Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 5.005
Filter
1.
Ann Acad Med Singap ; 53(7): 420-434, 2024 Jul 30.
Article in English | MEDLINE | ID: mdl-39132959

ABSTRACT

Introduction: Alcohol flushing syndrome (AFS) is experienced by up to 46% of East Asians. This study aimed to review the risk of cancers in AFS patients, elucidate an exposure-response relationship, and understand risk associated with alcohol intake and cancer. Method: An electronic database search of PubMed, Embase, Scopus and Cochrane Library was performed according to the Preferred Reporting Items for Systematic Reviews and Meta-analysis guidelines. Observational studies on AFS' effects and all cancers risk were included. Studies including patients with existing malignancy were excluded. Dichotomous variables were pooled using the Mantel-Haenszel method with a random effects model. Sensitivity and subgroup analyses were performed. PROSPERO (CRD42023392916) protocol was followed. Results: A total of 18 articles were included in the final analysis with a total of 387,521 participants. AFS was associated with an increased risk of all cancers (odds ratio [OR] 1.19, 95% confidence interval [CI] 1.06-1.34), esophageal squamous cell carcinoma (OR 1.47, 95% CI 1.05-2.05) and gastric adenocarci-noma (OR 1.40, 95% CI 1.14-1.72). Men with AFS exhibited an increased risk of all cancers (OR 1.34, 95% CI 1.13-1.59). However, this was not observed in women. All cancers risk was associated with AFS in those who consumed drink (i.e. consumed alcohol) more than 200 g of pure ethanol/week (OR 1.68, 95% CI 1.20-2.37) but not those who consumed less than 200 g of pure ethanol/week (OR 1.27, 95% CI 0.90-1.79) or non-drinkers (OR 0.99, 95% CI 0.67-1.47). Conclusion: AFS is associated with an increased risk of all cancers, particularly esophageal squamous cell carcinoma and gastric adenocarcinoma.


Subject(s)
Alcohol Drinking , Esophageal Neoplasms , Flushing , Humans , Flushing/epidemiology , Esophageal Neoplasms/epidemiology , Esophageal Neoplasms/etiology , Alcohol Drinking/adverse effects , Alcohol Drinking/epidemiology , Neoplasms/epidemiology , Neoplasms/etiology , Esophageal Squamous Cell Carcinoma/epidemiology , Esophageal Squamous Cell Carcinoma/etiology , Stomach Neoplasms/epidemiology , Stomach Neoplasms/etiology , Adenocarcinoma/epidemiology , Adenocarcinoma/etiology , Risk Factors
2.
Cancer Med ; 13(16): e70053, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39169774

ABSTRACT

BACKGROUND: Determining the effect of dietary factors on cancer is a crucial issue when accounting for the effect of other major risks, such as smoking and drinking. METHOD: A total of 15,563 adults from the Korean National Cancer Center Community Cohort were analyzed to determine and to compare the effect of dietary factors on stomach and colorectal cancer in overall and in the subgroup of non-smokers (or urinary cotinine concentrations <5 ng/mg) and non-drinkers with Cox proportional-hazard models. RESULTS: During the mean follow-up (13.7 years), 469 and 299 cases of stomach and colorectal cancer were identified, respectively. The preventive effect of vegetable, fish, and soybean/tofu intake on colorectal cancer was found in women after adjustment for smoking, drinking, BMI, and sociodemographic factors. In the subgroup analysis of non-smokers and non-drinkers, the effect on colorectal cancer was increased in women (≥1 time/week vs. almost never, vegetables: hazard ratio (HR) 0.30, 95% confidence interval (CI) 0.13-0.69; fish: HR 0.46, 95% CI 0.26-0.83), and the fresh fish intake effect on stomach cancer was newly identified in men (HR 0.36, 95% CI 0.15-0.86). These effects were more pronounced and additionally shown in other dietary factors such as soybean or tofu in women and vegetables and fish in men, when subjects with <5 ng/mg urinary cotinine concentrations applied. CONCLUSION: The protective effect of healthy eating on the risk of stomach and colorectal cancer were different by smoking and drinking status. Rigorous control of smoking and drinking effects is necessary when measuring the effect of dietary factors on cancer, properly.


