Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 4.995
Filter
2.
Am J Gastroenterol ; 119(6): 1056-1065, 2024 Jun 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
3.
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
4.
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
5.
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
6.
Radiol Oncol ; 58(2): 186-195, 2024 Jun 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
7.
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
8.
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
9.
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
10.
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
11.
Nurs Open ; 11(2): e2104, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38369669

ABSTRACT

AIM: This study aims to develop and validate a clinical nutrition risk screening tool to predict nutrition risk in home for the patients with gastric cancer after surgery at home so that high-risk patients can be targeted for preventive nutrition care. DESIGN: The development of self-screening tool for nutrition risk in patients with gastric cancer after gastrectomy (SNRSGC) through literature review, expert panel ratings and cognitive interview; the validation of SNRSGC is evaluated through prospective research on participants. METHODS: This research is divided into four parts: Step 1, Identification of a potential referred nutritional risk screening; Step 2, Item generation and scoring are selected through literature review methods to screen sensitive indicators which can reflect the nutritional characteristics of patients after gastric cancer surgery, establish the frame and update according to the latest guidelines; Step 3, Item reduction is determined by the rating of SNRSGC items by an expert panel and cognitive interview; Step 4, During the validation stage, we conducted research design based on the Consensus-based Standards for the selection of health Measurement Instruments checklist to evaluate the validity, reliability, interpretability and acceptability of SNRSGC. RESULTS: SNRSGC is the first screening tool specifically to predict nutrition risk for stay-at-home postoperative patients with gastric cancer, which can help patients at home detect nutritional risks at home in time and guide patients to seek medical treatment as soon as possible to improve their nutritional status.


Subject(s)
Nutritional Status , Stomach Neoplasms , Humans , Stomach Neoplasms/diagnosis , Stomach Neoplasms/surgery , Stomach Neoplasms/etiology , Prospective Studies , Reproducibility of Results , Early Detection of Cancer , Gastrectomy/adverse effects
12.
Front Immunol ; 15: 1321946, 2024.
Article in English | MEDLINE | ID: mdl-38361930

ABSTRACT

HER2 overexpression/amplification is a prevalent driver in various types of cancer, including gastric cancer (GC). Limited options are available for patients with HER2-positive metastatic gastric cancer, particularly those who do not respond to the standard therapy of HER2 antibody trastuzumab combined with chemotherapy. Previous research suggests that combining a PD-1 inhibitor with radiotherapy and granulocyte macrophage-colony stimulating factor (PRaG regimen) may enhance the antitumor effects in patients with chemotherapy-resistant metastatic solid tumors. In this case study, we presented a potential treatment strategy of a patient having HER2-positive and PD-L1-negative gastric adenocarcinoma. The patient showed rapid tumor progression even after surgery and multiple trastuzumab plus chemotherapy treatments. To address this, we employed a novel anti-HER2 antibody called RC48 in combination with PRaG regimen therapy (PRaG3.0). The patient demonstrated a positive response after two treatment cycles and achieved a progression-free survival time of 6.5 months. This case highlights the potential of four-combination therapies for treating refractory, multiorgan, HER2-positive, PD-L1-negative metastatic gastric cancer. Additionally, varying radiation doses in targeting dual foci is critical to enhance tumor immunotherapy.


Subject(s)
Stomach Neoplasms , Humans , Stomach Neoplasms/drug therapy , Stomach Neoplasms/etiology , Immune Checkpoint Inhibitors/therapeutic use , Granulocyte-Macrophage Colony-Stimulating Factor/therapeutic use , B7-H1 Antigen , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Trastuzumab/therapeutic use , Granulocytes , Macrophages
13.
Invest New Drugs ; 42(2): 171-178, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38347177

