OR 1.32 per 10 year increase, p < 0.0001), habitual smoking (adj. OR 1.68, p < 0.0001), and race (compared to NHW: Asian, adj. OR 2.34, p = 0.010; Hispanic, adj. OR 2.15, p < 0.001; Black, adj. OR 1.61, p < 0.001). CONCLUSION Asians, Hispanics, and African Americans have higher rates of GIM than NHW. Ethnicity should be an important consideration on determining who to screen for GIM in the US.
Collapse
Affiliation(s)
- Eugene Kligman
- Section of Gastroenterology and Hepatology, Baylor College of Medicine, 7200 Cambridge Street. Suite 8A, Houston, TX, 77030, USA
| | - Hiba Ali
- Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Ellie Chen
- Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Frederick Peng
- Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - David Szafron
- Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Kristen Staggers
- Institute for Clinical and Translational Research, Baylor College of Medicine, Houston, TX, USA
| | - Mimi C Tan
- Section of Gastroenterology and Hepatology, Baylor College of Medicine, 7200 Cambridge Street. Suite 8A, Houston, TX, 77030, USA
| | - Kalpesh Patel
- Section of Gastroenterology and Hepatology, Baylor College of Medicine, 7200 Cambridge Street. Suite 8A, Houston, TX, 77030, USA
| | - Mohamed O Othman
- Section of Gastroenterology and Hepatology, Baylor College of Medicine, 7200 Cambridge Street. Suite 8A, Houston, TX, 77030, USA.
- Baylor St Luke's Medical Center, Houston, TX, USA.
| |
Collapse
|
15
|
Torun C, Yavuz A, Akan K, Seneldir H, Toksoz AN, Ulasoglu HC, Tuncer I. Comparison of the diagnostic accuracy of the updated Sydney system and single biopsy. Saudi J Gastroenterol 2022; 28:441-447. [PMID: 35899924 PMCID: PMC9843512 DOI: 10.4103/sjg.sjg_146_22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
BACKGROUND Updated Sydney system (USS) recommends taking biopsies from certain areas of the stomach for the diagnosis of precancerous lesions associated with Helicobacter pylori. Our aim was to evaluate the contribution of each of the biopsy sites to the diagnosis. METHODS This prospective study included 97 patients aged 40 and over with dyspeptic complaints. Biopsies were taken from five regions: the lesser curvature of the antrum (LCA), the lesser curvature of the corpus (LCC), incisura angularis (IA), the greater curvature of the antrum (GCA), and the greater curvature of the corpus (GCC). Biopsy specimens were stained with hematoxylin-eosin stain, periodic acid Schiff-alcian blue, and Giemsa histochemical stain and evaluated according to the Sydney classification. RESULTS Thirty-seven (38%) patients were positive for H. pylori in at least one biopsy site. Atrophic gastritis without intestinal metaplasia (IM) was found in 17 (17.5%) of the patients (6.2% in IA, 5.2% in each of LCA, GCA, and LCC, and 2% in GCC). The prevalence of atrophic gastritis with IM was 42.3% (21.6% in LCA, 20.6% in GCA, 20.6% in IA, 14.4% in LCC, and 5.2% in GCC). Endoscopic follow-up was planned in 21 (22%) patients due to the presence of extensive atrophy or incomplete IM. If a single biopsy of the LCA or a biopsy of both LCA and GCA was taken, endoscopic follow-up would have been missed in 12 (57%) or 6 (29%) patients, respectively. CONCLUSION Taking biopsies in accordance with the USS had higher sensitivity in detecting atrophic gastritis with or without IM compared to single biopsy. One or two biopsies is not sufficient to identify patients for whom endoscopic follow-up is recommended.
Collapse
Affiliation(s)
- Cundullah Torun
- Department of Internal Medicine, Goztepe Training and Research Hospital, Istanbul Medeniyet University, Kadikoy/Istanbul, Turkey,Address for correspondence: Dr. Cundullah Torun, Goztepe Training and Research Hospital, Province of Istanbul, District of Kadıköy, Neighbourhood of Eğitim – 34722, Turkey. E-mail:
| | - Arda Yavuz
- Department of Gastroenterology, Goztepe Training and Research Hospital, Istanbul Medeniyet University, Kadikoy/Istanbul, Turkey
| | - Kubra Akan
- Department of Gastroenterology, Goztepe Training and Research Hospital, Istanbul Medeniyet University, Kadikoy/Istanbul, Turkey
| | - Hatice Seneldir
- Department of Medical Pathology, Goztepe Training and Research Hospital, Istanbul Medeniyet University, Kadikoy/Istanbul, Turkey
| | - Ayse Nur Toksoz
- Department of Medical Pathology, Goztepe Training and Research Hospital, Istanbul Medeniyet University, Kadikoy/Istanbul, Turkey
| | - Hak Celal Ulasoglu
- Department of Gastroenterology, Istanbul Okan University, Tuzla/Istanbul, Turkey
| | - Ilyas Tuncer
- Department of Gastroenterology, Goztepe Training and Research Hospital, Istanbul Medeniyet University, Kadikoy/Istanbul, Turkey
| |
Collapse
|
16
|
Zhou CB, Pan SY, Jin P, Deng JW, Xue JH, Ma XY, Xie YH, Cao H, Liu Q, Xie WF, Zou XP, Sheng JQ, Wang BM, Wang H, Ren JL, Liu SD, Sun YW, Meng XJ, Zhao G, Chen JX, Cui Y, Wang PQ, Guo HM, Yang L, Chen X, Ding J, Yang XN, Wang XK, Qian AH, Hou LD, Wang Z, Chen YX, Fang JY. Fecal Signatures of Streptococcus anginosus and Streptococcus constellatus for Noninvasive Screening and Early Warning of Gastric Cancer. Gastroenterology 2022; 162:1933-1947.e18. [PMID: 35167866 DOI: 10.1053/j.gastro.2022.02.015] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Revised: 01/25/2022] [Accepted: 02/06/2022] [Indexed: 12/30/2022]
Abstract
BACKGROUND & AIMS Most patients with gastric cancer (GCa) are diagnosed at an advanced stage. We aimed to investigate novel fecal signatures for clinical application in early diagnosis of GCa. METHODS This was an observational study that included 1043 patients from 10 hospitals in China. In the discovery cohort, 16S ribosomal RNA gene analysis was performed in paired samples (tissues and feces) from patients with GCa and chronic gastritis (ChG) to determine differential abundant microbes. Their relative abundances were detected using quantitative real-time polymerase chain reaction to test them as bacterial candidates in the training cohort. Their diagnostic efficacy was validated in the validation cohort. RESULTS Significant enrichments of Streptococcus anginosus (Sa) and Streptococcus constellatus (Sc) in GCa tumor tissues (P < .05) and feces (P < .0001) were observed in patients with intraepithelial neoplasia, early and advanced GCa. Either the signature parallel test Sa∪Sc or single signature Sa/Sc demonstrated superior sensitivity (Sa: 75.6% vs 72.1%, P < .05; Sc: 84.4% vs 64.0%, P < .001; and Sa∪Sc: 91.1% vs 81.4%, P < .01) in detecting early GCa compared with advanced GCa (specificity: Sa: 84.0% vs 83.9%, Sc: 70.4% vs 82.3%, and Sa∪Sc: 64.0% vs 73.4%). Fecal signature Sa∪Sc outperformed Sa∪CEA/Sc∪CEA in the discrimination of advanced GCa (sensitivity: 81.4% vs 74.2% and 81.4% vs 72.3%, P < .01; specificity: 73.4% vs 81.0 % and 73.4% vs 81.0%). The performance of Sa∪Sc in the diagnosis of both early and advanced GCa was verified in the validation cohort. CONCLUSION Fecal Sa and Sc are noninvasive, accurate, and sensitive signatures for early warning in GCa. (ClinicalTrials.gov, Number: NCT04638959).