Subject(s)
Alcohol Drinking , Colorectal Neoplasms , Diet, Healthy , Smoking , Stomach Neoplasms , Humans , Female , Male , Stomach Neoplasms/epidemiology , Stomach Neoplasms/prevention & control , Stomach Neoplasms/etiology , Middle Aged , Colorectal Neoplasms/epidemiology , Colorectal Neoplasms/prevention & control , Colorectal Neoplasms/etiology , Republic of Korea/epidemiology , Alcohol Drinking/epidemiology , Smoking/epidemiology , Smoking/adverse effects , Smoking/urine , Adult , Aged , Risk Factors , Cohort Studies , Proportional Hazards Models
3.
Sheng Li Xue Bao ; 76(4): 547-560, 2024 Aug 25.
Article in Chinese | MEDLINE | ID: mdl-39192788

ABSTRACT

Helicobacter pylori (Hp) is a Gram-negative bacterium that colonizes in the gastric mucosa. Hp induces the production of cancer-associated fibroblasts (CAF) in the stomach. The virulence factors of Hp and CAF trigger epithelial-mesenchymal transition (EMT), leading to local inflammation, damage to the gastric mucosa, and the occurrence of chronic gastritis. Here, we summarize the molecular mechanisms of CAF mediated gastric EMT after Hp infection, providing new insights into potential molecular targets and strategies for the future treatment of Hp infection associated gastric cancer.


Subject(s)
Epithelial-Mesenchymal Transition , Gastric Mucosa , Helicobacter Infections , Helicobacter pylori , Stomach Neoplasms , Humans , Helicobacter Infections/complications , Helicobacter pylori/physiology , Stomach Neoplasms/microbiology , Stomach Neoplasms/pathology , Stomach Neoplasms/etiology , Gastric Mucosa/microbiology , Gastric Mucosa/pathology , Cancer-Associated Fibroblasts/metabolism , Cancer-Associated Fibroblasts/physiology , Animals
4.
Cancer Treat Res ; 192: 1-17, 2024.
Article in English | MEDLINE | ID: mdl-39212913

ABSTRACT

Gastric cancer is a heterogeneous and prevalent disease. The traditional environmental exposures associated with elevated risk of gastric cancer are less prevalent in the USA today. Genetic risks and risks associated with inflammation remain. Most cases are sporadic, and familial clustering is observed in about 10% of the cases. Hereditary gastric cancer accounts for a very low percentage of cases. Here we review the genetic and environmental risk factors associated with the disease. In addition, we will review screening guidelines and current modalities that are available for screening in high-risk populations.


Subject(s)
Genetic Predisposition to Disease , Stomach Neoplasms , Humans , Stomach Neoplasms/genetics , Stomach Neoplasms/etiology , Stomach Neoplasms/epidemiology , Risk Factors , Environmental Exposure/adverse effects
5.
Curr Treat Options Oncol ; 25(9): 1137-1152, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39083164

ABSTRACT

OPINION STATEMENT: Gastric neuroendocrine neoplasms (G-NENs) are a heterogeneous group of tumors that broadly fall into two groups. The first group, driven by oversecretion of gastrin, are generally multifocal, small, and behave indolently with a low (but non-zero) risk of progression and metastatic spread. They are conventionally categorized into type 1, with endogenous gastric-based overproduction of gastrin, and type 2 G-NEN, with overproduction of gastrin from an extra-gastric gastrin-secreting tumor. The second group, termed type 3 G-NEN, occur spontaneously and are potentially more aggressive, having a clinical course analogous to other neuroendocrine tumors of the gastrointestinal tract. Type 1 G-NEN can be managed with endoscopic surveillance and resection of visible lesions with great success, reserving surgery for the rare high-risk lesion, whereas surgical resection of the causative gastrin-secreting tumor in type 2 G-NEN is usually curative. Type 3 G-NEN is usually managed with formal surgical resection but there is growing evidence that limited surgery or even endoscopic resection in appropriately selected patients with low risk is both safe and effective. A novel subtype of G-NEN, associated with long-term proton pump inhibitor usage, is increasing in incidence. The pathophysiology seems to parallel type 1 G-NEN. In the setting of metastatic disease, which can occur in any subtype but is most common by far in type 3 G-NEN, the lack of trial data unique to G-NEN results in extrapolation of strategies and agents for treatment of non-gastric neuroendocrine disease. The rapid pace of development in this area is likely to benefit the metastatic G-NEN patient as well. As treatment is predicate on type of G-NEN, establishing the etiology of the lesion is crucial but growing knowledge of G-NEN pathophysiology and close collaboration between pathologists, gastroenterologists, radiologists, surgeons, and oncologists have enabled a growing trend towards de-escalation and less-invasive treatment paradigms.