ABSTRACT

BACKGROUND: Immune checkpoint inhibitors have been approved for first- and third-line treatment of advanced gastric cancer. However, pembrolizumab alone in the second line did not improve overall survival compared to chemotherapy in the KEYNOTE-061 study. In this study, we aimed to explore the efficacy and safety of a three-drug regimen of PD-1 inhibitor combined with albumin paclitaxel and apatinib (a VEGFR inhibitor) for the second-line treatment of patients with metastatic gastric cancer (mGC). METHODS: This was a single-center, single-arm, phase II clinical study. Patients with mGC with stable microsatellite and negative HER-2 expression who failed first-line chemotherapy were enrolled. The enrolled patients were treated with PD-1 inhibitor (selected according to patients' requirements) in combination with albumin paclitaxel (125 mg/m2, intravenously, days 1 and 8, or 250 mg/m2, intravenously, day 1) and apatinib (250 or 500 mg, orally, days 1-21) every 3 weeks. The primary endpoint was progression-free survival (PFS), and the secondary endpoints were overall survival (OS), objective response rate (ORR), disease control rate (DCR), duration of response, and adverse events (AEs). RESULTS: From July 11, 2019, to October 13, 2022, a total of 43 patients were enrolled, of whom 10 were PD-L1 negative, 11 were PD-L1 positive, and 22 had unknown PD-L1 expression. As of the data cutoff on April 1st, 2023, nine patients had partial response, 29 had stable disease, and five experienced progressive disease, with the ORR of 20.9% and DCR of 88.3%. The median PFS was 6.2 months (95% CI, 3.9-9.3), and the median OS was 10.1 months (95% CI, 7.5-14.1). All patients suffered from alopecia and neurotoxicity. The other main AEs of grade 1 or 2 were bone marrow suppression (N = 21, 48.8%), hand-foot reaction (N = 19, 44.2%), hypertension (N = 18, 41.9%), hypothyroidism (N = 11, 25.6%), gastrointestinal bleeding (N = 3, 7.0%), and liver function damage (N = 5, 11.6%). Two patients reported grade 3-4 immune-related liver damage. CONCLUSION: Second-line PD-1 inhibitor combined with albumin paclitaxel and apatinib showed certain efficacy and safety in patients with mGC. TRIAL REGISTRATION: Clinical trials, NCT04182724. Registered 27 November 2019; retrospectively registered, https://clinicaltrials.gov/study/NCT04182724.


Subject(s)
Paclitaxel , Pyridines , Stomach Neoplasms , Humans , Paclitaxel/adverse effects , Immune Checkpoint Inhibitors/therapeutic use , Stomach Neoplasms/drug therapy , Stomach Neoplasms/etiology , B7-H1 Antigen , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Albumins/therapeutic use
14.
J Gastroenterol Hepatol ; 39(6): 1027-1032, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38413187

ABSTRACT

Big data is characterized by three attributes: volume, variety,, and velocity. In healthcare setting, big data refers to vast dataset that is electronically stored and managed in an automated manner and has the potential to enhance human health and healthcare system. In this review, gastric cancer (GC) and postcolonoscopy colorectal cancer (PCCRC) will be used to illustrate application of big data approach in the field of gastrointestinal cancer research. Helicobacter pylori (HP) eradication only reduces GC risk by 46% due to preexisting precancerous lesions. Apart from endoscopy surveillance, identifying medications that modify GC risk is another strategy. Population-based cohort studies showed that long-term use of proton pump inhibitors (PPIs) associated with higher GC risk after HP eradication, while aspirin and statins associated with lower risk. While diabetes mellitus conferred 73% higher GC risk, metformin use associated with 51% lower risk, effect of which was independent of glycemic control. Nonetheless, nonsteroidal anti-inflammatory drugs (NA-NSAIDs) are not associated with lower GC risk. CRC can still occur after initial colonoscopy in which no cancer was detected (i.e. PCCRC). Between 2005 and 2013, the rate of interval-type PCCRC-3y (defined as CRC diagnosed between 6 and 36 months of index colonoscopy which was negative for CRC) was 7.9% in Hong Kong, with >80% being distal cancers and higher cancer-specific mortality compared with detected CRC. Certain clinical and endoscopy-related factors were associated with PCCRC-3 risk. Medications shown to have chemopreventive effects on PCCRC include statins, NA-NSAIDs, and angiotensin-converting enzyme inhibitors/angiotensin receptor blockers.


Subject(s)
Big Data , Colorectal Neoplasms , Stomach Neoplasms , Humans , Colorectal Neoplasms/epidemiology , Stomach Neoplasms/microbiology , Stomach Neoplasms/etiology , Helicobacter Infections/drug therapy , Helicobacter Infections/complications , Colonoscopy , Proton Pump Inhibitors/therapeutic use , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Hydroxymethylglutaryl-CoA Reductase Inhibitors/therapeutic use , Helicobacter pylori , Aspirin/therapeutic use , Metformin/therapeutic use , Risk , Biomedical Research , Risk Factors
15.
World J Gastroenterol ; 30(4): 286-289, 2024 Jan 28.
Article in English | MEDLINE | ID: mdl-38313231