Collapse
Affiliation(s)
- Cheng-Bei Zhou
- Division of Gastroenterology and Hepatology; Shanghai Institute of Digestive Disease; NHC Key Laboratory of Digestive Diseases; State Key Laboratory for Oncogenes and Related Genes, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Si-Yuan Pan
- Division of Gastroenterology and Hepatology; Shanghai Institute of Digestive Disease; NHC Key Laboratory of Digestive Diseases; State Key Laboratory for Oncogenes and Related Genes, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Peng Jin
- Department of Gastroenterology, The First Medical Center of Chinese People's Liberation Army General Hospital, The Seventh Medical Center of Chinese People's Liberation Army General Hospital, Beijing, China
| | - Jia-Wen Deng
- Division of Gastroenterology and Hepatology; Shanghai Institute of Digestive Disease; NHC Key Laboratory of Digestive Diseases; State Key Laboratory for Oncogenes and Related Genes, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Jin-Hui Xue
- Department of Clinical Research, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Xin-Yue Ma
- Division of Gastroenterology and Hepatology; Shanghai Institute of Digestive Disease; NHC Key Laboratory of Digestive Diseases; State Key Laboratory for Oncogenes and Related Genes, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yuan-Hong Xie
- Division of Gastroenterology and Hepatology; Shanghai Institute of Digestive Disease; NHC Key Laboratory of Digestive Diseases; State Key Laboratory for Oncogenes and Related Genes, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Hui Cao
- Department of Gastrointestinal Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Qiang Liu
- Department of Pathology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Wei-Fen Xie
- Department of Gastroenterology, Changzheng Hospital, Second Military Medical University, Shanghai, China
| | - Xiao-Ping Zou
- Department of Gastroenterology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
| | - Jian-Qiu Sheng
- Department of Gastroenterology, The First Medical Center of Chinese People's Liberation Army General Hospital, The Seventh Medical Center of Chinese People's Liberation Army General Hospital, Beijing, China
| | - Bang-Mao Wang
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin, China
| | - Hong Wang
- Department of Gastroenterology, Shanghai Jing'an District Central Hospital, Fudan University, Shanghai, China
| | - Jian-Lin Ren
- Department of Gastroenterology, Zhongshan Hospital, Xiamen University, Xiamen, China
| | - Si-De Liu
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yun-Wei Sun
- Department of Gastroenterology, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiang-Jun Meng
- Department of Gastroenterology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Gang Zhao
- Department of Gastrointestinal Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Jin-Xian Chen
- Department of Gastrointestinal Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yun Cui
- Division of Gastroenterology and Hepatology; Shanghai Institute of Digestive Disease; NHC Key Laboratory of Digestive Diseases; State Key Laboratory for Oncogenes and Related Genes, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Pei-Qin Wang
- Department of Gastroenterology, Changzheng Hospital, Second Military Medical University, Shanghai, China
| | - Hui-Min Guo
- Department of Gastroenterology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
| | - Lang Yang
- Department of Gastroenterology, The First Medical Center of Chinese People's Liberation Army General Hospital, The Seventh Medical Center of Chinese People's Liberation Army General Hospital, Beijing, China
| | - Xin Chen
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin, China
| | - Jia Ding
- Department of Gastroenterology, Shanghai Jing'an District Central Hospital, Fudan University, Shanghai, China
| | - Xiao-Ning Yang
- Department of Gastroenterology, Zhongshan Hospital, Xiamen University, Xiamen, China
| | - Xin-Ke Wang
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Ai-Hua Qian
- Department of Gastroenterology, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Li-Dan Hou
- Department of Gastroenterology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zheng Wang
- Department of Gastrointestinal Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
| | - Ying-Xuan Chen
- Division of Gastroenterology and Hepatology; Shanghai Institute of Digestive Disease; NHC Key Laboratory of Digestive Diseases; State Key Laboratory for Oncogenes and Related Genes, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
| | - Jing-Yuan Fang
- Division of Gastroenterology and Hepatology; Shanghai Institute of Digestive Disease; NHC Key Laboratory of Digestive Diseases; State Key Laboratory for Oncogenes and Related Genes, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
| |
Collapse
|
17
|
Role of Gastric Microorganisms Other than Helicobacter pylori in the Development and Treatment of Gastric Diseases. BIOMED RESEARCH INTERNATIONAL 2022; 2022:6263423. [PMID: 35321071 PMCID: PMC8938066 DOI: 10.1155/2022/6263423] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Revised: 12/02/2021] [Accepted: 02/18/2022] [Indexed: 12/15/2022]
Abstract
The microenvironment in the stomach is different from other digestive tracts, mainly because of the secretion of gastric acid and digestive enzymes, bile reflux, special mucus barrier, gastric peristalsis, and so on, which all contribute to the formation of antibacterial environment. Microecological disorders can lead to gastric immune disorders or lead to the decrease of dominant bacteria and the increase of the abundance and virulence of pathogenic microorganisms and then promote the occurrence of diseases. The body performs its immune function through innate and adaptive immunity and maintains microbial balance through the mechanism of immune homeostasis. Microecological imbalance can lead to the invasion of pathogenic microorganisms and damage mucosal barrier and immune system. The coexistence of gastric microorganisms (including viruses and fungi) may play a synergistic or antagonistic role in the pathogenesis of gastric diseases. Probiotics have the ability to compete with intestinal pathogens, increase the secretion of immunoglobulin A (IgA), stimulate the production of mucin, bacteriocin, and lactic acid, regulate the expression and secretion of cytokines, and regulate the growth of microbiota, which all have beneficial effects on the host microbial environment. At present, most studies focused on Helicobacter pylori, ignoring other stomach microbes and the overall stomach microecology. So, in this article, we reviewed advances in human gastric microecology, the relationship between gastric microecology and immunity or gastric diseases, and the treatment of probiotics in gastric diseases, in order to explore new area for further study of gastric microorganisms and treatment of gastric diseases.
Collapse
|
18
|
Basso L, Gallo G, Biacchi D, Carati MV, Cavallaro G, Esposito L, Giuliani A, Izzo L, Izzo P, Lamazza A, Polistena A, Tarallo M, Micarelli A, Fiori E. Role of New Anatomy, Biliopancreatic Reflux, and Helicobacter Pylori Status in Postgastrectomy Stump Cancer. J Clin Med 2022; 11:1498. [PMID: 35329824 PMCID: PMC8952228 DOI: 10.3390/jcm11061498] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Revised: 03/06/2022] [Accepted: 03/07/2022] [Indexed: 01/14/2023] Open
Abstract
Distal gastrectomy for benign gastroduodenal peptic disease has become rare, but it still represents a widely adopted procedure for advanced and, in some countries, even for early distal gastric cancer. Survival rates following surgery for gastric malignancy are constantly improving, hence the residual mucosa of the gastric stump is exposed for a prolonged period to biliopancreatic reflux and, possibly, to Helicobacter pylori (HP) infection. Biliopancreatic reflux and HP infection are considered responsible for gastritis and metachronous carcinoma in the gastric stump after oncologic surgery. For gastrectomy patients, in addition to eradication treatment for cases that are already HP positive, endoscopic surveillance should also be recommended, for prompt surveillance and detection in the residual mucosa of any metaplastic-atrophic-dysplastic features following surgery.
Collapse
Affiliation(s)
- Luigi Basso
- “Pietro Valdoni” Department of Surgery, Policlinico “Umberto I”, “Sapienza” University of Rome, 00161 Rome, Italy; (D.B.); (M.V.C.); (G.C.); (L.E.); (A.G.); (L.I.); (P.I.); (A.L.); (A.P.); (M.T.); (E.F.)
| | - Gaetano Gallo
- Department of Medicine, Surgery and Neurosciences, Operative Unit of General Surgery and Surgical Oncology, University of Siena, 53100 Siena, Italy
| | - Daniele Biacchi
- “Pietro Valdoni” Department of Surgery, Policlinico “Umberto I”, “Sapienza” University of Rome, 00161 Rome, Italy; (D.B.); (M.V.C.); (G.C.); (L.E.); (A.G.); (L.I.); (P.I.); (A.L.); (A.P.); (M.T.); (E.F.)
| | - Maria Vittoria Carati
- “Pietro Valdoni” Department of Surgery, Policlinico “Umberto I”, “Sapienza” University of Rome, 00161 Rome, Italy; (D.B.); (M.V.C.); (G.C.); (L.E.); (A.G.); (L.I.); (P.I.); (A.L.); (A.P.); (M.T.); (E.F.)
| | - Giuseppe Cavallaro
- “Pietro Valdoni” Department of Surgery, Policlinico “Umberto I”, “Sapienza” University of Rome, 00161 Rome, Italy; (D.B.); (M.V.C.); (G.C.); (L.E.); (A.G.); (L.I.); (P.I.); (A.L.); (A.P.); (M.T.); (E.F.)
| | - Luca Esposito
- “Pietro Valdoni” Department of Surgery, Policlinico “Umberto I”, “Sapienza” University of Rome, 00161 Rome, Italy; (D.B.); (M.V.C.); (G.C.); (L.E.); (A.G.); (L.I.); (P.I.); (A.L.); (A.P.); (M.T.); (E.F.)
| | - Andrea Giuliani
- “Pietro Valdoni” Department of Surgery, Policlinico “Umberto I”, “Sapienza” University of Rome, 00161 Rome, Italy; (D.B.); (M.V.C.); (G.C.); (L.E.); (A.G.); (L.I.); (P.I.); (A.L.); (A.P.); (M.T.); (E.F.)
| | - Luciano Izzo
- “Pietro Valdoni” Department of Surgery, Policlinico “Umberto I”, “Sapienza” University of Rome, 00161 Rome, Italy; (D.B.); (M.V.C.); (G.C.); (L.E.); (A.G.); (L.I.); (P.I.); (A.L.); (A.P.); (M.T.); (E.F.)
| | - Paolo Izzo
- “Pietro Valdoni” Department of Surgery, Policlinico “Umberto I”, “Sapienza” University of Rome, 00161 Rome, Italy; (D.B.); (M.V.C.); (G.C.); (L.E.); (A.G.); (L.I.); (P.I.); (A.L.); (A.P.); (M.T.); (E.F.)
| | - Antonietta Lamazza
- “Pietro Valdoni” Department of Surgery, Policlinico “Umberto I”, “Sapienza” University of Rome, 00161 Rome, Italy; (D.B.); (M.V.C.); (G.C.); (L.E.); (A.G.); (L.I.); (P.I.); (A.L.); (A.P.); (M.T.); (E.F.)
| | - Andrea Polistena
- “Pietro Valdoni” Department of Surgery, Policlinico “Umberto I”, “Sapienza” University of Rome, 00161 Rome, Italy; (D.B.); (M.V.C.); (G.C.); (L.E.); (A.G.); (L.I.); (P.I.); (A.L.); (A.P.); (M.T.); (E.F.)
| | - Mariarita Tarallo
- “Pietro Valdoni” Department of Surgery, Policlinico “Umberto I”, “Sapienza” University of Rome, 00161 Rome, Italy; (D.B.); (M.V.C.); (G.C.); (L.E.); (A.G.); (L.I.); (P.I.); (A.L.); (A.P.); (M.T.); (E.F.)
| | - Alessandro Micarelli
- ITER Center for Balance and Rehabilitation Research (ICBRR), 02032 Rome, Italy;
- Eurac Research, Institute of Mountain Emergency Medicine, 39100 Bolzano, Italy
| | - Enrico Fiori
- “Pietro Valdoni” Department of Surgery, Policlinico “Umberto I”, “Sapienza” University of Rome, 00161 Rome, Italy; (D.B.); (M.V.C.); (G.C.); (L.E.); (A.G.); (L.I.); (P.I.); (A.L.); (A.P.); (M.T.); (E.F.)
| |
Collapse
|
19
|
Shahini E, Maida M. Surveillance strategies for precancerous gastric conditions after Helicobacter pylori eradication: There is still need for a tailored approach. World J Gastroenterol 2021; 27:8033-8039. [PMID: 35046629 PMCID: PMC8678819 DOI: 10.3748/wjg.v27.i46.8033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2021] [Revised: 10/03/2021] [Accepted: 11/25/2021] [Indexed: 02/06/2023] Open
Abstract
Prevailing evidence declares that Helicobacter pylori (H. pylori) eradication therapy could shift precancerous gastric conditions (PGC) and positively confines gastric cancer (GC) risk during long-term endoscopic follow-up. Nonetheless, there is a yet unsolved controversy regarding the best-individualized surveillance strategies following H. pylori eradication, based on malignant risk stratification. This last dispute is due to the uncertainty of contemporary evidence and the role of H. pylori inflammatory changes in underestimating PGC at the index endoscopy. However, the current state of the art suggests that it is reasonable that high-quality endoscopy with histological assessment for the most accurate diagnosis of PGC may be delayed in selected high-risk patients without alarm signs for malignancy, following the eradication of H. pylori. Notwithstanding, these aspects need to be further examined in the next future to establish and optimize the most beneficial and cost-effective strategies for recognizing and managing H. pylori-positive patients with PGC in the short- and long-term follow-up. Accordingly, additional studies are yet required to sharpen the hazard stratification of patients with the greatest chance of GC evolution, also recognizing the evolving racial, ethnic, immigration factors and the necessity of novel biomarkers to limit GC development or accomplish a diagnosis of malignancy at an early stage.