Subject(s)
Disease Management , Neuroendocrine Tumors , Stomach Neoplasms , Humans , Stomach Neoplasms/therapy , Stomach Neoplasms/diagnosis , Stomach Neoplasms/etiology , Stomach Neoplasms/pathology , Neuroendocrine Tumors/therapy , Neuroendocrine Tumors/diagnosis , Neuroendocrine Tumors/etiology , Neuroendocrine Tumors/pathology , Combined Modality Therapy/adverse effects , Treatment Outcome , Gastrins/metabolism
6.
Int J Mol Sci ; 25(14)2024 Jul 18.
Article in English | MEDLINE | ID: mdl-39063094

ABSTRACT

Chronic infection of Helicobacter pylori is considered the principal cause of gastric cancers, but evidence has accumulated regarding the impact of tobacco smoking and alcohol consumption on the development of gastric cancers. Several possible mechanisms, including the activation of nicotinic acetylcholine receptors, have been proposed for smoking-induced gastric carcinogenesis. On the other hand, local acetaldehyde exposure and ethanol-induced mucosal inflammation have been proposed as the mechanisms involved in the development of gastric cancers in heavy alcohol drinkers. In addition, genetic polymorphisms are also considered to play a pivotal role in smoking-related and alcohol-related gastric carcinogenesis. In this review, we will discuss the molecular mechanisms involved in the development of gastric cancers in relation to tobacco smoking and alcohol consumption.


Subject(s)
Alcohol Drinking , Stomach Neoplasms , Tobacco Smoking , Humans , Stomach Neoplasms/etiology , Stomach Neoplasms/genetics , Alcohol Drinking/adverse effects , Tobacco Smoking/adverse effects , Helicobacter Infections/complications , Helicobacter pylori/pathogenicity , Animals
7.
Front Endocrinol (Lausanne) ; 15: 1388608, 2024.
Article in English | MEDLINE | ID: mdl-38904039

ABSTRACT

Background: Gastric cancer (GC) is the third leading cause of cancer death worldwide, and hypothyroidism has been identified as a potential influencing factor. Despite known associations between hypothyroidism and various cancers, the causal link between hypothyroidism and GC and potential mediators of this relationship remains unclear. This study aimed to clarify these relationships using Mendelian randomization (MR). Methods: Utilizing genetic variant information from the FinnGen and MRC Integrative Epidemiology Unit open genome-wide association studies (GWAS) databases, we conducted univariable and multivariable MR analyses to explore the causal relationship between hypothyroidism and GC risk. The analysis was adjusted for confounders such as BMI, smoking status, and alcohol intake, and included mediator MR analysis to examine the role of high cholesterol. Results: We identified a significant inverse association between hypothyroidism and GC risk (OR = 0.93, 95% CI= 0.89-0.98, P = 0.003), with no evidence of reverse causation or pleiotropy. Adjustments for Helicobacter pylori infection weakened this association. Mediator analysis highlighted high cholesterol levels, chronic hepatitis B infection, and diabetes/endocrine disease status as significant mediators of the protective effect of hypothyroidism on GC risk. Conclusion: Our findings suggest that hypothyroidism may confer a protective effect against GC, mediated in part by high cholesterol and other factors. These results underscore the importance of thyroid function and metabolic health in GC risk, offering new insights for preventive strategies and highlighting the need for further research into these complex associations.


