1
|
Seitz HK. A narrative review on alcohol and alimentary tract cancer with special emphasis on acetaldehyde and oxidative stress. ZEITSCHRIFT FUR GASTROENTEROLOGIE 2025. [PMID: 40378880 DOI: 10.1055/a-2588-6849] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/19/2025]
Abstract
Approximately 4% of all cancer cases worldwide are caused by alcohol consumption (oropharynx, larynx, esophagus, stomach, colorectum, liver and the female breast). Various mechanisms contribute to ethanol-mediated carcinogenesis, including the action of acetaldehyde, the first metabolite of ethanol oxidation and oxidative stress primarily promoted through the induction of cytochrome P4502E1. Acetaldehyde is toxic and carcinogenic, binds to DNA and proteins, inhibits the oxidative defense- and the nuclear repair system, and prevents DNA methylation. High levels of acetaldehyde occur through increased production in the presence of a hyperactive alcohol dehydrogenase (ADH1C*1,1) or decreased degradation in the presence of low active aldehyde dehydrogenase (ALDH2*1,2). In addition, microbes of the upper alimentary tract and the colorectum effectively produce acetaldehyde from ethanol. In addition, ethanol induces cytochrome P4502E1 resulting in an enhanced ethanol metabolism and the generation of reactive oxygen species (ROS). ROS may cause lipid peroxidation (LPO) with the LPO-products 4-hydroxynonenal or malondialdehyde, which may form highly carcinogenic etheno DNA-adducts CYP2E1 is also involved in the activation of a variety of dietary and tobacco procarcinogens and in the degradation of retinoic acid. Alcohol also influences tumor promotion, such as epigenetics with a change in DNA methylation and histone modification, and affects a variety of cancer genes and signaling pathways. Preventive measures include reducing alcohol consumption, quitting smoking and keeping good oral hygiene. Alcohol consumers - especially when they smoke or belong to genetic risk groups - should be regularly checked for cancer of the upper alimentary tract, for alcohol- associated liver disease, and for breast cancer. Cessation or reduction of alcohol consumption definitively reduces cancer risk.
Collapse
Affiliation(s)
- Helmut Karl Seitz
- Centre of Liver- and Alcohol Diseases, ETHIANUM Klinik, Heidelberg, Germany
- Internal Medicine, Gastroenetrology, Alcohol Research, Faculty of Medicine, University of Heidelberg, Heidelberg, Germany
| |
Collapse
|
2
|
Lee JH, Kim J, Lee DG. The Influence of Lifestyle Behaviors and Body Mass Index Changes on Long-term Outcomes After Gastric Cancer Surgery: A Population-Based Cohort Study. J Gastric Cancer 2025; 25:356-369. [PMID: 40200878 PMCID: PMC11982505 DOI: 10.5230/jgc.2025.25.e18] [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: 08/29/2024] [Revised: 10/26/2024] [Accepted: 02/03/2025] [Indexed: 04/10/2025] Open
Abstract
PURPOSE The present study investigated the impact of lifestyle behaviors and body mass index (BMI) on late recurrence, gastric remnant cancer (GRC), and long-term survival after curative gastrectomy. MATERIALS AND METHODS This retrospective study utilized data from the Korean National Health Insurance claims database. Among 71,014 patients with gastric cancer who underwent curative gastrectomy between January 2009 and December 2012, 23,359 remained cancer-free for five years. Of these, 7,735 patients with health examination data within 2 years before surgery and 5 years after surgery were analyzed for lifestyle behaviors, including smoking, alcohol consumption, and physical activity. Multivariable analysis was used to evaluate the independent effects of these factors and changes in BMI on late recurrence, GRC, and long-term survival. RESULTS Late recurrence or GRC occurred among 628 patients (8.1%). Older age (≥60 years) and total gastrectomy were identified as risk factors. Although lifestyle behaviors and BMI changes did not directly affect recurrence, they significantly affected mortality. In the total gastrectomy group, current underweight status (hazard ratio [HR], 1.586) was associated with increased mortality. Among the partial gastrectomy group, continued smoking (HR, 1.366) and current underweight status (HR, 1.915) increased mortality risk. Conversely, regular physical activity (starting: HR, 0.674; continuing: HR, 0.699) and postoperative overweight or obesity (BMI >25 kg/m²) (HR, 0.713) were associated with reduced mortality. Changes in alcohol consumption showed inconsistent effects between the partial and total gastrectomy groups. CONCLUSIONS The long-term survival of post-gastrectomy patients improved with smoking cessation, regular physical activity, and maintenance of body weight.
Collapse
Affiliation(s)
- Ju-Hee Lee
- Department of Surgery, Hanyang University College of Medicine, Seoul, Korea.
| | - Jiyeong Kim
- Department of Pre-Medicine, College of Medicine, and Biostatistics Laboratory, Medical Research Collaborating Center, Hanyang University, Seoul, Korea
| | - Dong-Gyu Lee
- Department of Surgery, Hanyang University College of Medicine, Seoul, Korea
| |
Collapse
|
3
|
Alorabi MO, El-Bassiouny M, El Khodary DAEG, El Din MMAE, Elsayed AMMA, Reda C. Clinical presentation and treatment outcomes of gastric adenocarcinoma patients: a retrospective study from Ain Shams Clinical Oncology Department. Ecancermedicalscience 2025; 19:1861. [PMID: 40259905 PMCID: PMC12010182 DOI: 10.3332/ecancer.2025.1861] [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: 09/11/2024] [Indexed: 04/23/2025] Open
Abstract
Background Gastric adenocarcinoma (GAC) has a different epidemiological profile in Egypt than in other countries. It ranks 11th in incidence, with 3,285 new cases and 10th in mortality, with 2,469 cases. This retrospective study aims to analyze gastric cancer epidemiology and clinical outcomes in Egyptian patients at Ain Shams University Clinical Oncology Department. Methods We conducted a retrospective analysis of the complete medical records of patients with confirmed GAC at the Ain Shams University Clinical Oncology Department from January 2017 to December 2020. Results This study included 70 patients with GAC. The median age was 52.5 years, with nearly half of cases under 50 years and males representing 53% of the cohort. 70% of patients were from urban areas. Nearly one-third were smokers, with 57.1% having medical comorbidities, mainly diabetes mellitus, hypertension and viral hepatitis. Additionally, 25.7% had a positive family history of GAC. Most Common presenting symptoms were vomiting (42.9%) and abdominal pain (57.1%). 40% of tumours were in the gastric body, and 64.3% were diffuse-type GAC, with 64.3% classified as high grade (III). At presentation, the majority of cases were metastatic (55.7%), with 15.7% presenting with stage II disease and 28.6% with stage III. Most patients (72.8%) had an Eastern Cooperative Oncology Group ≤2. Only 18.6% received neoadjuvant chemotherapy, while 48.6% underwent surgical resection with adequate lymph node dissection in 55.9% of cases. Adjuvant chemotherapy or chemoradiation was administered to 19 patients. The median overall survival (OS) was 11 months, 36 months for stage II, 17 months for stage III and 7 months for stage IV. Univariate analysis indicated that female gender, higher stage (Stage III-IV), higher grade (G IV), absence of neoadjuvant chemotherapy and intestinal type were significantly associated with increased mortality. However, multivariate analysis adjusting for these factors identified the advanced stage as a significant independent predictor of mortality. Conclusion This study identified the distinct GAC profile of Egyptian patients, younger age, aggressive tumours and frequent metastases. These factors contributed to lower OS. Further research and targeted interventions are needed to improve outcomes.
Collapse
Affiliation(s)
- Mohamed Osama Alorabi
- Department of Clinical Oncology and Nuclear Medicine, Faculty of Medicine, Ain Shams University, Cairo 11566, Egypt
| | - Mohamed El-Bassiouny
- Department of Clinical Oncology and Nuclear Medicine, Faculty of Medicine, Ain Shams University, Cairo 11566, Egypt
| | | | - Mai Mohamed Ali Ezz El Din
- Department of Clinical Oncology and Nuclear Medicine, Faculty of Medicine, Ain Shams University, Cairo 11566, Egypt
| | | | - Christine Reda
- Department of Clinical Oncology and Nuclear Medicine, Faculty of Medicine, Ain Shams University, Cairo 11566, Egypt
| |
Collapse
|
4
|
Gonçalves N, Chaves J, Marques- Sá I, Dinis-Ribeiro M, Libânio D. Early diagnosis of gastric cancer: Endoscopy and artificial intelligence. Best Pract Res Clin Gastroenterol 2025:101979. [DOI: 10.1016/j.bpg.2025.101979] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/11/2025]
|
5
|
Wu LW, Jang SJ, Shapiro C, Fazlollahi L, Wang TC, Ryeom SW, Moy RH. Diffuse Gastric Cancer: A Comprehensive Review of Molecular Features and Emerging Therapeutics. Target Oncol 2024; 19:845-865. [PMID: 39271577 PMCID: PMC11557641 DOI: 10.1007/s11523-024-01097-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/30/2024] [Indexed: 09/15/2024]
Abstract
Diffuse-type gastric cancer (DGC) accounts for approximately one-third of gastric cancer diagnoses but is a more clinically aggressive disease with peritoneal metastases and inferior survival compared with intestinal-type gastric cancer (IGC). The understanding of the pathogenesis of DGC has been relatively limited until recently. Multiomic studies, particularly by The Cancer Genome Atlas, have better characterized gastric adenocarcinoma into molecular subtypes. DGC has unique molecular features, including alterations in CDH1, RHOA, and CLDN18-ARHGAP26 fusions. Preclinical models of DGC characterized by these molecular alterations have generated insight into mechanisms of pathogenesis and signaling pathway abnormalities. The currently approved therapies for treatment of gastric cancer generally provide less clinical benefit in patients with DGC. Based on recent phase II/III clinical trials, there is excitement surrounding Claudin 18.2-based and FGFR2b-directed therapies, which capitalize on unique biomarkers that are enriched in the DGC populations. There are numerous therapies targeting Claudin 18.2 and FGFR2b in various stages of preclinical and clinical development. Additionally, there have been preclinical advancements in exploiting unique therapeutic vulnerabilities in several models of DGC through targeting of the focal adhesion kinase (FAK) and Hippo pathways. These preclinical and clinical advancements represent a promising future for the treatment of DGC.