Collapse
Affiliation(s)
- Endrit Shahini
- Division of Gastroenterology, National Institute of Research "Saverio De Bellis", Castellana Grotte (Bari) 70013, Italy
| | - Marcello Maida
- Section of Gastroenterology, S.Elia - Raimondi Hospital, Caltanissetta 93017, Italy
| |
Collapse
|
20
|
Chen LW, Chang LC, Hua CC, Cheng TC, Lee CC. Comparing the Expressions of Vitamin D Receptor, Cell Proliferation, and Apoptosis in Gastric Mucosa With Gastritis, Intestinal Metaplasia, or Adenocarcinoma Change. Front Med (Lausanne) 2021; 8:766061. [PMID: 34881266 PMCID: PMC8645899 DOI: 10.3389/fmed.2021.766061] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2021] [Accepted: 10/27/2021] [Indexed: 12/20/2022] Open
Abstract
Background: This study aimed to compare the expression of vitamin D receptor (VDR), cell proliferation, and apoptosis in the gastric mucosa of patients with gastritis, intestinal metaplasia (IM), and adenocarcinoma using artificial intelligence. Material and Methods: This study retrospectively enrolled patients at the Keelung Chang Gung Memorial Hospital from November of 2016 to June, 2017, who were diagnosed with gastric adenocarcinoma. The inclusion criteria were patients' pathologic reports that revealed all compartments of Helicobacter pylori infection, gastritis, IM, and adenocarcinoma simultaneously in the same gastric sample. Tissue slides after immunohistochemical (IHC) staining were transformed into digital images using a scanner and counted using computer software (QuPath and ImageJ). IHC staining included PA1-711 antibody for VDR, Ki67 antigen for proliferation, and M30 antibody CK18 for apoptosis. Results: Twenty-nine patients were included in the IHC staining quantitative analysis. The mean age was 69.1 ± 11.3 y/o. Most (25/29, 86.2%) patients had poorly differentiated adenocarcinoma. The mean expression of Ki67 and CK18 increased progressively from gastritis and IM to adenocarcinoma, with statistical significance (P < 0.05). VDR expression did not correlate with Ki67 or CK18 expression. Survival time was only correlated with tumor stage (correlation coefficient = −0.423, P value < 0.05), but was not correlated with the expression of VDR, Ki67, and CK18. Conclusion: Ki67 expression and CK18 expression progressively increased in the areas of gastritis, IM, and adenocarcinoma. No correlation between VDR expression and Ki67 or CK18 expression was found in this study.
Collapse
Affiliation(s)
- Li-Wei Chen
- Department of Gastroenterology and Hepatology, Chang-Gung Memorial Hospital and University at Keelung, Keelung, Taiwan.,Community Medicine Research Center, Chang-Gung Memorial Hospital and University at Keelung, Keelung, Taiwan
| | - Liang-Che Chang
- Department of Pathology, Pathology Chang-Gung Memorial Hospital and University at Keelung, Keelung, Taiwan
| | - Chung-Ching Hua
- Department of Internal Medicine, Internal Medicine Chang-Gung Memorial Hospital and University at Keelung, Keelung, Taiwan
| | - Tzu-Chien Cheng
- Department of Pathology, Pathology Chang-Gung Memorial Hospital and University at Keelung, Keelung, Taiwan
| | - Chin-Chan Lee
- Community Medicine Research Center, Chang-Gung Memorial Hospital and University at Keelung, Keelung, Taiwan.,Department of Internal Medicine, Internal Medicine Chang-Gung Memorial Hospital and University at Keelung, Keelung, Taiwan
| |
Collapse
|
21
|
Al Hennawi H, Khedr A, Maan RK, Albarazi M, Atluri P. Gastric Intestinal Metaplasia and Its Rapid Progression Toward Gastric Adenocarcinoma: A Call for Clear Patient Management and Awareness. Cureus 2021; 13:e18751. [PMID: 34796052 PMCID: PMC8589342 DOI: 10.7759/cureus.18751] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/12/2021] [Indexed: 11/05/2022] Open
Abstract
Gastric cancer is one of the leading causes of cancer-related death worldwide. Helicobacter pylori (H. pylori) infection is known to cause gastric adenocarcinoma in a stepwise fashion. Gastric intestinal metaplasia is a known premalignant stage. We report a case of a 70-year-old male patient with active chronic H. pylori-associated gastritis and focal intestinal metaplasia on the initial presentation, who rapidly developed diffuse, poorly differentiated gastric adenocarcinoma 20 months after the loss to follow-up. Our case highlights the premalignant nature of gastric intestinal metaplasia (GIM) and the extreme importance of early eradication of H. pylori. We also address the lack of definitive GIM surveillance guidelines.
Collapse
Affiliation(s)
- Hussam Al Hennawi
- Internal Medicine, Alfaisal University College of Medicine, Riyadh, SAU
| | - Anwar Khedr
- Internal Medicine, Tanta University Faculty of Medicine, Tanta, EGY
| | | | | | - Purna Atluri
- Gastroenterology, University Hospital of Brooklyn, State University of New York Downstate Medical Center, Brooklyn, USA
| |
Collapse
|
22
|
Weng CY, Xu JL, Sun SP, Wang KJ, Lv B. Helicobacter pylori eradication: Exploring its impacts on the gastric mucosa. World J Gastroenterol 2021; 27:5152-5170. [PMID: 34497441 PMCID: PMC8384747 DOI: 10.3748/wjg.v27.i31.5152] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 06/14/2021] [Accepted: 07/15/2021] [Indexed: 02/06/2023] Open
Abstract
Helicobacter pylori (H. pylori) infects approximately 50% of all humans globally. Persistent H. pylori infection causes multiple gastric and extragastric diseases, indicating the importance of early diagnosis and timely treatment. H. pylori eradication produces dramatic changes in the gastric mucosa, resulting in restored function. Consequently, to better understand the importance of H. pylori eradication and clarify the subsequent recovery of gastric mucosal functions after eradication, we summarize histological, endoscopic, and gastric microbiota changes to assess the therapeutic effects on the gastric mucosa.
Collapse
Affiliation(s)
- Chun-Yan Weng
- Department of Gastroenterology, The First Clinical Medical College of Zhejiang Chinese Medical University, Hangzhou 310053, Zhejiang Province, China
| | - Jing-Li Xu
- Department of Gastrointestinal Surgery, The First Clinical Medical College of Zhejiang Chinese Medical University, Hangzhou 310053, Zhejiang Province, China
| | - Shao-Peng Sun
- Department of Gastroenterology, The First Clinical Medical College of Zhejiang Chinese Medical University, Hangzhou 310053, Zhejiang Province, China
| | - Kai-Jie Wang
- Department of Gastroenterology, The First Clinical Medical College of Zhejiang Chinese Medical University, Hangzhou 310053, Zhejiang Province, China
| | - Bin Lv
- Department of Gastroenterology, The First Clinical Medical College of Zhejiang Chinese Medical University, Hangzhou 310053, Zhejiang Province, China
- Department of Gastroenterology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou 310006, Zhejiang Province, China
| |
Collapse
|
23
|
Bae DH, Kim HJ, Yoon BH, Park JL, Kim M, Kim SK, Kim SY, Lee SI, Song KS, Kim YS. STK31 upregulation is associated with chromatin remodeling in gastric cancer and induction of tumorigenicity in a xenograft mouse model. Oncol Rep 2021; 45:42. [PMID: 33649810 PMCID: PMC7934220 DOI: 10.3892/or.2021.7993] [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: 10/05/2020] [Accepted: 01/19/2021] [Indexed: 12/26/2022] Open
Abstract
Pathological changes in the epigenetic landscape of chromatin are hallmarks of cancer. Our previous study showed that global methylation of promoters may increase or decrease during the transition from gastric mucosa to intestinal metaplasia (IM) to gastric cancer (GC). Here, CpG hypomethylation of the serine/threonine kinase STK31 promoter in IM and GC was detected in a reduced representation bisulfite sequencing database. STK31 hypomethylation, which resulted in its upregulation in 120 cases of primary GC, was confirmed. Using public genome-wide histone modification data, upregulation of STK31 promoter activity was detected in primary GC but not in normal mucosae, suggesting that STK31 may be repressed in gastric mucosa but activated in GC as a consequence of hypomethylation-associated chromatin remodeling. STK31 knockdown suppressed the proliferation, colony formation and migration activities of GC cells in vitro, whereas stable overexpression of STK31 promoted the proliferation, colony formation, and migration activities of GC cells in vitro and tumorigenesis in nude mice. Patients with GC in which STK31 was upregulated exhibited significantly shorter survival times in a combined cohort. Thus, activation of STK31 by chromatin remodeling may be associated with gastric carcinogenesis and also may help predict GC prognosis.