Subject(s)
Genome-Wide Association Study , Hypothyroidism , Mediation Analysis , Mendelian Randomization Analysis , Stomach Neoplasms , Humans , Stomach Neoplasms/genetics , Stomach Neoplasms/epidemiology , Stomach Neoplasms/etiology , Hypothyroidism/genetics , Hypothyroidism/epidemiology , Hypothyroidism/complications , Risk Factors , Female , Male , Polymorphism, Single Nucleotide
8.
Neoplasia ; 55: 101012, 2024 09.
Article in English | MEDLINE | ID: mdl-38875930

ABSTRACT

Increased mutational burden and EBV load have been revealed from normal tissues to Epstein-Barr virus (EBV)-associated gastric carcinomas (EBVaGCs). BPLF1, encoded by EBV, is a lytic cycle protein with deubiquitinating activity has been found to participate in disrupting repair of DNA damage. We first confirmed that BPLF1 gene in gastric cancer (GC) significantly increased the DNA double strand breaks (DSBs). Ubiquitination mass spectrometry identified histones as BPLF1 interactors and potential substrates, and co-immunoprecipitation and in vitro experiments verified that BPLF1 regulates H2Bub by targeting Rad6. Over-expressing Rad6 restored H2Bub but partially reduced γ-H2AX, suggesting that other downstream DNA repair processes were affected. mRNA expression of BRCA2 were significantly down-regulated by next-generation sequencing after over-expression of BPLF1, and over-expression of p65 facilitated the repair of DSBs. We demonstrated BPLF1 may lead to the accumulation of DSBs by two pathways, reducing H2B ubiquitination (H2Bub) and blocking homologous recombination which may provide new ideas for the treatment of gastric cancer.


Subject(s)
DNA Breaks, Double-Stranded , Epstein-Barr Virus Infections , Genomic Instability , Herpesvirus 4, Human , Histones , Stomach Neoplasms , Ubiquitination , Humans , Herpesvirus 4, Human/genetics , Stomach Neoplasms/virology , Stomach Neoplasms/genetics , Stomach Neoplasms/metabolism , Stomach Neoplasms/pathology , Stomach Neoplasms/etiology , Epstein-Barr Virus Infections/virology , Epstein-Barr Virus Infections/metabolism , Epstein-Barr Virus Infections/complications , Epstein-Barr Virus Infections/genetics , Histones/metabolism , Cell Line, Tumor , DNA Repair , Deubiquitinating Enzymes/metabolism , Deubiquitinating Enzymes/genetics , Gene Expression Regulation, Neoplastic , BRCA2 Protein/genetics , BRCA2 Protein/metabolism , Carcinogenesis/genetics , Viral Regulatory and Accessory Proteins
9.
Am J Gastroenterol ; 119(6): 1056-1065, 2024 06 01.
Article in English | MEDLINE | ID: mdl-38832708

ABSTRACT

INTRODUCTION: Ultra-processed food (UPF) intake has been associated with a higher risk of obesity, hypertension, type 2 diabetes, and cardiovascular diseases. The initial data on the relationship between UPF consumption and cancer risk were derived from retrospective observational studies with conflicting results. This systematic review and meta-analysis of prospective cohort studies aimed to investigate the association between UPF consumption and gastrointestinal cancer risk. METHODS: PubMed, Embase, and Cochrane databases were searched for prospective cohort studies that compared the highest vs the lowest level of UPF consumption according to NOVA food classification and reported the risk of gastrointestinal cancers by subsite. The association with cancer was quantified as hazard ratios (HR) using a random-effects model. RESULTS: Five prospective cohort studies were included in this review comprising 1,128,243 participants (241,201 participants in the highest and 223,366 in the lowest levels of UPF consumption). The mean follow-up ranged from 5.4 to 28 years. The highest UPF consumption was significantly associated with an increased risk of colorectal cancer (HR 1.11; 95% confidence interval [CI] 1.03-1.21; P = 0.01; I2 = 31%), colon cancer (HR 1.12; 95% CI 1.02-1.23; P = 0.02; I2 = 0%), and non-cardia gastric cancer (HR 1.43; 95% CI 1.02-2.00; P = 0.04; I2 = 0%) compared with the lowest UPF intake. However, no association was found between high UPF consumption and hepatocellular, esophageal, pancreatic, gastric cardia, and rectal cancer. DISCUSSION: The highest level of UPF consumption was significantly associated with colorectal and non-cardia gastric cancer.