Collapse
Affiliation(s)
- Lawrence W Wu
- Division of Hematology/Oncology, Department of Medicine, Columbia University Irving Medical Center, 161 Fort Washington Avenue, Room 956, New York, NY, 10032, USA
| | - Sung Joo Jang
- Division of Surgical Sciences, Department of Surgery, Columbia University Irving Medical Center, New York, NY, USA
| | - Cameron Shapiro
- Division of Surgical Sciences, Department of Surgery, Columbia University Irving Medical Center, New York, NY, USA
| | - Ladan Fazlollahi
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY, USA
| | - Timothy C Wang
- Division of Digestive and Liver Diseases, Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
| | - Sandra W Ryeom
- Division of Surgical Sciences, Department of Surgery, Columbia University Irving Medical Center, New York, NY, USA
| | - Ryan H Moy
- Division of Hematology/Oncology, Department of Medicine, Columbia University Irving Medical Center, 161 Fort Washington Avenue, Room 956, New York, NY, 10032, USA.
| |
Collapse
|
6
|
Gonzalez-Palacios S, Compañ-Gabucio LM, Torres-Collado L, Oncina-Canovas A, García-de-la-Hera M, Collatuzzo G, Negri E, Pelucchi C, Rota M, López-Carrillo L, Lunet N, Morais S, Ward MH, Martin V, Lozano-Lorca M, Malekzadeh R, Pakseresht M, Hernández-Ramírez RU, Bonzi R, Patel L, López-Cervantes M, Rabkin CS, Tsugane S, Hidaka A, Trichopoulou A, Karakatsani A, Camargo MC, Curado MP, Zhang ZF, La Vecchia C, Boffetta P, Vioque J. The protective effect of dietary folate intake on gastric cancer is modified by alcohol consumption: A pooled analysis of the StoP Consortium. Int J Cancer 2024; 155:1367-1375. [PMID: 38757245 PMCID: PMC11326987 DOI: 10.1002/ijc.35004] [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: 12/11/2023] [Revised: 04/11/2024] [Accepted: 04/17/2024] [Indexed: 05/18/2024]
Abstract
Dietary folate intake has been identified as a potentially modifiable factor of gastric cancer (GC) risk, although the evidence is still inconsistent. We evaluate the association between dietary folate intake and the risk of GC as well as the potential modification effect of alcohol consumption. We pooled data for 2829 histologically confirmed GC cases and 8141 controls from 11 case-control studies from the international Stomach Cancer Pooling Consortium. Dietary folate intake was estimated using food frequency questionnaires. We used linear mixed models with random intercepts for each study to calculate adjusted odds ratios (OR) and 95% confidence interval (CI). Higher folate intake was associated with a lower risk of GC, although this association was not observed among participants who consumed >2.0 alcoholic drinks/day. The OR for the highest quartile of folate intake, compared with the lowest quartile, was 0.78 (95% CI, 0.67-0.90, P-trend = 0.0002). The OR per each quartile increment was 0.92 (95% CI, 0.87-0.96) and, per every 100 μg/day of folate intake, was 0.89 (95% CI, 0.84-0.95). There was a significant interaction between folate intake and alcohol consumption (P-interaction = 0.02). The lower risk of GC associated with higher folate intake was not observed in participants who consumed >2.0 drinks per day, ORQ4v Q1 = 1.15 (95% CI, 0.85-1.56), and the OR100 μg/day = 1.02 (95% CI, 0.92-1.15). Our study supports a beneficial effect of folate intake on GC risk, although the consumption of >2.0 alcoholic drinks/day counteracts this beneficial effect.
Collapse
Affiliation(s)
- Sandra Gonzalez-Palacios
- Epidemiología de la Nutrición, Universidad Miguel Hernández (UMH), Alicante, Spain
- Nutritional Epidemiology Research Group, Instituto de Investigación Sanitaria y Biomédica de Alicante (ISABIAL), Alicante, Spain
- Group 6, Consortium for Biomedical Research in Epidemiology and Public Health (CIBERESP), Madrid, Spain
| | - Laura-María Compañ-Gabucio
- Epidemiología de la Nutrición, Universidad Miguel Hernández (UMH), Alicante, Spain
- Nutritional Epidemiology Research Group, Instituto de Investigación Sanitaria y Biomédica de Alicante (ISABIAL), Alicante, Spain
- Group 6, Consortium for Biomedical Research in Epidemiology and Public Health (CIBERESP), Madrid, Spain
| | - Laura Torres-Collado
- Epidemiología de la Nutrición, Universidad Miguel Hernández (UMH), Alicante, Spain
- Nutritional Epidemiology Research Group, Instituto de Investigación Sanitaria y Biomédica de Alicante (ISABIAL), Alicante, Spain
- Group 6, Consortium for Biomedical Research in Epidemiology and Public Health (CIBERESP), Madrid, Spain
| | - Alejandro Oncina-Canovas
- Epidemiología de la Nutrición, Universidad Miguel Hernández (UMH), Alicante, Spain
- Nutritional Epidemiology Research Group, Instituto de Investigación Sanitaria y Biomédica de Alicante (ISABIAL), Alicante, Spain
- Group 6, Consortium for Biomedical Research in Epidemiology and Public Health (CIBERESP), Madrid, Spain
| | - Manuela García-de-la-Hera
- Epidemiología de la Nutrición, Universidad Miguel Hernández (UMH), Alicante, Spain
- Nutritional Epidemiology Research Group, Instituto de Investigación Sanitaria y Biomédica de Alicante (ISABIAL), Alicante, Spain
- Group 6, Consortium for Biomedical Research in Epidemiology and Public Health (CIBERESP), Madrid, Spain
| | - Giulia Collatuzzo
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - Eva Negri
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - Claudio Pelucchi
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| | - Matteo Rota
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Lizbeth López-Carrillo
- Population Health Research Center, Mexico National Institute of Public Health, Morelos, Mexico
| | - Nuno Lunet
- Cancer Epidemiology, EPIUnit - Instituto de Saúde Pública da Universidade do Porto, Porto, Portugal
- Departamento de Ciências da Saúde Pública e Forenses e Educação Médica, Faculdade de Medicina da Universidade do Porto, Porto, Portugal
- Laboratório para a Investigação Integrativa e Translacional em Saúde Populacional (ITR), Porto, Portugal
| | - Samantha Morais
- Cancer Epidemiology, EPIUnit - Instituto de Saúde Pública da Universidade do Porto, Porto, Portugal
- Departamento de Ciências da Saúde Pública e Forenses e Educação Médica, Faculdade de Medicina da Universidade do Porto, Porto, Portugal
- Laboratório para a Investigação Integrativa e Translacional em Saúde Populacional (ITR), Porto, Portugal
| | - Mary H Ward
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, Maryland, USA
| | - Vicente Martin
- Nutritional Epidemiology Research Group, Instituto de Investigación Sanitaria y Biomédica de Alicante (ISABIAL), Alicante, Spain
- Group of Investigation in Interactions Gene-Environment and Health (GIIGAS), Institute of Biomedicine (IBIOMED), Universidad de León, León, Spain
| | - Macarena Lozano-Lorca
- Departamento de Medicina Preventiva y Salud Pública, Universidad de Granada, Granada, Spain
- Instituto de Investigación Biosanitaria ibs, GRANADA, Granada, Spain
| | - Reza Malekzadeh
- Digestive Oncology Research Center, Digestive Disease Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammadreza Pakseresht
- Digestive Oncology Research Center, Digestive Disease Research Institute, Tehran University of Medical Sciences, Tehran, Iran
- Department of Agricultural, Food and Nutritional Sciences, University of Alberta, Edmonton, Alberta, Canada
- Nutritional Epidemiology Group, Centre for Epidemiology and Biostatistics, University of Leeds, Leeds, UK
| | | | - Rossella Bonzi
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| | - Linia Patel
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| | | | - Charles S Rabkin
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, Maryland, USA
| | - Shoichiro Tsugane
- Division of Cohort Research, National Cancer Center Institute for Cancer Control, Tokyo, Japan
- Graduate School of Public Health, International University of Health and Welfare Graduate School of Public Health, Tokyo, Japan
| | - Akihisa Hidaka
- Division of Cohort Research, National Cancer Center Institute for Cancer Control, Tokyo, Japan
- Department of Diabetes and Endocrinology, JCHO Tokyo Yamate Medical Centre, Tokyo, Japan
| | | | - Anna Karakatsani
- Hellenic Health Foundation, Athens, Greece
- 2nd Pulmonary Medicine Department, Medical School, "ATTIKON" University Hospital, National and Kapodistrian University of Athens, Haidari, Greece
| | - M Constanza Camargo
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, Maryland, USA
| | - Maria Paula Curado
- Centro Internacional de Pesquisa, A. C. Camargo Cancer Center, São Paulo, Brazil
| | - Zuo-Feng Zhang
- Department of Epidemiology, UCLA Fielding School of Public Health and Jonsson Comprehensive Cancer Center, Los Angeles, California, USA
| | - Carlo La Vecchia
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| | - Paolo Boffetta
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
- Stony Brook Cancer Center, Stony Brook University, Stony Brook, New York, USA
| | - Jesús Vioque
- Epidemiología de la Nutrición, Universidad Miguel Hernández (UMH), Alicante, Spain
- Nutritional Epidemiology Research Group, Instituto de Investigación Sanitaria y Biomédica de Alicante (ISABIAL), Alicante, Spain
- Group 6, Consortium for Biomedical Research in Epidemiology and Public Health (CIBERESP), Madrid, Spain
| |
Collapse
|
7
|
Ji K, Shi L, Feng Y, Wang L, Guo H, Li H, Xing J, Xia S, Xu B, Liu E, Zheng Y, Li C, Liu M. Construction and interpretation of machine learning-based prognostic models for survival prediction among intestinal-type and diffuse-type gastric cancer patients. World J Surg Oncol 2024; 22:275. [PMID: 39407221 PMCID: PMC11481450 DOI: 10.1186/s12957-024-03550-y] [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] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Accepted: 10/01/2024] [Indexed: 10/19/2024] Open
Abstract
BACKGROUND Gastric cancer is one of the most common malignant tumors worldwide, with high incidence and mortality rates, and it has a complex etiology and complex pathological features. Depending on the tumor type, gastric cancer can be classified as intestinal-type and diffuse-type gastric cancer, each with distinct pathogenic mechanisms and clinical presentations. In recent years, machine learning techniques have been widely applied in the medical field, offering new perspectives for the diagnosis, treatment, and prognosis of gastric cancer patients. METHODS This study recruited 2158 gastric cancer patients and constructed prognostic prediction models for both intestinal-type and diffuse-type gastric cancer. Clinical pathological data were collected from patients, and machine learning algorithms were used for feature selection and model construction. The performance of the models was validated with training and testing datasets. The Shapley additive explanations (SHAP) values were used to interpret the model predictions and identify the main factors that influence patient survival. RESULTS In the prognostic model for intestinal-type gastric cancer, the gradient boosting decision tree (GBDT) model demonstrated the best performance, with key features including pTNM, CA125, tumor size, CA199, and PALB. Similarly, in the prognostic model for diffuse-type gastric cancer, the GBDT model was utilized, with key features comprising pTNM, Borrmann type IV disease, lymphocyte (LYM), lactate dehydrogenase (LDH), potassium (K), perineural invasion (PNI), tumor size, and whole stomach location. Risk stratification analysis revealed that the prognosis of high-risk patients was significantly worse than that of low-risk patients. CONCLUSION Machine learning shows great potential in predicting survival outcomes of gastric cancer patients, providing strong support for the development of personalized treatment plans.