Collapse
Affiliation(s)
- Dong Hyuck Bae
- Genome Editing Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon 34141, Republic of Korea
| | - Hee-Jin Kim
- Genome Editing Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon 34141, Republic of Korea
| | - Byoung-Ha Yoon
- Personalized Genomic Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon 34141, Republic of Korea
| | - Jong-Lyul Park
- Genome Editing Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon 34141, Republic of Korea
| | - Mirang Kim
- Department of Functional Genomics, Korea University of Science and Technology, Daejeon 34113, Republic of Korea
| | - Seon-Kyu Kim
- Personalized Genomic Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon 34141, Republic of Korea
| | - Seon-Young Kim
- Department of Functional Genomics, Korea University of Science and Technology, Daejeon 34113, Republic of Korea
| | - Sang-Il Lee
- Department of Surgery, College of Medicine, Chungnam National University, Daejeon 35015, Republic of Korea
| | - Kyu-Sang Song
- Department of Pathology, College of Medicine, Chungnam National University, Daejeon 35015, Republic of Korea
| | - Yong Sung Kim
- Genome Editing Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon 34141, Republic of Korea
| |
Collapse
|
24
|
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
|
25
|
Pereira-Marques J, Ferreira RM, Machado JC, Figueiredo C. The influence of the gastric microbiota in gastric cancer development. Best Pract Res Clin Gastroenterol 2021; 50-51:101734. [PMID: 33975676 DOI: 10.1016/j.bpg.2021.101734] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Revised: 02/12/2021] [Accepted: 02/15/2021] [Indexed: 01/31/2023]
Abstract
Colonization of the stomach by Helicobacter pylori is the trigger for a series of gastric mucosal changes that culminate in gastric cancer. Infection with this bacterium is considered the major risk factor for this malignancy. The introduction of high-throughput sequencing technologies coupled to advanced computational pipelines offered an improved understanding of the microbiome, and it is now currently accepted that, besides H. pylori, the stomach harbours a complex microbial community. While it is well established that H. pylori plays a central role in gastric carcinogenesis, the significance of the non-H. pylori microbiota is yet to be clarified. This review will address the state of the art on the relationship between the gastric microbiota and gastric cancer development, and identify areas where additional research is needed before translating microbiome research into preventive and therapeutic strategies to reduce gastric cancer burden.
Collapse
Affiliation(s)
- Joana Pereira-Marques
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135, Porto, Portugal; Ipatimup - Institute of Molecular Pathology and Immunology of the University of Porto, Rua Júlio Amaral de Carvalho 45, 4200-135, Porto, Portugal.
| | - Rui M Ferreira
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135, Porto, Portugal; Ipatimup - Institute of Molecular Pathology and Immunology of the University of Porto, Rua Júlio Amaral de Carvalho 45, 4200-135, Porto, Portugal.
| | - Jose C Machado
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135, Porto, Portugal; Ipatimup - Institute of Molecular Pathology and Immunology of the University of Porto, Rua Júlio Amaral de Carvalho 45, 4200-135, Porto, Portugal; Department of Pathology, Faculty of Medicine of the University of Porto, Alameda Prof. Hernâni Monteiro, 4200 - 319, Porto, Portugal.
| | - Ceu Figueiredo
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135, Porto, Portugal; Ipatimup - Institute of Molecular Pathology and Immunology of the University of Porto, Rua Júlio Amaral de Carvalho 45, 4200-135, Porto, Portugal; Department of Pathology, Faculty of Medicine of the University of Porto, Alameda Prof. Hernâni Monteiro, 4200 - 319, Porto, Portugal.
| |
Collapse
|
26
|
Sung JJY, Coker OO, Chu E, Szeto CH, Luk STY, Lau HCH, Yu J. Gastric microbes associated with gastric inflammation, atrophy and intestinal metaplasia 1 year after Helicobacter pylori eradication. Gut 2020; 69:1572-1580. [PMID: 31974133 PMCID: PMC7456733 DOI: 10.1136/gutjnl-2019-319826] [Citation(s) in RCA: 148] [Impact Index Per Article: 29.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Revised: 12/03/2019] [Accepted: 12/14/2019] [Indexed: 12/12/2022]
Abstract
OBJECTIVE Helicobacter pylori is associated with gastric inflammation, precancerous gastric atrophy (GA) and intestinal metaplasia (IM). We aimed to identify microbes that are associated with progressive inflammation, GA and IM 1 year after H. pylori eradication. DESIGN A total of 587 H. pylori-positive patients were randomised to receive H. pylori eradication therapy (295 patients) or placebo (292 patients). Bacterial taxonomy was analysed on 404 gastric biopsy samples comprising 102 pairs before and after 1 year H. pylori eradication and 100 pairs before and after 1 year placebo by 16S rRNA sequencing. RESULTS Analysis of microbial sequences confirmed the eradication of H. pylori in treated group after 1 year. Principal component analysis revealed distinct microbial clusters reflected by increase in bacterial diversity (p<0.00001) after H. pylori eradication. While microbial interactions remained largely unchanged after placebo treatment, microbial co-occurrence was less in treated group. Acinetobacter lwoffii, Streptococcus anginosus and Ralstonia were enriched while Roseburia and Sphingomonas were depleted in patients with persistent inflammation 1 year after H. pylori eradication. A distinct cluster of oral bacteria comprising Peptostreptococcus, Streptococcus, Parvimonas, Prevotella, Rothia and Granulicatella were associated with emergence and persistence of GA and IM. Probiotic Faecalibacterium praustznii was depleted in subjects who developed GA following H. pylori eradication. Functional pathways including amino acid metabolism and inositol phosphate metabolism were enriched while folate biosynthesis and NOD-like receptor signalling decreased in atrophy/IM-associated gastric microbiota. CONCLUSION This study identified that gastric microbes contribute to the progression of gastric carcinogenesis after H. pylori eradication.
Collapse
Affiliation(s)
- Joseph J Y Sung
- Department of Medicine and Therapeutics, Chinese University of Hong Kong, New Territories, Hong Kong
| | - Olabisi Oluwabukola Coker
- Department of Medicine and Therapeutics, Chinese University of Hong Kong, New Territories, Hong Kong
| | - Eagle Chu
- Department of Medicine and Therapeutics, Chinese University of Hong Kong, New Territories, Hong Kong
| | - Chun Ho Szeto
- Department of Medicine and Therapeutics, Chinese University of Hong Kong, New Territories, Hong Kong
| | - Simson Tsz Yat Luk
- Department of Medicine and Therapeutics, Chinese University of Hong Kong, New Territories, Hong Kong
| | - Harry Cheuk Hay Lau
- Department of Medicine and Therapeutics, Chinese University of Hong Kong, New Territories, Hong Kong
| | - Jun Yu
- Department of Medicine and Therapeutics, Chinese University of Hong Kong, New Territories, Hong Kong
| |
Collapse
|
27
|
Ford AC, Yuan Y, Forman D, Hunt R, Moayyedi P. Helicobacter pylori eradication for the prevention of gastric neoplasia. Cochrane Database Syst Rev 2020; 7:CD005583. [PMID: 32628791 PMCID: PMC7389270 DOI: 10.1002/14651858.cd005583.pub3] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
BACKGROUND Gastric cancer is the third most common cause of cancer death worldwide. Individuals infected with Helicobacter pylori have a higher likelihood of developing gastric cancer than individuals who are not infected. Eradication of H. pylori in healthy asymptomatic individuals in the general population may reduce the incidence of gastric cancer, but the magnitude of this effect is unclear. OBJECTIVES To assess the effectiveness of eradication of H. pylori in healthy asymptomatic individuals in the general population in reducing the incidence of gastric cancer. SEARCH METHODS We identified trials by searching the Cochrane Central Register of Controlled Trials (CENTRAL; 2020, Issue 1), MEDLINE (1946 to February 2020), and EMBASE (1974 to February 2020). We handsearched reference lists from trials selected by electronic searching to identify further relevant trials. We handsearched published abstracts from conference proceedings from the United European Gastroenterology Week (published in Gut) and Digestive Disease Week (published in Gastroenterology) between 2001 and 2019. We contacted members of the Cochrane Upper Gastrointestinal and Pancreatic Diseases Review Group and experts in the field and asked them to provide details of outstanding clinical trials and any relevant unpublished materials. SELECTION CRITERIA We analysed randomised controlled trials comparing at least one week of H. pylori therapy with placebo or no treatment in preventing subsequent development of gastric cancer in otherwise healthy and asymptomatic H. pylori-positive adults. Trials had to follow up participants for at least two years and needed to have at least two participants with gastric cancer as an outcome. We defined gastric cancer as any gastric adenocarcinoma, including intestinal (differentiated) or diffuse (undifferentiated) type, with or without specified histology. DATA COLLECTION AND ANALYSIS We collected data on incidence of gastric cancer, incidence of oesophageal cancer, deaths from gastric cancer, deaths from any cause, and adverse effects arising due to therapy. MAIN RESULTS Six trials met all our eligibility criteria and provided extractable data in the previous version. Following our updated search, one new RCT was identified, meaning that seven trials were included in this updated review. In addition, one previously included trial provided fully published data out to 10 years, and another previously included trial provided fully published data out to 22 years of follow-up. Four trials were at low risk of bias, one trial was at unclear risk, and two trials were at high risk of bias. Six trials were conducted in Asian populations. In preventing development of subsequent gastric cancer, H. pylori eradication therapy was superior to placebo or no treatment (RR 0.54, 95% confidence interval (CI) 0.40 to 0.72, 7 trials, 8323 participants, moderate certainty evidence). Only two trials reported the effect of eradication of H. pylori on the development of subsequent oesophageal cancer. Sixteen (0.8%) of 1947 participants assigned to eradication therapy subsequently developed oesophageal cancer compared with 13 (0.7%) of 1941 participants allocated to placebo (RR 1.22, 95% CI 0.59 to 2.54, moderate certainty evidence). H. pylori eradication reduced mortality from gastric cancer compared with placebo or no treatment (RR 0.61, 95% CI 0.40 to 0.92, 4 trials, 6301 participants, moderate certainty evidence). There was little or no evidence in all-cause mortality (RR 0.97, 95% CI 0.85 to 1.12, 5 trials, 7079 participants, moderate certainty evidence). Adverse events data were poorly reported. AUTHORS' CONCLUSIONS We found moderate certainty evidence that searching for and eradicating H. pylori reduces the incidence of gastric cancer and death from gastric cancer in healthy asymptomatic infected Asian individuals, but we cannot necessarily extrapolate this data to other populations.