Subject(s)
Fast Foods , Gastrointestinal Neoplasms , Humans , Gastrointestinal Neoplasms/epidemiology , Gastrointestinal Neoplasms/etiology , Fast Foods/adverse effects , Risk Factors , Colorectal Neoplasms/epidemiology , Colorectal Neoplasms/etiology , Stomach Neoplasms/epidemiology , Stomach Neoplasms/etiology , Food, Processed
11.
Int J Mol Sci ; 25(10)2024 May 09.
Article in English | MEDLINE | ID: mdl-38791187

ABSTRACT

Non-coding RNAs (ncRNAs) represent a broad family of molecules that regulate gene expression, including microRNAs, long non-coding RNAs and circular RNAs, amongst others. Dysregulated expression of ncRNAs alters gene expression, which is implicated in the pathogenesis of several malignancies and inflammatory diseases. Gastric cancer is the fifth most frequently diagnosed cancer and the fourth most common cause of cancer-related death. Studies have found that altered expression of ncRNAs may contribute to tumourigenesis through regulating proliferation, apoptosis, drug resistance and metastasis. This review describes the potential use of ncRNAs as diagnostic and prognostic biomarkers. Moreover, we discuss the involvement of ncRNAs in the pathogenesis of gastric cancer, including their interactions with the members of major signalling pathways.


Subject(s)
Biomarkers, Tumor , Gene Expression Regulation, Neoplastic , RNA, Untranslated , Stomach Neoplasms , Humans , Stomach Neoplasms/genetics , Stomach Neoplasms/metabolism , Stomach Neoplasms/pathology , Stomach Neoplasms/etiology , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , RNA, Untranslated/genetics , RNA, Untranslated/metabolism , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Prognosis , Signal Transduction , Animals , MicroRNAs/genetics , MicroRNAs/metabolism , RNA, Circular/genetics , RNA, Circular/metabolism
12.
World J Gastroenterol ; 30(17): 2308-2310, 2024 May 07.
Article in English | MEDLINE | ID: mdl-38813049

ABSTRACT

Gastric cystica profunda (GCP) is an uncommon but underestimated gastric lesion. Its precancerous potential determines its significance. In addition to previous mucosa injury due to operations, biopsy or polypectomy, chronic active and atrophic gastritis may also lead to the development of GCPs. By carefully examining the stomach and taking biopsy samples from the susceptible regions, the stage of atrophy can be determined. Chronic atrophic gastritis is a risk factor for cancer evolvement and it can also contribute to GCPs formation. GCPs frequently occur close to early gastric cancers (EGCs) or EGC can arise from the cystic glands. Endoscopic resection is an effective and minimally invasive treatment in GCP.


Subject(s)
Gastric Mucosa , Gastritis, Atrophic , Precancerous Conditions , Stomach Neoplasms , Humans , Biopsy , Chronic Disease , Cysts/surgery , Cysts/pathology , Cysts/etiology , Gastric Mucosa/pathology , Gastric Mucosa/surgery , Gastric Mucosa/diagnostic imaging , Gastritis, Atrophic/pathology , Gastritis, Atrophic/complications , Gastritis, Atrophic/surgery , Gastroscopy , Precancerous Conditions/pathology , Precancerous Conditions/surgery , Precancerous Conditions/etiology , Risk Factors , Stomach Diseases/etiology , Stomach Diseases/surgery , Stomach Diseases/pathology , Stomach Neoplasms/pathology , Stomach Neoplasms/surgery , Stomach Neoplasms/etiology
13.
Int J Cancer ; 155(7): 1203-1211, 2024 Oct 01.
Article in English | MEDLINE | ID: mdl-38712628