Collapse
Affiliation(s)
- Kunxiang Ji
- Department of Oncology IV, Beidahuang Industry Group General Hospital, Harbin, China
| | - Lei Shi
- Department of Oncology IV, Beidahuang Industry Group General Hospital, Harbin, China
| | - Yan Feng
- Department of Oncology IV, Beidahuang Industry Group General Hospital, Harbin, China
| | - Linna Wang
- Department of Oncology IV, Beidahuang Industry Group General Hospital, Harbin, China
| | - HuanNan Guo
- Department of Oncology IV, Beidahuang Industry Group General Hospital, Harbin, China
| | - Hui Li
- Department of Oncology IV, Beidahuang Industry Group General Hospital, Harbin, China
| | - Jiacheng Xing
- Department of Oncology IV, Beidahuang Industry Group General Hospital, Harbin, China
| | - Siyu Xia
- Department of Oncology IV, Beidahuang Industry Group General Hospital, Harbin, China
| | - Boran Xu
- Department of Oncology III, Beidahuang Industry Group General Hospital, Harbin, China
| | - Eryu Liu
- Department of Oncology III, Beidahuang Industry Group General Hospital, Harbin, China
| | - YanDan Zheng
- Department of Oncology, Anda City Hospital, Anda, China
| | - Chunfeng Li
- Department of Gastrointestinal Surgery, Harbin Medical University Cancer Hospital, Harbin, China.
| | - Mingyang Liu
- Department of Oncology IV, Beidahuang Industry Group General Hospital, Harbin, China.
| |
Collapse
|
8
|
Xu L, Lyu J, Zheng X, Wang A. Risk Prediction Models for Gastric Cancer: A Scoping Review. J Multidiscip Healthc 2024; 17:4337-4352. [PMID: 39257385 PMCID: PMC11385365 DOI: 10.2147/jmdh.s479699] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Accepted: 08/27/2024] [Indexed: 09/12/2024] Open
Abstract
Background Gastric cancer is a significant contributor to the global cancer burden. Risk prediction models aim to estimate future risk based on current and past information, and can be utilized for risk stratification in population screening programs for gastric cancer. This review aims to explore the research design of existing models, as well as the methods, variables, and performance of model construction. Methods Six databases were searched through to November 4, 2023 to identify appropriate studies. PRISMA extension for scoping reviews and the Arksey and O'Malley framework were followed. Data sources included PubMed, Embase, Web of Science, CNKI, Wanfang, and VIP, focusing on gastric cancer risk prediction model studies. Results A total of 29 articles met the inclusion criteria, from which 28 original risk prediction models were identified that met the analysis criteria. The risk prediction model is screened, and the data extracted includes research characteristics, prediction variables selection, model construction methods and evaluation indicators. The area under the curve (AUC) of the models ranged from 0.560 to 0.989, while the C-statistics varied between 0.684 and 0.940. The number of predictor variables is mainly concentrated between 5 to 11. The top 5 most frequently included variables were age, helicobacter pylori (Hp), precancerous lesion, pepsinogen (PG), sex, and smoking. Age and Hp were the most consistently included variables. Conclusion This review enhances understanding of current gastric cancer risk prediction research and its future directions. The findings provide a strong scientific basis and technical support for developing more accurate gastric cancer risk models. We expect that these conclusions will point the way for future research and clinical practice in this area to assist in the early prevention and treatment of gastric cancer.
Collapse
Affiliation(s)
- Linyu Xu
- Department of Public Service, The First Affiliated Hospital of China Medical University, Shenyang, 110001, People’s Republic of China
| | - Jianxia Lyu
- Department of Public Service, The First Affiliated Hospital of China Medical University, Shenyang, 110001, People’s Republic of China
| | - Xutong Zheng
- Department of Public Service, The First Affiliated Hospital of China Medical University, Shenyang, 110001, People’s Republic of China
| | - Aiping Wang
- Department of Public Service, The First Affiliated Hospital of China Medical University, Shenyang, 110001, People’s Republic of China
| |
Collapse
|
9
|
Pelucchi C, La Vecchia C, Bonzi R, Negri E, Corso G, Boccia S, Boffetta P, Camargo MC, Curado MP, Lunet N, Vioque J, Zhang ZF, StoP Project Working Group. The global gastric cancer consortium: an update from the Stomach cancer Pooling (StoP) project. Eur J Cancer Prev 2024; 33:433-437. [PMID: 38373049 DOI: 10.1097/cej.0000000000000874] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Collaborators] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/21/2024]
Abstract
We updated to December 2023 the main findings of the stomach cancer pooling (StoP) project including about 13 000 cases and 31 000 controls from 29 case-control and 5 nested studies. The StoP project quantified more precisely than previously available the positive associations of tobacco smoking, high alcohol consumption, meat intake, selected occupations (e.g. agricultural and miners), gastric ulcer and family history with gastric cancer and the inverse associations with socioeconomic status and selected aspects of diet (fruits, including citrus fruits, vegetables, including allium and mushrooms, and polyphenols). No consistent associations were found with coffee, yoghurt and leisure-time physical activity, metformin or proton pump inhibitors use.
Collapse
Affiliation(s)
- Claudio Pelucchi
- Department of Clinical Sciences and Community Health, Branch of Medical Statistics, Biometry and Epidemiology 'G.A. Maccacaro', Università degli Studi di Milano, Milan, Italy
| | - Carlo La Vecchia
- Department of Clinical Sciences and Community Health, Branch of Medical Statistics, Biometry and Epidemiology 'G.A. Maccacaro', Università degli Studi di Milano, Milan, Italy
| | - Rossella Bonzi
- Department of Clinical Sciences and Community Health, Branch of Medical Statistics, Biometry and Epidemiology 'G.A. Maccacaro', Università degli Studi di Milano, Milan, Italy
| | - Eva Negri
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - Giovanni Corso
- Division of Breast Surgery, IEO European Institute of Oncology IRCCS
- European Cancer Prevention Organization (ECP)
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Stefania Boccia
- Section of Hygiene, Department of Life Sciences and Public Health, Università Cattolica del Sacro Cuore
- Department of Woman and Child Health and Public Health, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Paolo Boffetta
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
- Stony Brook Cancer Center, Stony Brook University, Stony Brook, New York, USA
| | - M Constanza Camargo
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Rockville, Maryland, USA
| | - Maria Paula Curado
- Centro Internacional de Pesquisa, A. C. Camargo Cancer Center, São Paulo, Brasil
| | - Nuno Lunet
- EPIUnit - Instituto de Saúde Pública da Universidade do Porto
- Laboratório para a Investigação Integrativa e Translacional em Saúde Populacional (ITR)
- Departamento de Ciências da Saúde Pública e Forenses e Educação Médica, Faculdade de Medicina da Universidade do Porto, Porto, Portugal
| | - Jesus Vioque
- Instituto de Investigación Sanitaria y Biomédica de Alicante, Universidad Miguel Hernandez (ISABIAL-UMH), Alicante, Spain
- Consortium for Biomedical Research in Epidemiology and Public Health (CIBERESP), Madrid, Spain
| | - Zuo-Feng Zhang
- Department of Epidemiology, UCLA Fielding School of Public Health and Jonsson Comprehensive Cancer Center, Los Angeles, California, USA
| | | |
Collapse
Collaborators
Alicja Wolk, Amelie Plymoth, Akihisa Hidaka, Anna Karakatsani, Antonia Trichopoulou, Areti Lagiou, Carlo La Vecchia, Charles Rabkin, Claudio Pelucchi, Carlotta Galeone, David Zaridze, Demetrius Albanes, Dmitry Maximovich, Domenico Palli, Emmanuel Dias-Neto, Eva Negri, Evita Gašenko, Farhad Pourfarzi, Federica Turati, Francesca Bravi, Gemma Castaño-Vinyals, Gerson Shigueaki Hamada, Gianfranco Alicandro, Giulia Collatuzzo, Guo-Pei Yu, Jesus Vioque, Jinfu Hu, Joshua Muscat, Juozas Kupcinskas, Kenneth C Johnson, Lina Mu, Linda M Liao, Linia Patel, Lizbeth López-Carrillo, Malaquias López-Cervantes, Marcis Leja, Margherita Pizzato, M Constanza Camargo, Maria Paula Curado, Marta Rossi, Mary H Ward, Matteo Rota, Michele Sassano, Mohammad Derakhshan, Mohammadreza Pakseresht, Monica Ferraroni, Nuno Lunet, Nuria Aragonés, Pagona Lagiou, Paolo Boffetta, Rashmi Sinha, Raul Ulises Hernández-Ramirez, Reza Malekzadeh, Roberta Pastorino, Rossella Bonzi, Samantha Morais, Sandra Gonzalez-Palacios, Shailja Shah, Shoichiro Tsugane, Stefania Boccia, Stephanie Weinstein, Vicente Martin, Weimin Ye, Zuo-Feng Zhang,
Collapse
|
10
|
Hatta W, Koike T, Asano N, Hatayama Y, Ogata Y, Saito M, Jin X, Uno K, Imatani A, Masamune A. The Impact of Tobacco Smoking and Alcohol Consumption on the Development of Gastric Cancers. Int J Mol Sci 2024; 25:7854. [PMID: 39063094 PMCID: PMC11276971 DOI: 10.3390/ijms25147854] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Revised: 07/09/2024] [Accepted: 07/12/2024] [Indexed: 07/23/2024] Open
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.
Collapse
Affiliation(s)
- Waku Hatta
- Division of Gastroenterology, Tohoku University Graduate School of Medicine, Sendai 980-8575, Miyagi, Japan; (T.K.); (Y.H.); (Y.O.); (M.S.); (X.J.); (K.U.); (A.I.); (A.M.)
| | - Tomoyuki Koike
- Division of Gastroenterology, Tohoku University Graduate School of Medicine, Sendai 980-8575, Miyagi, Japan; (T.K.); (Y.H.); (Y.O.); (M.S.); (X.J.); (K.U.); (A.I.); (A.M.)
| | - Naoki Asano
- Division of Gastroenterology, Tohoku University Graduate School of Medicine, Sendai 980-8575, Miyagi, Japan; (T.K.); (Y.H.); (Y.O.); (M.S.); (X.J.); (K.U.); (A.I.); (A.M.)