Collapse
Affiliation(s)
- Alexander C Ford
- Leeds Gastroenterology Unit, St. James's University Hospital, Leeds, UK
| | - Yuhong Yuan
- Department of Medicine, Division of Gastroenterology, McMaster University, Hamilton, Canada
| | - David Forman
- International Agency for Research on Cancer, Lyon, France
| | - Richard Hunt
- Department of Medicine, Division of Gastroenterology, McMaster University, Hamilton, Canada
| | - Paul Moayyedi
- Department of Medicine, Division of Gastroenterology, McMaster University, Hamilton, Canada
| |
Collapse
|
28
|
Seo EH, Kim HJ, Kim JH, Lim B, Park JL, Kim SY, Lee SI, Jeong HY, Song KS, Kim YS. ONECUT2 upregulation is associated with CpG hypomethylation at promoter-proximal DNA in gastric cancer and triggers ACSL5. Int J Cancer 2020; 146:3354-3368. [PMID: 32129880 PMCID: PMC7217064 DOI: 10.1002/ijc.32946] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2019] [Revised: 02/16/2020] [Accepted: 02/19/2020] [Indexed: 12/14/2022]
Abstract
Many studies have focused on global hypomethylation or hypermethylation of tumor suppressor genes, but less is known about the impact of promoter hypomethylation of oncogenes. We previously showed that promoter methylation may gradually increase or decrease during the transition from gastric mucosa (GM) to intestinal metaplasia (IM) to gastric cancer (GC). In our study, we focused on regional CpG hypomethylation of the promoter‐proximal DNA of the transcription factor ONECUT2 (OC2) in IM and GC cells. We validated the hypomethylation of promoter‐proximal DNA of OC2 in 160 primary GCs, in which methylation level correlated negatively with OC2 mRNA level. IM and GC cells stained positively for OC2, whereas GM cells did not. Stable transfection of OC2 in GC cells promoted colony formation, cell migration, invasion and proliferation. Moreover, OC2 knockdown with a short hairpin RNA suppressed tumorigenesis in nude mice. In addition, chromatin immunoprecipitation coupled with DNA sequencing and RNA‐seq analyses revealed that OC2 triggered ACSL5, which is strongly expressed in IM of the stomach but not in GM, indicating that OC2 and ACSL5 are early‐stage biomarkers for GC. We also observed a high correlation between the levels of OC2 and ACSL5 mRNAs in the GENT database These results suggest that epigenetic alteration of OC2 upregulates its expression, which then activates ACSL5; thus, OC2 is induced in IM by epigenetic alteration and triggers ACSL5 expression, and thus OC2 and ACSL5 may cooperatively promote intestinal differentiation and GC progression. What's new? DNA hypomethylation can promote cancer development through activation of genes with oncogenic potential. Here, the authors found that CpGs in the promoter‐proximal DNA of ONECUT2 were hypomethylated in intestinal metaplasia and gastric cancers, and that hypomethylation was associated with ONECUT2 upregulation. Functional analysis demonstrated that ONECUT2 has oncogenic potential and could activate ACSL5, which is also expressed in intestinal metaplasia, suggesting that ONECUT2 and ACSL5 may cooperate to promote intestinal differentiation or development of gastric cancer. Taken together, the findings suggest that ONECUT2 and its downstream target ACSL5 could be used to develop early detection biomarkers and prevent gastric carcinogenesis.
Collapse
Affiliation(s)
- Eun-Hye Seo
- Genome Editing Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Republic of Korea.,Department of Functional Genomics, Korea University of Science and Technology, Daejeon, Republic of Korea
| | - Hee-Jin Kim
- Genome Editing Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Republic of Korea
| | - Jong-Hwan Kim
- Personalized Genomic Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Republic of Korea
| | - Byungho Lim
- Division of Drug Discovery Research, Research Center for Drug Discovery Technology, Korea Research Institute of Chemical Technology, Daejeon, Republic of Korea
| | - Jong-Lyul Park
- Genome Editing Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Republic of Korea
| | - Seon-Young Kim
- Department of Functional Genomics, Korea University of Science and Technology, Daejeon, Republic of Korea.,Personalized Genomic Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Republic of Korea
| | - Sang-Il Lee
- Department of Surgery, College of Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Hyun-Yong Jeong
- Department of Internal Medicine, College of Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Kyu-Sang Song
- Department of Pathology, College of Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Yong-Sung Kim
- Genome Editing Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Republic of Korea.,Department of Functional Genomics, Korea University of Science and Technology, Daejeon, Republic of Korea
| |
Collapse
|
29
|
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
|
30
|
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]
|
31
|
Basen-Engquist K, Brown P, Coletta AM, Savage M, Maresso KC, Hawk E. Lifestyle and Cancer Prevention. ABELOFF'S CLINICAL ONCOLOGY 2020:337-374.e12. [DOI: 10.1016/b978-0-323-47674-4.00022-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
32
|
Terasawa T, Hamashima C, Kato K, Miyashiro I, Yoshikawa T, Takaku R, Nishida H. Helicobacterpylori eradication treatment for gastric carcinoma prevention in asymptomatic or dyspeptic adults: systematic review and Bayesian meta-analysis of randomised controlled trials. BMJ Open 2019; 9:e026002. [PMID: 31542733 PMCID: PMC6756423 DOI: 10.1136/bmjopen-2018-026002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Revised: 07/29/2019] [Accepted: 09/02/2019] [Indexed: 12/11/2022] Open
Abstract
OBJECTIVES Recent meta-analyses of eradication therapy in Helicobacter pylori-infected adults reported significant reductions in gastric carcinoma risk. However, concerns about supporting unfocused screening and eradication programme in healthy, asymptomatic populations have arisen. We performed a systematic review and Bayesian meta-analysis to provide an accurate interpretation of randomised evidence on the preventive effectiveness of eradication therapy on gastric carcinoma risk. METHODS We searched databases including PubMed, Cochrane Central and Embase for reference and citation tracking without language restrictions, from inception through 31 July 2018. Paired investigators independently selected randomised controlled trials (RCTs) comparing eradication therapy with placebo or no treatment for asymptomatic or dyspeptic H. pylori-infected adults with no previous gastric carcinoma. The main outcome was gastric carcinoma incidence; secondary outcomes included gastric carcinoma-specific, non-gastric carcinoma and all-cause mortality. RESULTS A total of 5 population-based and 2 outpatient care-based RCTs involving 7303 adults were eligible. Eradication algorithms were heterogeneous, and unsuccessful eradication and reinfection were frequently observed. A Bayesian meta-analysis with competing risk outcomes found low-certainty evidence that eradication therapy might be more likely than control to reduce gastric carcinoma risk (HR=0.65; 95% credible interval (CrI) 0.41 to 1.0; I2 =11%). The CrIs included the null effects across the subgroup and sensitivity analyses, apart from those based on particular models that excluded two RCTs that enrolled subjects with specific histological findings only (HR=0.55; CrI 0.30 to 0.89; I2 =14%). The uncertainty of the average 41% risk reduction in gastric carcinoma-specific mortality included a clinically important mortality risk increase (HR=0.59 favouring eradication therapy; CrI 0.25 to 1.20; I2 =13%; low certainty). CONCLUSIONS There is insufficient evidence to support or refute the effectiveness of eradication therapy in preventing gastric carcinoma in H. pylori-infected, high-risk populations. Rigorously conducted large RCTs of healthy infected adults only would provide evidence of the true efficacy of successful eradication. PROSPERO registration number: CRD42014009245.
Collapse
Affiliation(s)
- Teruhiko Terasawa
- Emergency and General Internal Medicine, Fujita Health University, Toyoake, Japan
| | - Chisato Hamashima
- Department of Nursing, Faculty of Medical Technology, Teikyo University, Tokyo, Japan
| | - Katsuaki Kato
- Cancer Detection Center, Miyagi Cancer Society, Sendai, Japan
| | - Isao Miyashiro
- Department of Cancer Registration and Survey, Osaka Medical Center for Cancer and Cardiovascular Diseases, Osaka, Japan
| | - Takaki Yoshikawa
- Department of Gastrointestinal Surgery, Kanagawa Cancer Center, Yokohama, Japan
| | - Reo Takaku
- Institute for Health Economics and Policy, Tokyo, Japan
| | | |
Collapse
|
33
|
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
|
34
|
Alboraie M, Elhossary W, Aly OA, Abbas B, Abdelsalam L, Ghaith D, Shady Z, Gaber Y, Adel E, Peura D, Armstrong D, Esmat G. Egyptian recommendations for management of Helicobacter pylori infection: 2018 report. Arab J Gastroenterol 2019; 20:175-179. [PMID: 31564518 DOI: 10.1016/j.ajg.2019.09.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Accepted: 09/12/2019] [Indexed: 02/07/2023]
Affiliation(s)
- Mohamed Alboraie
- Department of Internal Medicine, Al-Azhar University, Cairo, Egypt.
| | - Walied Elhossary
- Endemic Medicine and Hepatology Department, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Osama Aboelfotoh Aly
- General Medicine, Gastroenterology & Hepatology Department, Faculty of Medicine, Ain Shams University, Egypt
| | - Bahaa Abbas
- Gastroenterology Department, Air Force Specialized Hospital, Military Medical Academy, Egypt
| | | | - Doaa Ghaith
- Clinical and Chemical Pathology Department, Faculty of Medicine, Cairo University, Egypt
| | - Zakarya Shady
- Department of Internal Medicine, Al-Azhar University, Cairo, Egypt
| | - Yasmine Gaber
- Endemic Medicine and Hepatology Department, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Eman Adel
- Department of Anatomy and Embryology, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - David Peura
- Department of Medicine, University of Virginia, School of Medicine, Charlottesville, VA, USA
| | | | - Gamal Esmat
- Endemic Medicine and Hepatology Department, Faculty of Medicine, Cairo University, Cairo, Egypt
| |
Collapse
|
35
|
Kim N. Chemoprevention of gastric cancer by Helicobacter pylori eradication and its underlying mechanism. J Gastroenterol Hepatol 2019; 34:1287-1295. [PMID: 30828872 DOI: 10.1111/jgh.14646] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2019] [Revised: 02/23/2019] [Accepted: 03/01/2019] [Indexed: 12/11/2022]
Abstract
The cascade of gastric cancer, a leading cause of cancer incidence and mortality, is multifactorial. Helicobacter pylori (HP) infection plays a major role in gastric cancer (GC), and there has been an accumulation of data regarding the chemopreventive effect of HP eradication. However, it remains unclear how HP infection causes GC and how HP eradication prevents GC. To clarify this issue, the following approaches were performed in this review article. First, how HP-induced atrophic gastritis (AG) and intestinal metaplasia (IM) provoke the development of GC is shown, followed by how long HP eradication takes to induce a reversible change in AG and IM. Second, epigenetic studies of PTPN6, MOS, DCC, CRK, and VAV1 were performed in noncancerous gastric specimens in terms of HP status. Among these genes, MOS was found to be a possible surrogate marker for GC development. HP eradication decreased aberrant DNA methylation in a gene-specific manner, and MOS played a role in metachronous gastric neoplasms. Third, transforming growth factor-β1 (TGF-β1) and TGF-β1-induced epithelial-mesenchymal transition (EMT) markers were investigated in gastric mucosa. HP infection triggered the TGF-β1-induced EMT pathway and caused the emergence of GC stem cells, such as CD44v8-10. When HP was eradicated, these two pathways were inhibited. Finally, a 2222 cohort study showed that HP eradication significantly decreased the risk of noncardiac GC. Taken together, HP eradication is effective as a primary GC prevention method, and its underlying mechanism includes reversibility of AG and IM, methylation, EMT, and stem cells.