ABSTRACT

The relationship between Helicobacter pylori (H. pylori) infection and upper gastrointestinal (UGI) cancers is complex. This multicenter, population-based cohort study conducted in seven areas in China aimed to assess the correlation between current H. pylori infection and the severity of UGI lesions, as well as its association with the risk of gastric cancer (GC) and esophageal cancer (EC). From 2015 to 2017, 27,085 participants (aged 40-69) completed a standardized questionnaire, and underwent a 13C-urea breath test. Then a subset underwent UGI endoscopy to assess the UGI lesion detection rates. All individuals were followed up until December 2021 to calculate the hazard ratios (HRs) for UGI cancers. H. pylori infection prevalence was 45.9%, and among endoscopy participants, 22.2% had gastric lesions, 19.2% had esophageal lesions. Higher detection rates of gastric lesions were noted in the H. pylori-positive population across all lesion severity levels. Over a median follow-up of 6.3 years, 104 EC and 179 GC cases were observed, including 103 non-cardia gastric cancer (NCGC) cases and 76 cardia gastric cancer (CGC) cases. H. pylori-infected individuals exhibited a 1.78-fold increased risk of GC (HR 1.78, 95% confidence interval [CI] 1.32-2.40) but no significant increase in EC risk (HR 1.07, 95% CI 0.73-1.57). Notably, there was a higher risk for both NCGC and CGC in H. pylori-infected individuals. This population-based cohort study provides valuable evidence supporting the association between current H. pylori infection and the risk of both NCGC and CGC. These findings contribute to the empirical basis for risk stratification and recommendations for UGI cancer screening.


Subject(s)
Esophageal Neoplasms , Helicobacter Infections , Helicobacter pylori , Stomach Neoplasms , Humans , Helicobacter Infections/complications , Helicobacter Infections/epidemiology , Middle Aged , Male , Female , Helicobacter pylori/isolation & purification , Adult , Stomach Neoplasms/microbiology , Stomach Neoplasms/epidemiology , Stomach Neoplasms/etiology , Stomach Neoplasms/pathology , Aged , Esophageal Neoplasms/epidemiology , Esophageal Neoplasms/microbiology , Esophageal Neoplasms/etiology , China/epidemiology , Cohort Studies , Risk Factors , Prevalence , Gastrointestinal Neoplasms/microbiology , Gastrointestinal Neoplasms/epidemiology , Gastrointestinal Neoplasms/etiology , Upper Gastrointestinal Tract/pathology , Upper Gastrointestinal Tract/microbiology
14.
Chirurgia (Bucur) ; 119(eCollection): 1-10, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38657111

ABSTRACT

Helicobacter pylori (H. pylori), classified as a Group 1 carcinogen by the International Agency for Research on Cancer (IARC), is linked to gastric cancer. The progression from atrophy to metaplasia, dysplasia, and carcinoma constitutes the pathway for intestinal-type gastric carcinoma development. H. pylori infection significantly increases gastric cancer risk, particularly in individuals with atrophic gastritis. Virulence factors like CagA and VacA disrupt host signaling pathways, contributing to chronic inflammation and carcinogenesis. Pro-inflammatory cytokines and dysregulated tumor suppressor genes further fuel this process. Eradicating H. pylori reduces gastric cancer incidence, especially in patients with atrophic gastritis and/or intestinal metaplasia. However, it may not prevent cancer in those with advanced pre-neoplastic lesions. Early detection and management of H. pylori infection are crucial in mitigating gastric cancer risk, offering significant benefits.


Subject(s)
Antigens, Bacterial , Bacterial Proteins , Helicobacter Infections , Helicobacter pylori , Stomach Neoplasms , Humans , Stomach Neoplasms/microbiology , Stomach Neoplasms/etiology , Helicobacter Infections/complications , Risk Factors , Incidence , Gastritis, Atrophic/microbiology , Treatment Outcome , Virulence Factors
15.
Radiol Oncol ; 58(2): 186-195, 2024 06 01.
Article in English | MEDLINE | ID: mdl-38643513

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.