ABSTRACT

Gastric cancer, a prevalent malignancy worldwide, ranks sixth in terms of frequency and third in fatality, causing over a million new cases and 769000 annual deaths. Predominant in Eastern Europe and Eastern Asia, risk factors include family medical history, dietary habits, tobacco use, Helicobacter pylori, and Epstein-Barr virus infections. Unfortunately, gastric cancer is often diagnosed at an advanced stage, leading to a grim prognosis, with a 5-year overall survival rate below 5%. Surgical intervention, particularly with D2 Lymphadenectomy, is the mainstay for early-stage cases but offers limited success. For advanced cases, the National Comprehensive Cancer Network recommends chemotherapy, radiation, and targeted therapy. Emerging immunotherapy presents promise, especially for unresectable or metastatic cases, with strategies like immune checkpoint inhi-bitors, tumor vaccines, adoptive immunotherapy, and nonspecific immunomodulators. In this Editorial, with regards to the article "Advances and key focus areas in gastric cancer immunotherapy: A comprehensive scientometric and clinical trial review", we address the advances in the field of immunotherapy in gastric cancer and its future prospects.


Subject(s)
Epstein-Barr Virus Infections , Stomach Neoplasms , Humans , Stomach Neoplasms/therapy , Stomach Neoplasms/etiology , Herpesvirus 4, Human , Immunotherapy/adverse effects , Immunotherapy, Adoptive
16.
Gut Liver ; 18(2): 257-264, 2024 Mar 15.
Article in English | MEDLINE | ID: mdl-38384180

ABSTRACT

Background/Aims: : Tegoprazan is a novel potassium-competitive acid blocker that has beneficial effects on acid-related disorders such as gastroesophageal reflux and peptic ulcer diseases. This study aimed to validate the effect of tegoprazan on endoscopic submucosal dissection (ESD)-induced artificial ulcers. Methods: : Patients from 16 centers in Korea who underwent ESD for gastric neoplasia were enrolled. After ESD, pantoprazole was administered intravenously for 48 hours. The patients were randomly allocated to either the tegoprazan or esomeprazole group. Tegoprazan 50 mg or esomeprazole 40 mg were administered for 4 weeks, after which gastroscopic evaluation was performed. If the artificial ulcer had not healed, the same dose of tegoprazan or esomeprazole was administered for an additional 4 weeks, and a gastroscopic evaluation was performed. Results: : One hundred sixty patients were enrolled in this study. The healing rates of artificial ulcers at 4 weeks were 30.3% (23/76) and 22.1% (15/68) in the tegoprazan and esomeprazole groups, respectively (p=0.006). At 8 weeks after ESD, the cumulative ulcer healing rates were 73.7% (56/76) and 77.9% (53/68) in the tegoprazan and esomeprazole groups, respectively (p=0.210). Delayed bleeding occurred in two patients in the tegoprazan group (2.6%) and in one patient in the esomeprazole group (1.5%). Other adverse events were negligible in both groups. Conclusions: : Tegoprazan showed similar effects on post-ESD artificial ulcer healing in comparison with esomeprazole.


Subject(s)
Benzene Derivatives , Endoscopic Mucosal Resection , Imidazoles , Stomach Neoplasms , Stomach Ulcer , Humans , Esomeprazole/therapeutic use , Ulcer/drug therapy , Ulcer/etiology , Proton Pump Inhibitors/therapeutic use , Stomach Ulcer/drug therapy , Stomach Ulcer/surgery , Stomach Ulcer/etiology , Stomach Neoplasms/etiology , Endoscopic Mucosal Resection/adverse effects
17.
Int J Mol Sci ; 25(3)2024 Feb 03.
Article in English | MEDLINE | ID: mdl-38339127

ABSTRACT

Gastric cancer (GC) is the fourth leading cause of death worldwide, with more than 1 million cases diagnosed every year. Helicobacter pylori represents the main risk factor, being responsible for 78% of the cases. Increased amounts of salt, pickled food, red meat, alcohol, smoked food, and refined sugars negatively affect the stomach wall, contributing to GC development. Several gene mutations, including PIK3CA, TP53, ARID1A, CDH1, Ras, Raf, and ERBB3 are encountered in GC pathogenesis, leading to phosphatidylinositol 3-kinase (PI3K) protein kinase B (AKT)/mammalian target of rapamycin (mTOR)-PI3K/AKT/mTOR-and mitogen-activated protein kinase (MAPK) signaling pathway activation and promoting tumoral activity. Helicobacter pylori, growth factors, cytokines, hormones, and oxidative stress also activate both pathways, enhancing GC development. In clinical trials, promising results have come from monoclonal antibodies such as trastuzumab and ramucirumab. Dual inhibitors targeting the PI3K/AKT/mTOR and MAPK signaling pathways were used in vitro studies, also with promising results. The main aim of this review is to present GC incidence and risk factors and the dysregulations of the two protein kinase complexes together with their specific inhibitors.