Collapse
Affiliation(s)
- Nayoung Kim
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, South Korea.,Department of Internal Medicine and Liver Research Institute, Seoul National University College of Medicine, Seoul, South Korea.,Tumor Microenvironment Global Core Research Center, Seoul National University, Seoul, South Korea
| |
Collapse
|
36
|
Kuo HY, Chang WL, Yeh YC, Cheng HC, Tsai YC, Wu CT, Lin SH, Yang HB, Lu CC, Sheu BS. Spasmolytic polypeptide-expressing metaplasia associated with higher expressions of miR-21, 155, and 223 can be regressed by Helicobacter pylori eradication in the gastric cancer familial relatives. Helicobacter 2019; 24:e12578. [PMID: 30990573 DOI: 10.1111/hel.12578] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2018] [Revised: 01/19/2019] [Accepted: 02/07/2019] [Indexed: 12/12/2022]
Abstract
BACKGROUND AND AIMS Spasmolytic polypeptide-expressing metaplasia (SPEM) is a preneoplastic gastric cancer lesion related to epigenetic microRNA (miRNA) expression. This study elucidated whether Helicobacter pylori-infected first-degree relatives of patients with gastric cancer (GCF) are susceptible to have SPEM and correlated with miR-21, 155, and 223 expressions. We also validated whether SPEM and these miRNAs can be regressed after H pylori eradication. METHODS We prospectively enrolled 148 GCF and 148 nonulcer dyspepsia (NUD) subjects without gastric cancer familial history as controls. Each case had received a panendoscopy to determine H pylori status and gastric histology, including SPEM. The cases with SPEM were followed after H pylori eradication to determine SPEM regression. The total RNA was extracted to analyze tissues miR-21, 155, and 223 before and after eradication. RESULTS GCF subjects had a higher prevalence of H pylori infection (73% vs 32%) and SPEM (42% vs 14%, P < 0.01) than controls. The tissue miR-21, 155, and 223 in antrum were higher in cases with SPEM than in those without SPEM (P <= 0.05). There was similar SPEM reversibility after H pylori eradication between GCF subjects and controls (72% vs 69%, P = 0.852). In the SPEM regressed cases, tissue miR-21, 155, and 223 decreased after H pylori eradication (P < 0.05). CONCLUSION The H pylori-infected GCF subjects were prone to have SPEM with higher tissues miR-21, 155, and 223 expressions. H pylori eradication can result in a 70% SPEM regression, accompanied by a decline in miR-21, 155, and 233 expression levels.
Collapse
Affiliation(s)
- Hsin-Yu Kuo
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University Hospital, National Cheng Kung University, Tainan, Taiwan.,Department of Internal Medicine, College of Medicine, National Cheng Kung University Hospital, National Cheng Kung University, Tainan, Taiwan
| | - Wei-Lun Chang
- Department of Internal Medicine, College of Medicine, National Cheng Kung University Hospital, National Cheng Kung University, Tainan, Taiwan
| | - Yi-Chun Yeh
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University Hospital, National Cheng Kung University, Tainan, Taiwan
| | - Hsiu-Chi Cheng
- Department of Internal Medicine, College of Medicine, National Cheng Kung University Hospital, National Cheng Kung University, Tainan, Taiwan
| | - Yu-Ching Tsai
- Department of Internal Medicine, College of Medicine, National Cheng Kung University Hospital, National Cheng Kung University, Tainan, Taiwan.,Department of Internal Medicine, Tainan Hospital, Department of Health, Ministry of Welfare and Health, Tainan, Taiwan
| | - Chung-Tai Wu
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University Hospital, National Cheng Kung University, Tainan, Taiwan.,Department of Internal Medicine, College of Medicine, National Cheng Kung University Hospital, National Cheng Kung University, Tainan, Taiwan
| | - Sheng-Hsiang Lin
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University Hospital, National Cheng Kung University, Tainan, Taiwan
| | - Hsiao-Bai Yang
- Department of Pathology, College of Medicine, National Cheng Kung University Hospital, National Cheng Kung University, Tainan, Taiwan.,Department of Pathology, Ton-Yen General Hospital, Hsin-chu, Taiwan
| | - Cheng-Chang Lu
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University Hospital, National Cheng Kung University, Tainan, Taiwan.,Department of Pathology, College of Medicine, National Cheng Kung University Hospital, National Cheng Kung University, Tainan, Taiwan
| | - Bor-Shyang Sheu
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University Hospital, National Cheng Kung University, Tainan, Taiwan.,Department of Internal Medicine, College of Medicine, National Cheng Kung University Hospital, National Cheng Kung University, Tainan, Taiwan.,Department of Internal Medicine, Tainan Hospital, Department of Health, Ministry of Welfare and Health, Tainan, Taiwan
| |
Collapse
|
37
|
Kwon YJ, Kim N, Baek SM, Lee HS, Lee J, Hwang YJ, Yoon H, Shin CM, Park YS, Kim JW, Lee DH. The prevalence of histologic atrophy and intestinal metaplasia in the corpus has decreased over 15 years in females in the Korean population. Helicobacter 2019; 24:e12579. [PMID: 30920087 DOI: 10.1111/hel.12579] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/26/2018] [Revised: 02/12/2019] [Accepted: 02/13/2019] [Indexed: 12/19/2022]
Abstract
BACKGROUND The aim of this study was to investigate the trends of atrophy and intestinal metaplasia (IM) in 2002 subjects without significant gastroduodenal diseases. MATERIALS AND METHODS A total of 2002 subjects were prospectively enrolled and divided into three periods (2003-2007, 2008-2012, and 2013-2018). Trends of H pylori and atrophy/IM scored by Updated Sydney System were analyzed according to sex, and multivariate logistic analysis was performed for the risk factors for atrophy/IM. RESULTS H pylori-negative and H pylori-positive subjects were 1220 (61.0%) and 782 (38.0%), respectively. H pylori positivity decreased from 149/303 (49.2%), 207/515 (40.2%) and 426/1184 (36.0%), in the three periods, respectively (P < 0.001). The prevalence of atrophy (P < 0.001) and IM in the corpus (P < 0.001) significantly decreased over 15 years in females, but not in males. The mean grade of atrophy and IM was higher in males (0.36 and 0.51) than in females (0.28 and 0.41) in the corpus (P = 0.027) and in the antrum (P = 0.006), respectively. Similarly, the mean grade of IM in males (0.34) was higher in females (0.19; P < 0.001) in the corpus. Multivariate analysis showed that old age, study period, and H pylori were statistically significant in atrophy of antrum and corpus, and IM in the corpus. In cases of IM of antrum, old age, H pylori, and smoking were statistically significant. CONCLUSION A significant decrease in atrophy and IM in the corpus in females over 15 years suggests sex- or gender-specific characteristics.
Collapse
Affiliation(s)
- Young Jae Kwon
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Republic of Korea
| | - Nayoung Kim
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Republic of Korea.,Department of Internal Medicine and Liver Research Institute, Seoul National University, Seoul, Republic of Korea.,Tumor Microenvironment Global Core Research Center, Seoul National University, Seoul, South Korea
| | - Sung Min Baek
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Republic of Korea
| | - Hye Seung Lee
- Department of Pathology, Seoul National University Bundang Hospital, Seongnam, Republic of Korea
| | - Jaebong Lee
- Division of Statistics in Medical Research Collaborating Center, Seoul National University Bundang Hospital, Seongnam, Republic of Korea
| | - Young Jae Hwang
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Republic of Korea
| | - Hyuk Yoon
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Republic of Korea.,Tumor Microenvironment Global Core Research Center, Seoul National University, Seoul, South Korea
| | - Cheol Min Shin
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Republic of Korea.,Tumor Microenvironment Global Core Research Center, Seoul National University, Seoul, South Korea
| | - Young Soo Park
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Republic of Korea.,Tumor Microenvironment Global Core Research Center, Seoul National University, Seoul, South Korea
| | - Jin-Wook Kim
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Republic of Korea.,Department of Internal Medicine and Liver Research Institute, Seoul National University, Seoul, Republic of Korea
| | - Dong Ho Lee
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Republic of Korea.,Department of Internal Medicine and Liver Research Institute, Seoul National University, Seoul, Republic of Korea.,Tumor Microenvironment Global Core Research Center, Seoul National University, Seoul, South Korea
| |
Collapse
|
38
|
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
|
39
|
Chen F, Liu Y, Tsay A, McAllister BP, Karamchandani DM. Hit or a miss: Concordance between histopathologic-endoscopic findings in gastric mucosal biopsies. Ann Diagn Pathol 2019; 38:106-114. [DOI: 10.1016/j.anndiagpath.2018.12.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Revised: 11/30/2018] [Accepted: 12/13/2018] [Indexed: 12/21/2022]
|
40
|
Khatoon J, Prasad KN, Rai RP, Shukla SK, Krishnani N, Ghoshal UC. Expression levels of A disintegrin and metalloproteases (ADAMs), and Th17-related cytokines and their association with Helicobacter pylori infection in patients with gastroduodenal diseases. Pathog Dis 2018; 76:5145580. [PMID: 30371773 DOI: 10.1093/femspd/fty078] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Accepted: 10/25/2018] [Indexed: 02/06/2023] Open
Abstract
Expression levels of A disintegrin and metalloproteases (ADAMs) (10 and 17) and Th17-related cytokines [interleukin (IL) 17A, IL-17F, IL-33, IL-23, IL-23R] were investigated by quantitative real time polymerase chain reaction in gastric biopsies of patients with different gastroduodenal pathologies in the presence and absence of Helicobacter pylori infection. Patients with gastric cancer (GC) (n = 70, intestinal-type 38 and diffuse type 32), peptic ulcer disease [n = 50, duodenal ulcer (DU) 16 and gastric ulcer (GU) 34] and functional dyspepsia (n = 120) were included in the study. Further, the expression levels of ADAMs and Th17 cytokines were correlated with H. pylori cytotoxin-associated genes pathogenicity island (cagPAI) status. Expression levels of ADAMs (10 and 17) and Th17-related cytokines (IL-17A, IL-23, IL-23R) were significantly higher in H. pylori-positive than in H. pylori-negative gastric biopsies. Significant increase in ADAM17 and Th17 cytokines (IL-17A and IL-23) expressions was observed in patients with GU and intestinal-type GC in the presence of H. pylori infection and in strains harbouring intact cagPAI. Expression levels of IL-17A, IL-23 and ADAM17 were strongly correlated with GU and intestinal-type GC and weakly with DU and diffuse-type GC in the presence of H. pylori infection. Higher expression levels of ADAM17 and Th17 cytokines (IL-17A and IL-23), and their strong correlation with GU and intestinal-type GC patients in the presence of H. pylori and its intact cagPAI status, suggest a possible role of strain specificity in the pathogenesis of these diseases.