- Division of Cancer Stem Cell, Miyagi Cancer Center Research Institute, 47-1 Nodayama, Medeshima-Shiode, Natori 981-1293, Miyagi, Japan
- Division of Carcinogenesis and Senescence Biology, Tohoku University Graduate School of Medicine, Natori 981-1293, Miyagi, Japan
| | - Yutaka Hatayama
- Division of Gastroenterology, Tohoku University Graduate School of Medicine, Sendai 980-8575, Miyagi, Japan; (T.K.); (Y.H.); (Y.O.); (M.S.); (X.J.); (K.U.); (A.I.); (A.M.)
| | - Yohei Ogata
- Division of Gastroenterology, Tohoku University Graduate School of Medicine, Sendai 980-8575, Miyagi, Japan; (T.K.); (Y.H.); (Y.O.); (M.S.); (X.J.); (K.U.); (A.I.); (A.M.)
| | - Masahiro Saito
- Division of Gastroenterology, Tohoku University Graduate School of Medicine, Sendai 980-8575, Miyagi, Japan; (T.K.); (Y.H.); (Y.O.); (M.S.); (X.J.); (K.U.); (A.I.); (A.M.)
| | - Xiaoyi Jin
- Division of Gastroenterology, Tohoku University Graduate School of Medicine, Sendai 980-8575, Miyagi, Japan; (T.K.); (Y.H.); (Y.O.); (M.S.); (X.J.); (K.U.); (A.I.); (A.M.)
| | - Kaname Uno
- Division of Gastroenterology, Tohoku University Graduate School of Medicine, Sendai 980-8575, Miyagi, Japan; (T.K.); (Y.H.); (Y.O.); (M.S.); (X.J.); (K.U.); (A.I.); (A.M.)
| | - Akira Imatani
- Division of Gastroenterology, Tohoku University Graduate School of Medicine, Sendai 980-8575, Miyagi, Japan; (T.K.); (Y.H.); (Y.O.); (M.S.); (X.J.); (K.U.); (A.I.); (A.M.)
| | - Atsushi Masamune
- Division of Gastroenterology, Tohoku University Graduate School of Medicine, Sendai 980-8575, Miyagi, Japan; (T.K.); (Y.H.); (Y.O.); (M.S.); (X.J.); (K.U.); (A.I.); (A.M.)
| |
Collapse
|
11
|
Lin JL, Lin JX, Lin GT, Huang CM, Zheng CH, Xie JW, Wang JB, Lu J, Chen QY, Li P. Global incidence and mortality trends of gastric cancer and predicted mortality of gastric cancer by 2035. BMC Public Health 2024; 24:1763. [PMID: 38956557 PMCID: PMC11221210 DOI: 10.1186/s12889-024-19104-6] [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: 07/18/2023] [Accepted: 06/11/2024] [Indexed: 07/04/2024] Open
Abstract
OBJECTIVE To study the historical global incidence and mortality trends of gastric cancer and predicted mortality of gastric cancer by 2035. METHODS Incidence data were retrieved from the Cancer Incidence in Five Continents (CI5) volumes I-XI, and mortality data were obtained from the latest update of the World Health Organization (WHO) mortality database. We used join-point regression analysis to examine historical incidence and mortality trends and used the package NORDPRED in R to predict the number of deaths and mortality rates by 2035 by country and sex. RESULTS More than 1,089,000 new cases of gastric cancer and 769,000 related deaths were reported in 2020. The average annual percent change (AAPC) in the incidence of gastric cancer from 2003 to 2012 among the male population, South Korea, Japan, Malta, Canada, Cyprus, and Switzerland showed an increasing trend (P > 0.05); among the female population, Canada [AAPC, 1.2; (95%Cl, 0.5-2), P < 0.05] showed an increasing trend; and South Korea, Ecuador, Thailand, and Cyprus showed an increasing trend (P > 0.05). AAPC in the mortality of gastric cancer from 2006 to 2015 among the male population, Thailand [3.5 (95%cl, 1.6-5.4), P < 0.05] showed an increasing trend; Malta Island, New Zealand, Turkey, Switzerland, and Cyprus had an increasing trend (P > 0.05); among the male population aged 20-44, Thailand [AAPC, 3.4; (95%cl, 1.3-5.4), P < 0.05] showed an increasing trend; Norway, New Zealand, The Netherlands, Slovakia, France, Colombia, Lithuania, and the USA showed an increasing trend (P > 0.05). It is predicted that the mortality rate in Slovenia and France's female population will show an increasing trend by 2035. It is predicted that the absolute number of deaths in the Israeli male population and in Chile, France, and Canada female population will increase by 2035. CONCLUSION In the past decade, the incidence and mortality of gastric cancer have shown a decreasing trend; however, there are still some countries showing an increasing trend, especially among populations younger than 45 years. Although mortality in most countries is predicted to decline by 2035, the absolute number of deaths due to gastric cancer may further increase due to population growth.
Collapse
Affiliation(s)
- Ju-Li Lin
- Department of Gastric Surgery, Fujian Medical University Union Hospital, No.29 Xinquan Road, Fuzhou , Fujian Province, 350001, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
| | - Jian-Xian Lin
- Department of Gastric Surgery, Fujian Medical University Union Hospital, No.29 Xinquan Road, Fuzhou , Fujian Province, 350001, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, Fujian Province, China
| | - Guang-Tan Lin
- Department of Gastric Surgery, Fujian Medical University Union Hospital, No.29 Xinquan Road, Fuzhou , Fujian Province, 350001, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
| | - Chang-Ming Huang
- Department of Gastric Surgery, Fujian Medical University Union Hospital, No.29 Xinquan Road, Fuzhou , Fujian Province, 350001, China.
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China.
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, Fujian Province, China.
| | - Chao-Hui Zheng
- Department of Gastric Surgery, Fujian Medical University Union Hospital, No.29 Xinquan Road, Fuzhou , Fujian Province, 350001, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
| | - Jian-Wei Xie
- Department of Gastric Surgery, Fujian Medical University Union Hospital, No.29 Xinquan Road, Fuzhou , Fujian Province, 350001, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
| | - Jia-Bin Wang
- Department of Gastric Surgery, Fujian Medical University Union Hospital, No.29 Xinquan Road, Fuzhou , Fujian Province, 350001, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, Fujian Province, China
| | - Jun Lu
- Department of Gastric Surgery, Fujian Medical University Union Hospital, No.29 Xinquan Road, Fuzhou , Fujian Province, 350001, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
| | - Qi-Yue Chen
- Department of Gastric Surgery, Fujian Medical University Union Hospital, No.29 Xinquan Road, Fuzhou , Fujian Province, 350001, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
| | - Ping Li
- Department of Gastric Surgery, Fujian Medical University Union Hospital, No.29 Xinquan Road, Fuzhou , Fujian Province, 350001, China.
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China.
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, Fujian Province, China.
| |
Collapse
|
12
|
Kang K, Bagaoisan MA, Zhang Y. Unveiling the Younger Face of Gastric Cancer: A Comprehensive Review of Epidemiology, Risk Factors, and Prevention Strategies. Cureus 2024; 16:e62826. [PMID: 39036206 PMCID: PMC11260356 DOI: 10.7759/cureus.62826] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/20/2024] [Indexed: 07/23/2024] Open
Abstract
Gastric cancer poses a significant global health challenge, with high incidence and mortality rates each year. Despite advancements in screening and treatment, late detection remains a critical issue. Efforts to address this include raising public awareness and implementing targeted screening programs for high-risk populations. The increasing incidence of gastric cancer among younger individuals underscores the need for lifestyle adjustments and targeted interventions to mitigate risks and improve outcomes. Understanding the various factors contributing to gastric cancer risk is essential for effective prevention strategies, including Helicobacter pylori eradication, lifestyle modifications, and regular screening for high-risk groups. A comprehensive approach addressing both individual behaviors and broader societal factors is crucial in the fight against gastric cancer. This review provides an in-depth examination of gastric cancer epidemiology, risk factors, preventive measures, and screening initiatives, with a particular focus on the rising incidence among younger demographics. Emphasizing the importance of early detection and intervention, the review highlights the need for proactive screening to improve patient outcomes and reduce mortality rates. By addressing these aspects comprehensively, this paper aims to enhance the understanding of gastric cancer dynamics, particularly its incidence among younger individuals, and to inform future strategies for prevention and control.
Collapse
Affiliation(s)
- Kai Kang
- Institute of Nursing, Angeles University Foundation, Angeles City, PHL
| | | | - YuXin Zhang
- Institute of Clinical Nursing, Gansu Health Vocational College, Lanzhou, CHN
| |
Collapse
|
13
|
Mok JW, Oh YH, Magge D, Padmanabhan S. Racial disparities of gastric cancer in the USA: an overview of epidemiology, global screening guidelines, and targeted screening in a heterogeneous population. Gastric Cancer 2024; 27:426-438. [PMID: 38436760 DOI: 10.1007/s10120-024-01475-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Accepted: 01/24/2024] [Indexed: 03/05/2024]
Abstract
Gastric cancer is the fifth most common cancer diagnosis and fourth leading cause of cancer-related death globally. The incidence of gastric cancer in the USA shows significant racial and ethnic disparities with gastric cancer incidence in Korean Americans being over five times higher than in non-Hispanic whites. Since gastric cancer is not common in the USA, there are no current screening guidelines. In countries with higher incidences of gastric cancer, screening guidelines have been implemented for early detection and intervention and this has been associated with a reduction in mortality. Immigrants from high incidence countries develop gastric cancer at lower rates once outside of their country of origin, but continue to be at higher risk for developing gastric cancer. This risk does seem to decrease with subsequent generations. With increasing availability of endoscopy, initiating gastric cancer screening guidelines for high-risk groups can have the potential to improve survival by diagnosing and treating gastric cancer at an earlier stage. This article aims to provide context to gastric cancer epidemiology globally, review risk factors for developing gastric cancer, highlight racial and ethnic disparities in gastric cancer burden in the USA, examine current guidelines that exist in high incidence countries, and suggest future studies examining the efficacy of additional screening in high-risk populations to reduce gastric cancer mortality and disparate burden on ethnic minorities in the USA.
Collapse
Affiliation(s)
- Jean Woo Mok
- Vanderbilt University School of Medicine, Nashville, TN, USA.
| | - Yeong Ha Oh
- Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Deepa Magge
- Division of Surgical Oncology and Endocrine Surgery, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Sekhar Padmanabhan
- Division of Surgical Oncology and Endocrine Surgery, Vanderbilt University Medical Center, Nashville, TN, USA
| |
Collapse
|
14
|
Song M, Jayasekara H, Pelucchi C, Rabkin CS, Johnson KC, Hu J, Palli D, Ferraroni M, Liao LM, Bonzi R, Zaridze D, Maximovitch D, Aragonés N, Martin V, Castaño-Vinyals G, Guevara M, Tsugane S, Hamada GS, Hidaka A, Negri E, Ward MH, Sinha R, Lagiou A, Lagiou P, Boffetta P, Curado MP, Lunet N, Vioque J, Zhang ZF, La Vecchia C, Camargo MC. Reproductive factors, hormonal interventions, and gastric cancer risk in the Stomach cancer Pooling (StoP) Project. Cancer Causes Control 2024; 35:727-737. [PMID: 38123742 PMCID: PMC12052039 DOI: 10.1007/s10552-023-01829-1] [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: 09/28/2023] [Accepted: 11/10/2023] [Indexed: 12/23/2023]
Abstract
BACKGROUND Gastric cancer incidence is higher in men, and a protective hormone-related effect in women is postulated. We aimed to investigate and quantify the relationship in the Stomach cancer Pooling (StoP) Project consortium. METHODS A total of 2,084 cases and 7,102 controls from 11 studies in seven countries were included. Summary odds ratios (ORs) and 95% confidence intervals (CIs) assessing associations of key reproductive factors and menopausal hormone therapy (MHT) with gastric cancer were estimated by pooling study-specific ORs using random-effects meta-analysis. RESULTS A duration of fertility of ≥ 40 years (vs. < 20), was associated with a 25% lower risk of gastric cancer (OR = 0.75; 95% CI: 0.58-0.96). Compared with never use, ever, 5-9 years and ≥ 10 years use of MHT in postmenopausal women, showed ORs of 0.73 (95% CI: 0.58-0.92), 0.53 (95% CI: 0.34-0.84) and 0.71 (95% CI: 0.50-1.00), respectively. The associations were generally similar for anatomical and histologic subtypes. CONCLUSION Our results support the hypothesis that reproductive factors and MHT use may lower the risk of gastric cancer in women, regardless of anatomical or histologic subtypes. Given the variation in hormones over the lifespan, studies should address their effects in premenopausal and postmenopausal women. Furthermore, mechanistic studies may inform potential biological processes.