Subject(s)
Proto-Oncogene Proteins c-akt , Stomach Neoplasms , Humans , Proto-Oncogene Proteins c-akt/metabolism , Phosphatidylinositol 3-Kinase/metabolism , Sirolimus , Phosphatidylinositol 3-Kinases/metabolism , Stomach Neoplasms/etiology , Stomach Neoplasms/genetics , TOR Serine-Threonine Kinases/metabolism , MAP Kinase Signaling System
18.
Int J Cancer ; 154(11): 1900-1910, 2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38339851

ABSTRACT

Air pollution has been shown to significantly impact human health including cancer. Gastric and upper aerodigestive tract (UADT) cancers are common and increased risk has been associated with smoking and occupational exposures. However, the association with air pollution remains unclear. We pooled European subcohorts (N = 287,576 participants for gastric and N = 297,406 for UADT analyses) and investigated the association between residential exposure to fine particles (PM2.5), nitrogen dioxide (NO2), black carbon (BC) and ozone in the warm season (O3w) with gastric and UADT cancer. We applied Cox proportional hazards models adjusting for potential confounders at the individual and area-level. During 5,305,133 and 5,434,843 person-years, 872 gastric and 1139 UADT incident cancer cases were observed, respectively. For gastric cancer, we found no association with PM2.5, NO2 and BC while for UADT the hazard ratios (95% confidence interval) were 1.15 (95% CI: 1.00-1.33) per 5 µg/m3 increase in PM2.5, 1.19 (1.08-1.30) per 10 µg/m3 increase in NO2, 1.14 (1.04-1.26) per 0.5 × 10-5 m-1 increase in BC and 0.81 (0.72-0.92) per 10 µg/m3 increase in O3w. We found no association between long-term ambient air pollution exposure and incidence of gastric cancer, while for long-term exposure to PM2.5, NO2 and BC increased incidence of UADT cancer was observed.


Subject(s)
Air Pollutants , Air Pollution , Stomach Neoplasms , Humans , Particulate Matter/adverse effects , Particulate Matter/analysis , Nitrogen Dioxide/adverse effects , Stomach Neoplasms/epidemiology , Stomach Neoplasms/etiology , Incidence , Environmental Exposure/adverse effects , Air Pollution/adverse effects , Air Pollution/analysis , Air Pollutants/adverse effects , Air Pollutants/analysis
20.
Cleve Clin J Med ; 91(1): 33-39, 2024 Jan 02.
Article in English | MEDLINE | ID: mdl-38167394

ABSTRACT

Gastric intestinal metaplasia (GIM), a common histologic finding, is associated with increased risk of gastric cancer, and GIM associated with Helicobacter pylori infection is classified as an environmental metaplastic atrophic gastritis. Patients may be asymptomatic or present with various dyspeptic symptoms. Autoimmune metaplastic atrophic gastritis is a less common but important cause of chronic gastritis. The Correa cascade describes the evolution of precancerous mucosal changes that lead to development of GIM, with differentiation of 2 histologic types of GIM (complete and incomplete) and the consequences of each type. The risk of progression to malignancy is higher with incomplete GIM. It is also higher for those who immigrate from regions with a high incidence of H pylori infection to areas where the incidence is low. Guidelines regarding endoscopic management of GIM vary by geographic region.


Subject(s)
Gastritis, Atrophic , Gastritis , Helicobacter Infections , Helicobacter pylori , Precancerous Conditions , Stomach Neoplasms , Humans , Stomach Neoplasms/etiology , Stomach Neoplasms/prevention & control , Gastritis, Atrophic/complications , Helicobacter Infections/complications , Watchful Waiting , Gastritis/complications , Gastritis/diagnosis , Gastritis/pathology , Precancerous Conditions/complications , Precancerous Conditions/diagnosis , Precancerous Conditions/epidemiology , Metaplasia/complications
SELECTION OF CITATIONS
SEARCH DETAIL
...