Collapse
Affiliation(s)
- Jahanarah Khatoon
- Department of Microbiology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow (U.P.) 226014, India
| | - Kashi Nath Prasad
- Department of Microbiology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow (U.P.) 226014, India
| | - Ravi Prakash Rai
- Department of Microbiology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow (U.P.) 226014, India
| | - Sanket Kumar Shukla
- Department of Medicine, Center of Translational Medicine, Thomas Jefferson University, Philadelphia, PA-19107 USA
| | - Narendra Krishnani
- Department of Pathology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow (U.P.) 226014, India
| | - Uday Chand Ghoshal
- Department of Gastroenterology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow (U.P.) 226014, India
| |
Collapse
|
41
|
Zhang G, Li J, Li S, Wang Y. Exploring Spatial Trends and Influencing Factors for Gastric Cancer Based on Bayesian Statistics: A Case Study of Shanxi, China. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2018; 15:E1824. [PMID: 30142954 PMCID: PMC6165541 DOI: 10.3390/ijerph15091824] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Revised: 08/13/2018] [Accepted: 08/22/2018] [Indexed: 02/06/2023]
Abstract
Gastric cancer (GC) is the fourth most common type of cancer and the second leading cause of cancer-related deaths worldwide. To detect the spatial trends of GC risk based on hospital-diagnosed patients, this study presented a selection probability model and integrated it into the Bayesian spatial statistical model. Then, the spatial pattern of GC risk in Shanxi Province in north central China was estimated. In addition, factors influencing GC were investigated mainly using the Bayesian Lasso model. The spatial variability of GC risk in Shanxi has the conspicuous feature of being 'high in the south and low in the north'. The highest GC relative risk was 1.291 (95% highest posterior density: 0.789⁻4.002). The univariable analysis and Bayesian Lasso regression results showed that a diverse dietary structure and increased consumption of beef and cow milk were significantly (p ≤ 0.08) and in high probability (greater than 68%) negatively associated with GC risk. Pork production per capita has a positive correlation with GC risk. Moreover, four geographic factors, namely, temperature, terrain, vegetation cover, and precipitation, showed significant (p < 0.05) associations with GC risk based on univariable analysis, and associated with GC risks in high probability (greater than 60%) inferred from Bayesian Lasso regression model.
Collapse
Affiliation(s)
- Gehong Zhang
- Medical Imaging Department, Shanxi Medical University, Taiyuan 030001, Shanxi, China.
| | - Junming Li
- School of Statistics, Shanxi University of Finance and Economics, Taiyuan 030006, Shanxi, China.
| | - Sijin Li
- Medical Imaging Department, Shanxi Medical University, Taiyuan 030001, Shanxi, China.
| | - Yang Wang
- Medical Imaging Department, Shanxi Medical University, Taiyuan 030001, Shanxi, China.
| |
Collapse
|
42
|
Shams AZ, Haug U. Strategies for prevention of gastrointestinal cancers in developing countries: a systematic review. J Glob Health 2018; 7:020405. [PMID: 29250323 PMCID: PMC5718709 DOI: 10.7189/jogh.07.020405] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Background Gastrointestinal cancers account for one third of total cancer incidence and mortality in developing countries. To date, there is no systematic synthesis of evidence regarding strategies to prevent gastrointestinal cancers in developing countries. We aimed to provide a systematic overview of studies evaluating strategies for prevention or early detection of the three most common gastrointestinal cancers (gastric, liver and colorectal cancer) in developing countries. Methods We searched MEDLINE, Web of Science and WHO Global Index Medicus databases for relevant articles published until October 2016 using combinations of the search terms “gastrointestinal”, “digestive system”, “gastric”, “liver”, “colorectal”, “cancer”, “prevention”, “early detection” and “developing country” (including names). Results Overall, 73 articles met the inclusion criteria, providing information on short– and long–term outcomes (up to 30 years) from various intervention studies (∼45% randomized). Trials on hepatitis B vaccination consistently showed vaccine efficacy over time and indicated long–term preventive effects on liver cancer incidence that start to become measurable at the population level. Studies on anti–H. pylori treatment suggested a reduction in gastric cancer incidence reaching statistical significance after long–term follow–up, while evidence regarding a preventive effect in persons with precancerous lesions is still inconclusive. The studies regarding colorectal cancer focused on early detection, ∼90% of which were restricted to intermediate endpoints. Conclusion In conclusion, there were a number of studies on gastric and liver cancer prevention in developing countries showing promising results after long–term follow–up. Important next steps include pooled meta–analyses as far as possible given the heterogeneity between studies as well as implementation research.
Collapse
Affiliation(s)
- Ahmad Zia Shams
- Epidemiological Cancer Registry Baden-Wuerttemberg, German Cancer Research Centre, Heidelberg, Germany.,Department of Clinical Epidemiology, Leibniz Institute for Prevention Research and Epidemiology, Bremen, Germany
| | - Ulrike Haug
- Department of Clinical Epidemiology, Leibniz Institute for Prevention Research and Epidemiology, Bremen, Germany.,Faculty of Human and Health Sciences, University of Bremen, Bremen, Germany
| |
Collapse
|
43
|
Hwang YJ, Kim N, Lee HS, Lee JB, Choi YJ, Yoon H, Shin CM, Park YS, Lee DH. Reversibility of atrophic gastritis and intestinal metaplasia after Helicobacter pylori eradication - a prospective study for up to 10 years. Aliment Pharmacol Ther 2018; 47:380-390. [PMID: 29193217 DOI: 10.1111/apt.14424] [Citation(s) in RCA: 117] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2017] [Revised: 07/31/2017] [Accepted: 10/27/2017] [Indexed: 12/12/2022]
Abstract
BACKGROUND Atrophic gastritis and intestinal metaplasia are premalignant conditions for gastric cancer. Their reversibility by Helicobacter pylori eradication remains controversial. AIM To evaluate the reversibility of atrophic gastritis and intestinal metaplasia by H. pylori eradication with long-term follow-up. METHODS 598 subjects were prospectively enrolled and followed for up to 10 years. They were categorised as H. pylori-negative (n = 65), H. pylori non-eradicated (n = 91), and H. pylori-eradicated (n = 442). Histological assessment was performed for antrum and corpus by Sydney classification. RESULTS Histological follow-up was performed regularly at 1, 2, 3-4 and ≥5 years, with mean follow-up of 1.07 ± 0.21, 2.29 ± 0.83, 3.93 ± 1.02, and 6.45 ± 1.28 years, respectively. Atrophic gastritis in antrum and corpus gradually and significantly (both P < .05 for all timepoints) improved only in the H. pylori-eradicated group compared to that at baseline. Significant difference in atrophic gastritis between H. pylori-eradicated and H. pylori-negative groups disappeared from 1-year follow-up. Similarly, intestinal metaplasia in antrum and corpus improved significantly (both P < .05 for all timepoints) only in the H. pylori-eradicated group in comparison with that at baseline. Significant difference in intestinal metaplasia between H. pylori-eradicated and H. pylori-negative groups disappeared from ≥5 years of follow-up in the antrum and from 3 years of follow-up in the corpus. CONCLUSION H. pylori eradication may be a preventative strategy for intestinal-type gastric cancer by regression of atrophic gastritis and intestinal metaplasia.