Collapse
Affiliation(s)
- Minkyo Song
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD, USA
| | - Harindra Jayasekara
- Cancer Epidemiology Division, Cancer Council Victoria, Melbourne, VIC, Australia
- School of Public Health and Preventive Medicine, Faculty of Medicine, Nursing and Health Sciences, Monash University, Clayton, VIC, Australia
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Melbourne, VIC, Australia
| | - Claudio Pelucchi
- Branch of Medical Statistics, Biometry, and Epidemiology "G. A. Maccacaro", Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| | - Charles S Rabkin
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD, USA
| | - Kenneth C Johnson
- School of Epidemiology and Public Health, Department of Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Jinfu Hu
- Harbin Medical University, Harbin, China
| | - Domenico Palli
- Cancer Risk Factors and Life-Style Epidemiology Unit, Institute for Cancer Research, Prevention and Clinical Network, ISPRO, Florence, Italy
| | - Monica Ferraroni
- Branch of Medical Statistics, Biometry, and Epidemiology "G. A. Maccacaro", Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| | - Linda M Liao
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD, USA
| | - Rossella Bonzi
- Branch of Medical Statistics, Biometry, and Epidemiology "G. A. Maccacaro", Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| | - David Zaridze
- Department of Clinical Epidemiology, N.N. Blokhin National Medical Research Center for Oncology, Moscow, Russia
| | - Dmitry Maximovitch
- Department of Clinical Epidemiology, N.N. Blokhin National Medical Research Center for Oncology, Moscow, Russia
| | - Nuria Aragonés
- Consortium for Biomedical Research in Epidemiology and Public Health (CIBERESP), Madrid, Spain
- Cancer Registration and Surveillance Unit, Public Health Division, Department of Health of Madrid, Madrid, Spain
| | - Vicente Martin
- Consortium for Biomedical Research in Epidemiology and Public Health (CIBERESP), Madrid, Spain
- Instituto de Biomedicina (IBIOMED), Universidad de León, León, Spain
| | - Gemma Castaño-Vinyals
- Consortium for Biomedical Research in Epidemiology and Public Health (CIBERESP), Madrid, Spain
- Barcelona Institute for Global Health-ISGlobal, Barcelona, Spain
- IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain
- Universitat Pompeu Fabra (UPF), Barcelona, Spain
| | - Marcela Guevara
- Consortium for Biomedical Research in Epidemiology and Public Health (CIBERESP), Madrid, Spain
- Instituto de Salud Pública y Laboral de Navarra, 31003, Pamplona, Spain
- Navarra Institute for Health Research (IdiSNA), 31008, Pamplona, Spain
| | - Shoichiro Tsugane
- Epidemiology and Prevention Group, Center for Public Health Sciences, National Cancer Center, Tokyo, Japan
- National Institute of Health and Nutrition, National Institutes of Biomedical Innovation, Health and Nutrition, Tokyo, Japan
| | | | - Akihisa Hidaka
- Epidemiology and Prevention Group, Center for Public Health Sciences, National Cancer Center, Tokyo, Japan
| | - Eva Negri
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - Mary H Ward
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD, USA
| | - Rashmi Sinha
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD, USA
| | - Areti Lagiou
- Department of Public and Community Health, School of Public Health, University of West Attica, Athens, Greece
| | - Pagona Lagiou
- Department of Hygiene, Epidemiology and Medical Statistics, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Paolo Boffetta
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
- Stony Brook Cancer Center, Stony Brook University, Stony Brook, NY, USA
| | - Maria Paula Curado
- Centro Internacional de Pesquisa, A. C. Camargo Cancer Center, São Paulo, Brazil
| | - Nuno Lunet
- EPIUnit - Instituto de Saúde Pública da Universidade do Porto, Porto, Portugal
- Laboratório Para a Investigação Integrativa e Translacional em Saúde Populacional (ITR), Porto, Portugal
- Departamento de Ciências da Saúde Pública e Forenses e Educação Médica, Faculdade de Medicina da Universidade do Porto, Porto, Portugal
| | - Jesus Vioque
- Consortium for Biomedical Research in Epidemiology and Public Health (CIBERESP), Madrid, Spain
- Instituto de Investigación Sanitaria y Biomédica de Alicante, Universidad Miguel Hernandez (ISABIAL-UMH), Alicante, Spain
| | - Zuo-Feng Zhang
- Department of Epidemiology, UCLA Fielding School of Public Health and Jonsson Comprehensive Cancer Center, Los Angeles, CA, USA
| | - Carlo La Vecchia
- Branch of Medical Statistics, Biometry, and Epidemiology "G. A. Maccacaro", Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| | - M Constanza Camargo
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD, USA.
| |
Collapse
|
15
|
Kang SJ, Shin CM, Han K, Jung JH, Jin EH, Lim JH, Choi YJ, Yoon H, Park YS, Kim N, Lee DH. Impact of Smoking and Alcohol Consumption on Early-Onset Gastric Cancer Development in Young Koreans: A Population-Based Study. J Gastric Cancer 2024; 24:145-158. [PMID: 38575508 PMCID: PMC10995832 DOI: 10.5230/jgc.2024.24.e2] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 10/22/2023] [Accepted: 10/28/2023] [Indexed: 04/06/2024] Open
Abstract
PURPOSE Although smoking and alcohol consumption are known risk factors for gastric cancer (GC), studies assessing their effects on early-onset GC are limited. In this nationwide, population-based, prospective cohort study, we assessed the effects of smoking and alcohol consumption on early-onset GC in patients aged <50 years. MATERIALS AND METHODS We analyzed data of patients aged 20-39 years who underwent cancer and general health screening in the Korean National Health Screening Program between 2009 and 2012. We calculated the adjusted hazard ratios (aHR) and 95% confidence intervals (CI) for GC incidence until December 2020. RESULTS We enrolled 6,793,699 individuals (men:women=4,077,292:2,716,407) in this cohort. The mean duration of follow-up was 9.4 years. During follow-up, 9,893 cases of GC (men:women=6,304:3,589) were reported. Compared with the aHRs (95% CI) of never-smokers, those of former and current-smokers were 1.121 (1.044-1.205) and 1.282 (1.212-1.355), respectively. Compared with the aHRs (95% CI) of non-consumers, those of low-moderate- and high-risk alcohol consumers were 1.095 (1.046-1.146) and 1.212 (1.113-1.321), respectively. GC risk was the highest in current-smokers and high-risk alcohol consumers (1.447 [1.297-1.615]). Interestingly, alcohol consumption and smoking additively increased the GC risk in men but not in women (Pinteraction=0.002). CONCLUSION Smoking and alcohol consumption are significant risk factors for early-onset GC in young Koreans. Further studies are needed to investigate sex-based impact of alcohol consumption and smoking on GC incidence in young individuals.
Collapse
Affiliation(s)
- Seung Joo Kang
- Department of Internal Medicine, Healthcare Research Institute, Healthcare System Gangnam Center, Seoul National University Hospital, Seoul, Korea
| | - Cheol Min Shin
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Korea. ,
| | - Kyungdo Han
- Department of Statistics and Actuarial Science, Soongsil University, Seoul, Korea. ,
| | - Jin Hyung Jung
- Department of Medical Statistics, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Eun Hyo Jin
- Department of Internal Medicine, Healthcare Research Institute, Healthcare System Gangnam Center, Seoul National University Hospital, Seoul, Korea
| | - Joo Hyun Lim
- Department of Internal Medicine, Healthcare Research Institute, Healthcare System Gangnam Center, Seoul National University Hospital, Seoul, Korea
| | - Yoon Jin Choi
- Division of Gastroenterology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Korea
| | - Hyuk Yoon
- 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
| | - Nayoung Kim
- 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
| |
Collapse
|
16
|
Mishra Y, Ranjan A, Mishra V, Chattaraj A, Aljabali AAA, El-Tanani M, Hromić-Jahjefendić A, Uversky VN, Tambuwala MM. The role of the gut microbiome in gastrointestinal cancers. Cell Signal 2024; 115:111013. [PMID: 38113978 DOI: 10.1016/j.cellsig.2023.111013] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 12/06/2023] [Accepted: 12/13/2023] [Indexed: 12/21/2023]
Abstract
The gut microbiota present in the human digestive system is incredibly varied and is home to trillions of microorganisms. The gut microbiome is shaped at birth, while numerous genetic, dietary, and environmental variables primarily influence the microbiome composition. The importance of gut microbiota on host health is becoming more widely acknowledged. Digestion, intestinal permeability, and immunological and metabolism responses can all be affected by changes in the composition and function of the gut microbiota. There is mounting evidence that the microbial population's complex traits are important biomarkers and indicators of patient outcomes in cancer and its therapies. Numerous studies have demonstrated that changed commensal gut microorganisms contribute to the development and spread of cancer through various routes. Despite the ongoing controversy surrounding the gut microbiome and gastrointestinal cancer, accumulating evidence points to a potentially far more intricate connection than a simple cause-and-effect relationship. SIMPLE SUMMARY: Due to their high frequency and fatality rate, gastrointestinal cancers are regarded as a severe public health issue with complex medical and economic burdens. The gut microbiota may directly or indirectly interact with existing therapies like immunotherapy and chemotherapy, affecting how well a treatment works. The gut microbiome influences the immune response's activity, function, and development. Generally, certain gut bacteria impact the antitumor actions during cancer by creating particular metabolites or triggering T-cell responses. Yet, certain bacterial species have been found to promote cellular proliferation and metastasis in cancer, and comprehending these interactions in the context of cancer may help identify possible treatment targets. Notwithstanding the improvements in the field, additional research is still required to comprehend the underlying processes, examine the effects on existing therapies, and pinpoint certain bacteria and immune cells that can cause this interaction.