Subject(s)
Helicobacter Infections , Helicobacter pylori , Metaplasia , Precancerous Conditions , Stomach Neoplasms , Humans , Stomach Neoplasms/pathology , Stomach Neoplasms/microbiology , Stomach Neoplasms/etiology , Metaplasia/pathology , Helicobacter Infections/complications , Helicobacter Infections/pathology , Precancerous Conditions/pathology , Precancerous Conditions/microbiology , Carcinogenesis/pathology , Gastritis, Atrophic/pathology , Gastritis, Atrophic/microbiology , Gastric Mucosa/pathology , Gastric Mucosa/microbiology , Intestines/pathology , Intestines/microbiology
16.
J Gastroenterol Hepatol ; 39(8): 1580-1585, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38641971

ABSTRACT

BACKGROUND AND AIM: Since the first report of gastric adenocarcinoma of the fundic-gland type in 2010, the clinicopathological characteristics of gastric neoplasm of the fundic-gland type (GNFG) have become clearer; however, their risk factors remain unclear. This exploratory study aimed to identify the risk factors for GNFG. METHODS: We conducted a single-center, retrospective, matched case-control study using medical information recorded at our health management center from January 2014 to July 2023. During this period, 39 240 people underwent upper gastrointestinal endoscopy. GNFG were extracted as cases and matched to controls, according to age and sex, in a 1:8 ratio, excluding those with a history of gastrointestinal surgery and those with a history or comorbidity of cancer. Univariate analysis was used to compare patient background and endoscopic findings. Multivariable analysis was performed, adjusting for factors with P values < 0.1 and antacid use. RESULTS: A total of 20 GNFG cases and 160 matched healthy controls were included. In the univariate analysis, only reflux esophagitis was significantly more common in GNFG (40.0% vs 18.1%; P = 0.036). Factors antacids and duodenitis had P values < 0.1. Logistic regression analysis was performed, adjusting for antacids, reflux esophagitis, and duodenitis. Antacids and reflux esophagitis were the independent risk factors for GNFG (odds ratio = 3.68 [95% confidence interval: 1.04-11.91] and 3.25 [95% confidence interval: 1.11-9.35]). CONCLUSIONS: Although the sample of patients with GNFG was small, antacids and reflux esophagitis were identified as a risk factor. The pathogenesis of antacids and reflux esophagitis may be involved in the development of GNFG.


Subject(s)
Antacids , Esophagitis, Peptic , Stomach Neoplasms , Humans , Antacids/therapeutic use , Risk Factors , Retrospective Studies , Case-Control Studies , Male , Stomach Neoplasms/epidemiology , Stomach Neoplasms/etiology , Female , Esophagitis, Peptic/epidemiology , Esophagitis, Peptic/etiology , Middle Aged , Aged , Adenocarcinoma/etiology , Adenocarcinoma/epidemiology , Gastric Fundus/pathology , Adult
17.
Sci Rep ; 14(1): 8076, 2024 04 06.
Article in English | MEDLINE | ID: mdl-38580718

ABSTRACT

Postoperative hepatobiliary enzyme abnormalities often present as postoperative liver dysfunction in patients with gastric cancer (GC). This study aimed to identify the risk factors for postoperative liver dysfunction and their clinical impact after GC surgery. We retrospectively analyzed the data of 124 patients with GC who underwent laparoscopic or robotic surgery at Kyoto Prefectural University of Medicine between 2017 and 2019. Twenty (16.1%) patients with GC developed postoperative liver dysfunction (Common Terminology Criteria for Adverse Events (CTCAE) version 5.0 ≥ Grade 3). Univariate analyses identified robotic surgery as a risk factor for postoperative liver dysfunction (P = 0.005). There was no correlation between the postoperative liver dysfunction status and postoperative complications or postoperative hospital stays. Patients with postoperative liver dysfunction did not have significantly worse overall survival (P = 0.296) or recurrence-free survival (P = 0.565) than those without postoperative liver dysfunction. Robotic surgery is a risk factor for postoperative liver dysfunction; however, postoperative liver dysfunction does not affect short or long-term outcomes.