Collapse
Affiliation(s)
- Y-J Hwang
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seoungnam, South Korea
| | - N Kim
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seoungnam, South Korea.,Department of Internal Medicine and Institute of Liver Research, Seoul National University College of Medicine, Seoul, South Korea
| | - H S Lee
- Department of Pathology, Seoul National University Bundang Hospital, Seoungnam, South Korea
| | - J B Lee
- Division of Statistics in Medical Research Collaborating Center, Seoul National University Bundang Hospital, Seoungnam, South Korea
| | - Y J Choi
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seoungnam, South Korea
| | - H Yoon
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seoungnam, South Korea
| | - C M Shin
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seoungnam, South Korea
| | - Y S Park
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seoungnam, South Korea
| | - D H Lee
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seoungnam, South Korea.,Department of Internal Medicine and Institute of Liver Research, Seoul National University College of Medicine, Seoul, South Korea
| |
Collapse
|
44
|
Hwang YJ, Kim N, Kim SE, Baik GH, Lee JY, Park KS, Joo YE, Myung DS, Kim HJ, Song HJ, Kim HU, Nam K, Shin JE, Kim HJ, Kim GH, Lee J, Lim SH, Seo GS, Choi SC. Change in the Prevalences and Risk Factors of Atrophic Gastritis and Intestinal Metaplasia in Korea: Multicenter Clinical Trials. THE KOREAN JOURNAL OF HELICOBACTER AND UPPER GASTROINTESTINAL RESEARCH 2018. [DOI: 10.7704/kjhugr.2018.18.4.247] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Young-Jae Hwang
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Nayoung Kim
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Sung Eun Kim
- Department of Internal Medicine, Kosin University College of Medicine, Busan, Korea
| | - Gwang Ho Baik
- Department of Internal Medicine, College of Medicine, Hallym University, Chuncheon, Korea
| | - Ju Yup Lee
- Department of Internal Medicine, Keimyung University School of Medicine, Daegu, Korea
| | - Kyung Sik Park
- Department of Internal Medicine, Keimyung University School of Medicine, Daegu, Korea
| | - Young-Eun Joo
- Department of Internal Medicine, Chonnam National University Medical School, Gwangju, Korea
| | - Dae-Seong Myung
- Department of Internal Medicine, Chonnam National University Medical School, Gwangju, Korea
| | - Hyeon Ju Kim
- Department of Family Medicine, Jeju National University School of Medicine, Jeju, Korea
| | - Hyun Joo Song
- Department of Internal Medicine, Jeju National University School of Medicine, Jeju, Korea
| | - Heung Up Kim
- Department of Internal Medicine, Jeju National University School of Medicine, Jeju, Korea
| | - Kwangwoo Nam
- Department of Internal Medicine, Dankook University College of Medicine, Cheonan, Korea
| | - Jeong Eun Shin
- Department of Internal Medicine, Dankook University College of Medicine, Cheonan, Korea
| | - Hyun Jin Kim
- Department of Internal Medicine and Institute of Health Science, Gyeongsang National University School of Medicine, Jinju, Korea
| | - Gwang Ha Kim
- Department of Internal Medicine, Pusan National University School of Medicine, Busan, Korea
| | - Jongchan Lee
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Seon Hee Lim
- Department of Internal Medicine, Seoul National University Healthcare System Gangnam Center and Healthcare Research Institute, Seoul, Korea
| | - Geom Seog Seo
- Department of Internal Medicine, Wonkwang University School of Medicine, Iksan, Korea
| | - Suck Chei Choi
- Department of Internal Medicine, Wonkwang University School of Medicine, Iksan, Korea
| |
Collapse
|
45
|
Hwang YJ, Kim N, Yun CY, Kwon MG, Baek SM, Kwon YJ, Lee HS, Lee JB, Choi YJ, Yoon H, Shin CM, Park YS, Lee DH. Predictive Factors for Improvement of Atrophic Gastritis and Intestinal Metaplasia: A Long-term Prospective Clinical Study. THE KOREAN JOURNAL OF HELICOBACTER AND UPPER GASTROINTESTINAL RESEARCH 2018. [DOI: 10.7704/kjhugr.2018.18.3.186] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Young-Jae Hwang
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Nayoung Kim
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Korea
- Department of Internal Medicine and Institute of Liver Research, Seoul National University College of Medicine, Seoul, Korea
| | - Chang Yong Yun
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Min Gu Kwon
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Sung Min Baek
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Yeong Jae Kwon
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Hye Seung Lee
- Department of Pathology, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Jae Bong Lee
- Division of Statistics in Medical Research Collaborating Center, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Yoon Jin Choi
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Hyuk Yoon
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Cheol Min Shin
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Young Soo Park
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Dong Ho Lee
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Korea
- Department of Internal Medicine and Institute of Liver Research, Seoul National University College of Medicine, Seoul, Korea
| |
Collapse
|
46
|
Nie Y, Wu K, Yu J, Liang Q, Cai X, Shang Y, Zhou J, Pan K, Sun L, Fang J, Yuan Y, You W, Fan D. A global burden of gastric cancer: the major impact of China. Expert Rev Gastroenterol Hepatol 2017; 11:651-661. [PMID: 28351219 DOI: 10.1080/17474124.2017.1312342] [Citation(s) in RCA: 84] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Gastric cancer (GC) is a highly aggressive cancer and a major cause of cancer-related deaths worldwide. Approximately half of the world's GC cases and deaths occur in china. GC presents challenges in early diagnosis and effective therapy due to a lack of understanding of the underlying molecular biology. The primary goals of this review are to outline current GC research in china and describe future trends in this field. Areas covered: This review mainly focuses on a series of GC-related advances China has achieved. Considerable progress has been made in understanding the role of H. pylori in GC by a series of population-based studies in well-established high-risk areas; A few germline and somatic alterations have been identified by 'omics' studies; Studies on the mechanisms of malignant phenotypes have helped us to form an in-depth understanding of GC and advance drug discovery. Moreover, identification of potential biomarkers and targeted therapies have facilitated the diagnosis and treatment of GC. However, many challenges remain. Expert commentary: To combat GC, sufficient funding is important. More attention should be paid on early diagnosis and the discovery of novel efficient biomarkers and the development of biomarker-based or targeted therapeutics in GC.
Collapse
Affiliation(s)
- Yongzhan Nie
- a State Key Laboratory of Cancer Biology and Xijing Hospital of Digestive Diseases , Fourth Military Medical University , Xi'an , China
| | - Kaichun Wu
- a State Key Laboratory of Cancer Biology and Xijing Hospital of Digestive Diseases , Fourth Military Medical University , Xi'an , China
| | - Jun Yu
- b Department of Medicine and Therapeutics and Institute of Digestive Disease , Chinese University of Hong Kong , Hong Kong , China
| | - Qiaoyi Liang
- b Department of Medicine and Therapeutics and Institute of Digestive Disease , Chinese University of Hong Kong , Hong Kong , China
| | - Xiqiang Cai
- a State Key Laboratory of Cancer Biology and Xijing Hospital of Digestive Diseases , Fourth Military Medical University , Xi'an , China
| | - Yulong Shang
- a State Key Laboratory of Cancer Biology and Xijing Hospital of Digestive Diseases , Fourth Military Medical University , Xi'an , China
| | - Jinfeng Zhou
- a State Key Laboratory of Cancer Biology and Xijing Hospital of Digestive Diseases , Fourth Military Medical University , Xi'an , China
| | - Kaifeng Pan
- c Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Cancer Epidemiology, Peking University School of Oncology , Peking University Cancer Hospital & Institute , Beijing , China
| | - Liping Sun
- d Tumor Etiology and Screening, Department of Cancer Institute and General Surgery , The First Affiliated Hospital of China Medical University , Shenyang , China
| | - Jingyuan Fang
- e Renji Hospital , Shanghai Jiao-Tong University School of Medicine , Shanghai , China
| | - Yuan Yuan
- d Tumor Etiology and Screening, Department of Cancer Institute and General Surgery , The First Affiliated Hospital of China Medical University , Shenyang , China
| | - Weicheng You
- c Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Cancer Epidemiology, Peking University School of Oncology , Peking University Cancer Hospital & Institute , Beijing , China
| | - Daiming Fan
- a State Key Laboratory of Cancer Biology and Xijing Hospital of Digestive Diseases , Fourth Military Medical University , Xi'an , China
| |
Collapse
|
47
|
Wang L, Lin Z, Chen S, Li J, Chen C, Huang Z, Ye B, Ding J, Li W, Wu L, Jiang Y, Meng L, Du Q, Si J. Ten-day bismuth-containing quadruple therapy is effective as first-line therapy for Helicobacter pylori –related chronic gastritis: a prospective randomized study in China. Clin Microbiol Infect 2017; 23:391-395. [DOI: 10.1016/j.cmi.2016.12.032] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2016] [Revised: 12/25/2016] [Accepted: 12/28/2016] [Indexed: 12/14/2022]
|
48
|
周 林, 赵 阳, 张 伟. 幽门螺杆菌根除治疗在胃癌预防中的现状及前景. Shijie Huaren Xiaohua Zazhi 2017; 25:1327-1337. [DOI: 10.11569/wcjd.v25.i15.1327] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
幽门螺杆菌(Helicobacter pylori, H. pylori)与胃癌(gastric cancer, GC)发生发展密切相关, 根除H. pylori可降低GC发生率. 经济学分析表明, 在高危人群中根除治疗H. pylori作为预防GC的策略是成本效益较好的. 即使在GC低风险人群中, H. pylori筛查和治疗也能使上消化道其他非恶性疾病获益. 然而, 广泛开展H. pylori根除治疗可能带来其他问题, 包括抗生素耐药增加和与H. pylori负相关疾病(如胃食管反流病、Barrett食管、哮喘和肥胖等)的患病率升高. 本文将结合近年最新研究, 就上述问题展开论述, 进一步提出H. pylori根除治疗预防GC所面临的挑战以及将来可能的进展方向.
Collapse
|
49
|
Abstract
Helicobacter pylori is an important human pathogen, associated with a substantial burden from both malignant and non-malignant diseases. The bacterium is classed as a human carcinogen, being strongly linked with gastric cancer, the third most common cause of cancer death worldwide and is also associated with common conditions such as dyspepsia and peptic ulcer. Eradication of H. pylori reduces the incidence of gastric cancer and peptic ulcer, as well as the prevalence and costs of managing dyspepsia. Economic analyses suggest that eradication of H. pylori as a means of controlling gastric cancer is cost-effective in high-risk populations. Even in populations at low risk of gastric cancer, there might be other benefits arising from screening and treatment, owing to the effects on non-malignant upper gastrointestinal diseases. However, public health authorities have been slow to consider the benefits of population-based screening and treatment as a means of reducing the morbidity and mortality associated with the infection. There are also concerns about widespread use of eradication therapy, including antimicrobial resistance and a rise in the prevalence of diseases that are negatively associated with H. pylori, such as GERD, Barrett oesophagus, asthma and obesity. This Review summarizes these issues.
Collapse
|
50
|
Abstract
Introduction Many clinicians consider chronic gastritis to be equivalent to Helicobacter pylori infection. However, it is known that there are numerous other causes of the condition. Aim Determination of the incidence of gastritis in patients with dyspepsia referred for diagnostic endoscopy of the upper part of the digestive tract, identification of the parts of the stomach most frequently affected by the inflammation, as well as the impact of an insufficient number of collected samples on the correct diagnosis. Material and methods Upper gastrointestinal endoscopy due to dyspepsia was performed in 110 patients. In the course of gastroscopy two biopsy specimens were collected for histopathological examination and towards H. pylori infection from the lesser and greater curvature in the antrum 3 cm from the pyloric sphincter, in the body – 4 cm proximally to the stomach angular incisure on the lesser curvature, and in the middle of the greater curvature, as well as in the subcardiac region on the side of the lesser and greater curvature. Results In patients with dyspepsia H. pylori-negative chronic gastritis is more common than gastritis with accompanying H. pylori infection. Collection of too small a number of biopsy specimens results in failure to detect inflammatory changes and/or H. pylori infection, which may be limited to one part of the stomach. Biopsy specimens of gastric mucosa should be collected in compliance with the assumptions of the Sydney System. Helicobacter pylori infection in people with dyspepsia is now being reported more rarely than in the past (36%). Conclusions In patients with dyspepsia chronic H. pylori-negative gastritis is more common than gastritis with an accompanying H. pylori infection. Helicobacter pylori infection is not always equivalent to the presence of chronic gastritis.
Collapse
|