Collapse
Affiliation(s)
- Yachana Mishra
- School of Bioengineering and Biosciences, Lovely Professional University, Phagwara 144411, Punjab, India
| | - Abhigyan Ranjan
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara 144411, Punjab, India
| | - Vijay Mishra
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara 144411, Punjab, India
| | - Aditi Chattaraj
- School of Bioengineering and Biosciences, Lovely Professional University, Phagwara 144411, Punjab, India
| | - Alaa A A Aljabali
- Department of Pharmaceutical Sciences, Yarmouk University, Irbid, Jordan
| | - Mohamed El-Tanani
- College of Pharmacy, Ras Alkhama Medical and Health Sciences University, United Arab Emirates
| | - Altijana Hromić-Jahjefendić
- Department of Genetics and Bioengineering, Faculty of Engineering and Natural Sciences, International University of Sarajevo, Hrasnicka cesta 15, Sarajevo 71000, Bosnia and Herzegovina
| | - Vladimir N Uversky
- Department of Molecular Medicine and USF Health Byrd Alzheimer's Research Institute, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA
| | - Murtaza M Tambuwala
- Lincoln Medical School, University of Lincoln, Brayford Pool, Lincoln LN6 7TS, England, United Kingdom.
| |
Collapse
|
17
|
He F, Wang S, Zheng R, Gu J, Zeng H, Sun K, Chen R, Li L, Han B, Li X, Wei W, He J. Trends of gastric cancer burdens attributable to risk factors in China from 2000 to 2050. THE LANCET REGIONAL HEALTH. WESTERN PACIFIC 2024; 44:101003. [PMID: 38269331 PMCID: PMC10806286 DOI: 10.1016/j.lanwpc.2023.101003] [Citation(s) in RCA: 23] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 12/07/2023] [Accepted: 12/24/2023] [Indexed: 01/26/2024]
Abstract
Background The incidence of gastric cancer (GC) decreased in past decades, which was thought largely attributable to risk factors control, yet China still accounts for 44% of global GC burdens. We aimed to estimate changing trajectories of proportions of GC burdens attributable to modifiable risk factors from 2000 to 2050 in China, to inform future targeted preventive strategies. Methods The incidence and new cases of GC were predicted to 2050 using Bayesian age-period-cohort model based on incidence data by anatomical subsites drawn from 682 cancer registries from National Central Cancer Registry. Population attributable fractions (PAFs) were calculated based on prevalence of risk factors and relative risks with GC. Temporal trends of PAFs were described by sex and categories of risk factors using joinpoint analysis. Findings We observed declining trends of PAFs of Helicobacter pylori (H. pylori) infection, smoking, pickled vegetable and alcohol consumption, but increasing trends of PAFs of unhealthy body mass index and diabetes for GC in China. The combined PAFs of these risk factors were estimated to decrease by 10.57% from 2000 to 2050 for GC. We estimated there will be 279,707 GC (122,796 cardia gastric cancer [CGC] and 156,911 non-cardia gastric cancer [NCGC]) cases in 2050. Out of these cases, 70.18% of GC cases could be attributable to modifiable risk factors, while H. pylori infection was predicted to be responsible for 40.7% of CGC and 62.1% of NCGC cases in 2050. Interpretation More than half of GC remained attributable to modifiable risk factors in China. Continued effective strategies on risk factors control are needed to reduce the burden of this highly life-threatening cancer in future. Funding Beijing Nova Program (No. Z201100006820069), CAMS Innovation Fund for Medical Sciences (CIFMS, grant No. 2021-I2M-1-023), CAMS Innovation Fund for Medical Sciences (CIFMS, grant No. 2021-I2M-1-010), Talent Incentive Program of Cancer Hospital Chinese Academy of Medical Sciences (Hope Star).
Collapse
Affiliation(s)
- Feifan He
- Office of National Central Cancer Registry, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Shaoming Wang
- Office of National Central Cancer Registry, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Rongshou Zheng
- Office of National Central Cancer Registry, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jianhua Gu
- Office of National Central Cancer Registry, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Department of Emergency Medicine, Qilu Hospital of Shandong University, Jinan, China
| | - Hongmei Zeng
- Office of National Central Cancer Registry, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Kexin Sun
- Office of National Central Cancer Registry, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Ru Chen
- Office of National Central Cancer Registry, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Li Li
- Office of National Central Cancer Registry, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Bingfeng Han
- Office of National Central Cancer Registry, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xinqing Li
- Office of National Central Cancer Registry, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Wenqiang Wei
- Office of National Central Cancer Registry, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jie He
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
18
|
Ko KP. Risk Factors of Gastric Cancer and Lifestyle Modification for Prevention. J Gastric Cancer 2024; 24:99-107. [PMID: 38225769 PMCID: PMC10774756 DOI: 10.5230/jgc.2024.24.e10] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Revised: 12/14/2023] [Accepted: 12/15/2023] [Indexed: 01/17/2024] Open
Abstract
Gastric cancer has been consistently decreasing worldwide, whereas cardia gastric cancer is on the rise. This indicates that the exposure rates to epidemiological causes are changing. In this study, we aim to review the risk factors for gastric cancer with respect to cardia and non-cardia types. One of the most significant risk factors for gastric cancer is Helicobacter pylori infection. H. pylori infection is known as a risk factor for non-cardia gastric cancer, and there have been results indicating that H. pylori infection is not associated with cardia gastric cancer. However, in the East Asian region, there is epidemiological evidence suggesting that H. pylori infection might be a risk factor for cardia gastric cancer. Smoking and alcohol consumption are known risk factors for gastric cancer, regardless of anatomical location. Obesity is considered a factor in the development of cardia gastric cancer. However, further research is needed to understand the specific relationship with non-cardia gastric cancer. The consumption of high-salt and processed meat is more distinctly associated with non-cardia gastric cancer than in cardia gastric cancer. In addition to these factors, exposure to chemicals and radiation are considered risk factors for gastric cancer. Primary prevention of gastric cancer involves eliminating or avoiding risk factors such as H. pylori eradication and adopting a healthy lifestyle, including quitting smoking, reducing alcohol consumption, maintaining a healthy weight, and having a low-salt diet.
Collapse
Affiliation(s)
- Kwang-Pil Ko
- Clinical Preventive Medicine Center, Seoul National University Bundang Hospital, Seongnam, Korea.
| |
Collapse
|
19
|
Shah D, Bentrem D. Environmental and Genetic Risk Factors for Gastric Cancer. Cancer Treat Res 2024; 192:1-17. [PMID: 39212913 DOI: 10.1007/978-3-031-61238-1_1] [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] [Indexed: 09/04/2024]
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.
Collapse
Affiliation(s)
- Dhavan Shah
- Northwestern Quality Improvement, Research, and Education in Surgery, Department of Surgery, Feinberg School of Medicine, Northwestern University, Evanston, USA
| | - David Bentrem
- Department of Surgery, Feinberg School of Medicine, Northwestern University, Evanston, USA.
- Jesse Brown VA Medical Center, Chicago, USA.
| |
Collapse
|
20
|
Tan Y, Wei Z, Liu K, Qin Y, Hui W. Lifestyle habits and gastric cancer in an East Asian population: a Mendelian randomization study. Front Oncol 2023; 13:1224753. [PMID: 37731647 PMCID: PMC10507616 DOI: 10.3389/fonc.2023.1224753] [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: 06/25/2023] [Accepted: 08/14/2023] [Indexed: 09/22/2023] Open
Abstract
Background Epidemiological evidence suggests an association between lifestyle habits (smoking, alcohol consumption, tea, coffee intake, etc.) and gastric cancer (GC). However, the causal relationship remains uncertain. Therefore, the purpose of this study was to ascertain whether there is a causal connection between them. Methods Two-sample Mendelian randomization (MR) analysis was performed using the publicly available Genome Wide Association Study summary datasets using six methods: inverse variance weighting (IVW), weighted median, MR using a Robust Adjusted Profile Score (MR.Raps), MR using a Robust Adjusted Profile Score (MR-PRESSO), Radial regression of MR, and Causal Analysis Using Summary Effect Estimates (CAUSE). A sensitivity analysis was conducted to assess the robustness of the results. Results In an East Asian population, we found that increased tea intake reduced the risk of GC [odds ratio (OR)= 0.90, 95% confidence interval (CI)= 0.82-0.99, P = 0.037] while there was a positive association between smoking and GC (OR = 1.58, 95% CI = 1.04-2.39, P = 0.032). No causal relationship between alcohol and coffee intake and GC. Sensitivity analyses demonstrated the robustness of these causal associations. Conclusions Our study suggests that tea intake may reduce the risk of GC, for which smoking is a potential risk factor. Nevertheless, a larger and more diverse sample size is needed for further validation.
Collapse
Affiliation(s)
- Yuegui Tan
- Department of Pharmacy, Xi’an Fifth Hospital, Xian, Shaanxi, China
| | - Zhao Wei
- Department of Medicinal Chemistry, School of Pharmacy, Air Force Medical University, Xi’an, Shaanxi, China
| | - Kun Liu
- Department of Epidemiology, Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, School of Public Health, Air Force Medical University, Xi’an, Shaanxi, China
| | - Yuzhen Qin
- Xi’an Jiaotong-liverpool University, XJTLU Wisdom Lake Academy of Pharmacy, Xian, Shaanxi, China
| | - Wenqi Hui
- Department of Pharmacy, Xi’an Fifth Hospital, Xian, Shaanxi, China
| |
Collapse
|
21
|
Livzan MA, Gaus OV, Popello DV. Eating habits and stomach cancer risk. EXPERIMENTAL AND CLINICAL GASTROENTEROLOGY 2023:89-97. [DOI: 10.31146/1682-8658-ecg-211-3-89-97] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/17/2024]
Abstract
Stomach cancer (GC) ranks fifth in the structure of cancer incidence and remains the third leading cause of cancer mortality worldwide. The formation of gastric cancer occurs under the influence of genetic and epigenetic factors. Among the latter, eating habits play a significant role. Primary prevention of cancer through lifestyle and dietary changes is an important and high priority strategy in modern health care. This article presents an overview and systematization of the available data on the influence of nutritional factors on the risk of gastric cancer formation.
Collapse
Affiliation(s)
| | | | - D. V. Popello
- Central State Medical Academy of the Administration of President of the Russian Federation
| |
Collapse
|
22
|
Shin WS, Xie F, Chen B, Yu P, Yu J, To KF, Kang W. Updated Epidemiology of Gastric Cancer in Asia: Decreased Incidence but Still a Big Challenge. Cancers (Basel) 2023; 15:cancers15092639. [PMID: 37174105 PMCID: PMC10177574 DOI: 10.3390/cancers15092639] [Citation(s) in RCA: 42] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 05/02/2023] [Accepted: 05/02/2023] [Indexed: 05/15/2023] Open
Abstract
Despite the decline in incidence and mortality rates, gastric cancer (GC) is the fifth leading cause of cancer deaths worldwide. The incidence and mortality of GC are exceptionally high in Asia due to high H. pylori infection, dietary habits, smoking behaviors, and heavy alcohol consumption. In Asia, males are more susceptible to developing GC than females. Variations in H. pylori strains and prevalence rates may contribute to the differences in incidence and mortality rates across Asian countries. Large-scale H. pylori eradication was one of the effective ways to reduce GC incidences. Treatment methods and clinical trials have evolved, but the 5-year survival rate of advanced GC is still low. Efforts should be put towards large-scale screening and early diagnosis, precision medicine, and deep mechanism studies on the interplay of GC cells and microenvironments for dealing with peritoneal metastasis and prolonging patients' survival.