Subject(s)
Laparoscopy , Stomach Neoplasms , Humans , Stomach Neoplasms/etiology , Retrospective Studies , Gastrectomy/adverse effects , Clinical Relevance , Treatment Outcome , Laparoscopy/adverse effects , Postoperative Complications/etiology , Postoperative Complications/surgery , Risk Factors
18.
Maturitas ; 184: 107960, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38460415

ABSTRACT

OBJECTIVE: Gastric cancer (GC) is more common in men than women, but also more common among postmenopausal than premenopausal women. The protective effect of reproductive hormones against GC remains unclear. Therefore, we evaluated the association between menopausal hormone therapy (MHT) and the risk of GC in women. METHODS: We investigated the national cohort data of women aged over 40 years who underwent health checkups by the Korean National Health Insurance Service in 2009. After excluding individuals with missing data and those previously diagnosed with cancer, 1,354,621 postmenopausal women were included and divided into groups according to their MHT history. We followed the study population until 2018 and analyzed the hazard ratios (HR) with 95 % confidence intervals (CIs) for the incidence rate of GC in a multivariate adjusted model. RESULTS: The number of women with and without a history of MHT was 214,723 (15.9 %) and 1,139,898 (84.1 %), respectively. During the mean 8.32 ± 0.8 years of follow-up, a total of 12,496 GC cases developed in the study population (10,962 MHT non-users; 1534 MHT users). In the adjusted model, MHT was associated with a 12 % decrease in the development of GC relative to non-use of MHT (HR 0.88; 95 % CI 0.83-0.93). Exposure to MHT for >2 years was linked to a reduction in GC risk, particularly when initiated before the age of 50, giving a 45 % risk reduction. CONCLUSIONS: According to our large-scale prospective national cohort study, exogenous MHT is associated with a decreased risk of GC in postmenopausal women.


Subject(s)
Stomach Neoplasms , Humans , Stomach Neoplasms/epidemiology , Stomach Neoplasms/chemically induced , Stomach Neoplasms/etiology , Female , Republic of Korea/epidemiology , Middle Aged , Cohort Studies , Adult , Risk Factors , Incidence , Aged , Menopause , Postmenopause , Proportional Hazards Models , Estrogen Replacement Therapy/adverse effects , Estrogen Replacement Therapy/statistics & numerical data , Hormone Replacement Therapy/adverse effects
19.
P R Health Sci J ; 43(1): 9-17, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38512756

ABSTRACT

Of the chronic bacterial infections that affect humans, Helicobacter pylori (H. pylori) infection is one of the most common. It inhabits the stomachs of half of the adult human population. In Puerto Rico, a US territory, it has an overall prevalence of 33%, similar to the prevalence reported in the population of the US as a whole. Helicobacter pylori infection is responsible for mucosal inflammation that may lead to chronic gastritis, most peptic ulcers, gastric adenocarcinoma, and mucosa-associated lymphoid tissue lymphoma. The International Agency for Research on Cancer identified H. pylori as a definite carcinogen in 1994, the only bacterium to be given such a classification. Its oncogenic effect has been postulated to be caused by different mechanisms, including bacterial characteristics and host factors. Epidemiologic studies have shown that gastric cancer risk differs among regions. One of the top 10 causes of cancer death in Puerto Rico is gastric cancer. Although the eradication of H. pylori has well-known benefits, there are some concerns when considering mass screening and treatment of infected patients. These include the fact that such eradication could provoke an increase in antibiotic resistance rates, the disturbance of the gut microbiota, an increase in body weight, and the aggravation of existing gastroesophageal reflux symptoms. Gastric cancer is a major health concern, and we should understand the role of H. pylori eradication in its prevention. This article is geared to summarize current knowledge and controversies.


Subject(s)
Gastritis, Atrophic , Helicobacter Infections , Helicobacter pylori , Stomach Neoplasms , Adult , Humans , Stomach Neoplasms/epidemiology , Stomach Neoplasms/etiology , Helicobacter Infections/complications , Helicobacter Infections/epidemiology , Gastritis, Atrophic/complications , Gastritis, Atrophic/microbiology , Gastritis, Atrophic/pathology , Puerto Rico
SELECTION OF CITATIONS
SEARCH DETAIL