Collapse
Affiliation(s)
- Wing Sum Shin
- Department of Anatomical and Cellular Pathology, State Key Laboratory of Translational Oncology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong 999077, China
| | - Fuda Xie
- Department of Anatomical and Cellular Pathology, State Key Laboratory of Translational Oncology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong 999077, China
- State Key Laboratory of Digestive Disease, Institute of Digestive Disease, The Chinese University of Hong Kong, Hong Kong 999077, China
- CUHK-Shenzhen Research Institute, The Chinese University of Hong Kong, Shenzhen 518000, China
| | - Bonan Chen
- Department of Anatomical and Cellular Pathology, State Key Laboratory of Translational Oncology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong 999077, China
- State Key Laboratory of Digestive Disease, Institute of Digestive Disease, The Chinese University of Hong Kong, Hong Kong 999077, China
- CUHK-Shenzhen Research Institute, The Chinese University of Hong Kong, Shenzhen 518000, China
| | - Peiyao Yu
- Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Jun Yu
- State Key Laboratory of Digestive Disease, Institute of Digestive Disease, The Chinese University of Hong Kong, Hong Kong 999077, China
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong 999077, China
| | - Ka Fai To
- Department of Anatomical and Cellular Pathology, State Key Laboratory of Translational Oncology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong 999077, China
- State Key Laboratory of Digestive Disease, Institute of Digestive Disease, The Chinese University of Hong Kong, Hong Kong 999077, China
| | - Wei Kang
- Department of Anatomical and Cellular Pathology, State Key Laboratory of Translational Oncology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong 999077, China
- State Key Laboratory of Digestive Disease, Institute of Digestive Disease, The Chinese University of Hong Kong, Hong Kong 999077, China
- CUHK-Shenzhen Research Institute, The Chinese University of Hong Kong, Shenzhen 518000, China
| |
Collapse
|
23
|
Yang WJ, Zhao HP, Yu Y, Wang JH, Guo L, Liu JY, Pu J, Lv J. Updates on global epidemiology, risk and prognostic factors of gastric cancer. World J Gastroenterol 2023; 29:2452-2468. [PMID: 37179585 PMCID: PMC10167900 DOI: 10.3748/wjg.v29.i16.2452] [Citation(s) in RCA: 158] [Impact Index Per Article: 79.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/26/2022] [Revised: 03/19/2023] [Accepted: 04/07/2023] [Indexed: 04/24/2023] Open
Abstract
Gastric cancer (GC) is defined as the primary epithelial malignancy derived from the stomach, and it is a complicated and heterogeneous disease with multiple risk factors. Despite its overall declining trend of incidence and mortality in various countries over the past few decades, GC remains the fifth most common malignancy and the fourth leading cause of cancer-related death globally. Although the global burden of GC has shown a significant downward trend, it remains severe in certain areas, such as Asia. GC ranks third in incidence and mortality among all cancer types in China, and it accounts for nearly 44.0% and 48.6% of new GC cases and GC-related deaths in the world, respectively. The regional differences in GC incidence and mortality are obvious, and annual new cases and deaths are increasing rapidly in some developing regions. Therefore, early preventive and screening strategies for GC are urgently needed. The clinical efficacies of conventional treatments for GC are limited, and the developing understanding of GC pathogenesis has increased the demand for new therapeutic regimens, including immune checkpoint inhibitors, cell immunotherapy and cancer vaccines. The present review describes the epidemiology of GC worldwide, especially in China, summarizes its risk and prognostic factors, and focuses on novel immunotherapies to develop therapeutic strategies for the management of GC patients.
Collapse
Affiliation(s)
- Wen-Juan Yang
- Department of Clinical Laboratory, Honghui Hospital, Xi'an Jiaotong University, Xi'an 710054, Shaanxi Province, China
| | - He-Ping Zhao
- Department of Clinical Laboratory, Honghui Hospital, Xi'an Jiaotong University, Xi'an 710054, Shaanxi Province, China
| | - Yan Yu
- Department of Clinical Laboratory, Honghui Hospital, Xi'an Jiaotong University, Xi'an 710054, Shaanxi Province, China
| | - Ji-Han Wang
- Institute of Medical Research, Northwestern Polytechnical University, Xi'an 710072, Shaanxi Province, China
| | - Lei Guo
- Department of Spinal Surgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an 710054, Shaanxi Province, China
| | - Jun-Ye Liu
- Department of Clinical Laboratory, Honghui Hospital, Xi'an Jiaotong University, Xi'an 710054, Shaanxi Province, China
| | - Jie Pu
- Department of Cardiology, Shaanxi Provincial People’s Hospital, Xi'an 710068, Shaanxi Province, China
| | - Jing Lv
- Department of Clinical Laboratory, Honghui Hospital, Xi'an Jiaotong University, Xi'an 710054, Shaanxi Province, China
| |
Collapse
|
24
|
Thrift AP, Wenker TN, El-Serag HB. Global burden of gastric cancer: epidemiological trends, risk factors, screening and prevention. Nat Rev Clin Oncol 2023; 20:338-349. [PMID: 36959359 DOI: 10.1038/s41571-023-00747-0] [Citation(s) in RCA: 308] [Impact Index Per Article: 154.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/21/2023] [Indexed: 03/25/2023]
Abstract
Gastric cancer remains a major cause of cancer-related mortality worldwide. The temporal trends for this malignancy, however, are dynamic, and reports from the past decade indicate important declines in some regions and demographic groups, as well as a few notable exceptions in which gastric cancer rates are either stable or increasing. Two main anatomical subtypes of gastric cancer exist, non-cardia and cardia, with different temporal trends and risk factors (such as obesity and reflux for cardia gastric cancer and Helicobacter pylori infection for non-cardia gastric cancer). Shifts in the distribution of anatomical locations have been detected in several high-incidence regions. H. pylori is an important aetiological factor for gastric cancer; importantly, the anticipated long-term findings from studies examining the effect of H. pylori eradication on the risk of (re)developing gastric cancer have emerged in the past few years. In this Review, we highlight the latest trends in incidence and mortality using an evidence-based approach. We make the best possible inferences, including clinical and public health inference, on the basis of the quality of the evidence available, and highlight burning questions as well as gaps in knowledge and public health practice that need to be addressed to reduce gastric cancer burden worldwide.
Collapse
Affiliation(s)
- Aaron P Thrift
- Section of Epidemiology and Population Sciences, Department of Medicine, Baylor College of Medicine, Houston, TX, USA
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA
| | - Theresa Nguyen Wenker
- Section of Gastroenterology and Hepatology, Department of Medicine, Baylor College of Medicine, Houston, TX, USA
- Center for Innovations in Quality, Effectiveness and Safety (IQuESt), Michael E. DeBakey Veterans Affairs Medical Center, Houston, TX, USA
| | - Hashem B El-Serag
- Section of Gastroenterology and Hepatology, Department of Medicine, Baylor College of Medicine, Houston, TX, USA.
- Center for Innovations in Quality, Effectiveness and Safety (IQuESt), Michael E. DeBakey Veterans Affairs Medical Center, Houston, TX, USA.
| |
Collapse
|
25
|
Pham C, Nguyen Wenker T, El-Serag HB. Epidemiology of Gastric Intestinal Metaplasia and Gastric Cancer. FOREGUT: THE JOURNAL OF THE AMERICAN FOREGUT SOCIETY 2023; 3:80-88. [DOI: 10.1177/26345161231154024] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
Abstract
Gastric cancer remains one of the most common cancers globally. The pathogenesis of intestinal-type gastric cancer involves pre-malignant stages, including gastric intestinal metaplasia (GIM), the replacement of native gastric foveolar and/or glandular epithelium by intestinal-type epithelium. GIM prevalence is highly variable based on geography and race/ethnicity partly due to the varying prevalence of H. pylori, a potent risk factor. However, gastric cancer incidence does not mirror that of H. pylori, demonstrating a complex interaction between H. pylori and risk factors. We will discuss the epidemiology of GIM and gastric cancer, including incidence trends, risk factors, and implications for future management.
Collapse
Affiliation(s)
- Codey Pham
- Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Theresa Nguyen Wenker
- Department of Medicine, Baylor College of Medicine, Houston, TX, USA
- Center for Innovations in Quality, Effectiveness and Safety (IQuESt), Michael E DeBakey Veterans Affairs Medical Center, Houston, TX, USA
| | - Hashem B El-Serag
- Department of Medicine, Baylor College of Medicine, Houston, TX, USA
- Center for Innovations in Quality, Effectiveness and Safety (IQuESt), Michael E DeBakey Veterans Affairs Medical Center, Houston, TX, USA
| |
Collapse
|
26
|
Wang XY, Wang LL, Xu L, Liang SZ, Yu MC, Zhang QY, Dong QJ. Evaluation of polygenic risk score for risk prediction of gastric cancer. World J Gastrointest Oncol 2023; 15:276-285. [PMID: 36908320 PMCID: PMC9994049 DOI: 10.4251/wjgo.v15.i2.276] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Revised: 01/11/2023] [Accepted: 02/02/2023] [Indexed: 02/14/2023] Open
Abstract
Genetic variations are associated with individual susceptibility to gastric cancer. Recently, polygenic risk score (PRS) models have been established based on genetic variants to predict the risk of gastric cancer. To assess the accuracy of current PRS models in the risk prediction, a systematic review was conducted. A total of eight eligible studies consisted of 544842 participants were included for evaluation of the performance of PRS models. The overall accuracy was moderate with Area under the curve values ranging from 0.5600 to 0.7823. Incorporation of epidemiological factors or Helicobacter pylori (H. pylori) status increased the accuracy for risk prediction, while selection of single nucleotide polymorphism (SNP) and number of SNPs appeared to have little impact on the model performance. To further improve the accuracy of PRS models for risk prediction of gastric cancer, we summarized the association between gastric cancer risk and H. pylori genomic variations, cancer associated bacteria members in the gastric microbiome, discussed the potentials for performance improvement of PRS models with these microbial factors. Future studies on comprehensive PRS models established with human SNPs, epidemiological factors and microbial factors are indicated.
Collapse
Affiliation(s)
- Xiao-Yu Wang
- Central Laboratories and Department of Gastroenterology, Qingdao Municipal Hospital, Qingdao University, Qingdao 266071, Shandong Province, China
| | - Li-Li Wang
- Central Laboratories and Department of Gastroenterology, Qingdao Municipal Hospital, Qingdao University, Qingdao 266071, Shandong Province, China
| | - Lin Xu
- Central Laboratories and Department of Gastroenterology, Qingdao Municipal Hospital, Qingdao University, Qingdao 266071, Shandong Province, China
| | - Shu-Zhen Liang
- Central Laboratories and Department of Gastroenterology, Qingdao Municipal Hospital, Qingdao University, Qingdao 266071, Shandong Province, China
| | - Meng-Chao Yu
- Central Laboratories and Department of Gastroenterology, Qingdao Municipal Hospital, Qingdao University, Qingdao 266071, Shandong Province, China
| | - Qiu-Yue Zhang
- Department of Clinical Laboratory, the Eighth Medical Center of the General Hospital of the People’s Liberation Army, Beijing 100000, China
| | - Quan-Jiang Dong
- Central Laboratories and Department of Gastroenterology, Qingdao Municipal Hospital, Qingdao University, Qingdao 266071, Shandong Province, China
| |
Collapse
|
27
|
Backert S, Linz B, Tegtmeyer N. Helicobacter pylori-Induced Host Cell DNA Damage and Genetics of Gastric Cancer Development. Curr Top Microbiol Immunol 2023; 444:185-206. [PMID: 38231219 DOI: 10.1007/978-3-031-47331-9_7] [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] [Indexed: 01/18/2024]
Abstract
Gastric cancer is a very serious and deadly disease worldwide with about one million new cases every year. Most gastric cancer subtypes are associated with genetic and epigenetic aberrations caused by chromosome instability, microsatellite instability or Epstein-Barr virus infection. Another risk factor is an infection with Helicobacter pylori, which also triggers severe alterations in the host genome. This pathogen expresses an extraordinary repertoire of virulence determinants that take over control of important host cell signaling functions. In fact, H. pylori is a paradigm of persistent infection, chronic inflammation and cellular destruction. In particular, H. pylori profoundly induces chromosomal DNA damage by introducing double-strand breaks (DSBs) followed by genomic instability. DSBs appear in response to oxidative stress and pro-inflammatory transcription during the S-phase of the epithelial cell cycle, which mainly depends on the presence of the bacterial cag pathogenicity island (cagPAI)-encoded type IV secretion system (T4SS). This scenario is closely connected with the T4SS-mediated injection of ADP-glycero-β-D-manno-heptose (ADP-heptose) and oncoprotein CagA. While ADP-heptose links transcription factor NF-κB-induced innate immune signaling with RNA-loop-mediated DNA replication stress and introduction of DSBs, intracellular CagA targets the tumor suppressor BRCA1. The latter scenario promotes BRCAness, a disease characterized by the deficiency of effective DSB repair. In addition, genetic studies of patients demonstrated the presence of gastric cancer-associated single nucleotide polymorphisms (SNPs) in immune-regulatory and other genes as well as specific pathogenic germline variants in several crucial genes involved in homologous recombination and DNA repair, all of which are connected to H. pylori infection. Here we review the molecular mechanisms leading to chromosomal DNA damage and specific genetic aberrations in the presence or absence of H. pylori infection, and discuss their importance in gastric carcinogenesis.
Collapse
Affiliation(s)
- Steffen Backert
- Division of Microbiology, Department of Biology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Staudtstr. 5, 91058, Erlangen, Germany.
| | - Bodo Linz
- Division of Microbiology, Department of Biology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Staudtstr. 5, 91058, Erlangen, Germany
| | - Nicole Tegtmeyer
- Division of Microbiology, Department of Biology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Staudtstr. 5, 91058, Erlangen, Germany.
| |
Collapse
|
28
|
Wysokińska M, Kołota A. Assessment of the Prevalence of Alcoholic Beverage Consumption and Knowledge of the Impact of Alcohol on Health in a Group of Polish Young Adults Aged 18-35: A Cross-Sectional Study. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2022; 19:15425. [PMID: 36497500 PMCID: PMC9737381 DOI: 10.3390/ijerph192315425] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 11/02/2022] [Accepted: 11/18/2022] [Indexed: 06/17/2023]
Abstract
Alcoholic beverages are widely consumed worldwide, especially by young adults. Their excessive consumption is associated with numerous health, social and financial damages. The level of knowledge of young adults about the health effects of consuming alcoholic beverages is low, and research in this area is conducted on small, unrepresentative groups. This cross-sectional study aimed to assess the prevalence of alcoholic beverage consumption and the level of knowledge about the impact of ethyl alcohol on health in a group of people aged 18−35. The survey results indicate that the majority of respondents regularly consume alcoholic beverages (94.6%), and they are at a low risk of excessive consumption (p < 0.0001). The most frequently chosen alcoholic beverage in the studied group was beer, and the least chosen one was vodka. The main motive for reaching for alcoholic beverages was the desire to improve mood. Respondents did not indicate significant changes in alcohol consumption during the COVID-19 pandemic, but participants in the high-risk group more often indicated an increase in alcohol consumption (p = 0.0025). The analysis of the level of knowledge showed that the participants in the study had an average or low level of knowledge about the effects of ethanol on health, with no significant relationships between the study groups. The obtained results indicate a strong need for the continuous education of young people on the effects of the excessive consumption of alcoholic beverages on the body, with particular emphasis on the consequences of using alcohol as a mood-enhancing agent.
Collapse
|
29
|
The mediating role of combined lifestyle factors on the relationship between education and gastric cancer in the Stomach cancer Pooling (StoP) Project. Br J Cancer 2022; 127:855-862. [PMID: 35624300 PMCID: PMC9427973 DOI: 10.1038/s41416-022-01857-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 05/02/2022] [Accepted: 05/10/2022] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND The causal pathway between high education and reduced risk of gastric cancer (GC) has not been explained. The study aimed at evaluating the mediating role of lifestyle factors on the relationship between education and GC METHODS: Ten studies with complete data on education and five lifestyle factors (smoking, alcohol drinking, fruit and vegetable intake, processed meat intake and salt consumption) were selected from a consortium of studies on GC including 4349 GC cases and 8441 controls. We created an a priori score based on the five lifestyle factors, and we carried out a counterfactual-based mediation analysis to decompose the total effect of education on GC into natural direct effect and natural indirect effect mediated by the combined lifestyle factors. Effects were expressed as odds ratios (ORs) with a low level of education as the reference category. RESULTS The natural direct and indirect effects of high versus low education were 0.69 (95% CI: 0.62-0.77) and 0.96 (95% CI: 0.95-0.97), respectively, corresponding to a mediated percentage of 10.1% (95% CI: 7.1-15.4%). The mediation effect was limited to men. CONCLUSIONS The mediation effect of the combined lifestyle factors on the relationship between education and GC is modest. Other potential pathways explaining that relationship warrants further investigation.
Collapse
|
30
|
Uspenskiy YP, Baryshnikova NV, Krasnov AA, Petlenko SV, Apryatina VA. Topical issues of prevention of stomach cancer: A review. CONSILIUM MEDICUM 2022. [DOI: 10.26442/20751753.2022.5.201922] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
Prevention of gastric cancer, both primary and secondary, is an extremely important component of the management of gastroenterological patients. The correct collection of anamnesis with an assessment of the hereditary (family) cancer risk, the action of risk factors (eating disorders, habitual/chronic intoxication, obesity, Helicobacter pylori infection, etc.), as well as the use of gastroprotectors (in particular, the drug Regastim Gastro), especially in persons with potentially precancerous the condition is chronic atrophic gastritis. According to the data of a double-blind placebo-controlled randomized study Regastim Gastro (active ingredient alpha-glutamyl tryptophan) in the treatment of chronic atrophic gastritis, it was found that this drug has a powerful anti-inflammatory effect and regenerative activity. Taking the drug Regastim Gastro, compared with placebo, statistically significantly contributed to a decrease in the number of inflammatory infiltration cells per 1 mm2 of the gastric mucosa. Regastim Gastro decreases in eosinophilic (3 times) and neutrophilic (4 times) infiltration of the gastric mucosa and also reduced the number of macrophages, lymphocytes and plasmocytes. In addition to anti-inflammatory properties, the drug also had a pronounced regenerative effect. Taking of Regastim Gastro statistically significant (p=0.028) increases in the number of glands per 1 mm2 of the gastric mucosa by 26.1% compared with the initial screening indicators. In the group of patients taking placebo, on the contrary, there was a further progression of the pathological process, accompanied by a decrease in the number of glands per 1 mm2 of the gastric mucosa after the end of treatment in comparison with the initial indicators. After the course of therapy, the number of glands per 1 mm2 of the gastric mucosa in patients taking the drug Regastim Gastro was statistically significantly higher in comparison with the results in the placebo group (p=0.013). After the course of Regastim Gastro, there was an improvement in acid production: a shift in the acidic side of the average pH value (1.6 times) and an increase in the value of the acidity index, both when compared with the initial values (5.4 times) and in comparison with the placebo group (2.9 times). The intake of Regastim Gastro to patients with gastritis, both H. pylori (+) and H. pylori (-) before the development of atrophy of the gastric mucosa can reduce the inflammatory factor, prevent the occurrence of atrophy and may have maximum anti-carcinogenic action.
Collapse
|
31
|
Collatuzzo G, Alicandro G, Bertuccio P, Pelucchi C, Bonzi R, Palli D, Ferraroni M, Ye W, Plymoth A, Zaridze D, Maximovich D, Aragones N, Castaño-Vinyals G, Vioque J, Garcia de la Hera M, Zhang ZF, Hu J, Lopez-Carrillo L, López-Cervantes M, Dalmartello M, Mu L, Ward MH, Rabkin C, Yu GP, Camargo MC, Curado MP, Lunet N, Negri E, La Vecchia C, Boffetta P. Peptic ulcer as mediator of the association between risk of gastric cancer and socioeconomic status, tobacco smoking, alcohol drinking and salt intake. J Epidemiol Community Health 2022; 76:jech-2022-219074. [PMID: 35831132 DOI: 10.1136/jech-2022-219074] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 06/25/2022] [Indexed: 12/17/2022]
Abstract
BACKGROUND Peptic ulcer disease (PUD) and gastric cancer (GC) are more prevalent in individuals with low socioeconomic status (SES) and share several risk factors. The aim of this study was to investigate the mediating role of PUD in the association between established risk factors and GC. METHODS We conducted a pooled analysis of 12 studies from the Stomach Cancer Pooling Project Consortium, including a total of 4877 GC cases and 11 808 controls. We explored the mediating role of PUD in the association between SES, tobacco smoking, heavy alcohol drinking and salt intake, and GC. Also, we assessed the ORs and 95% CIs of the risk factors and both PUD and GC. RESULTS PUD mediated 36% of the smoking effect mainly among men. Other risk factors were only slightly mediated by PUD (SES, 5.3%; heavy alcohol drinking, 3.3%; and salt intake, 2.5%). No significant difference was found when excluding PUD diagnosed within 2 years from GC. CONCLUSIONS Our study provides innovative information on the mechanism of stomach mucosal damage leading to PUD and GC, with respect to the effect of tobacco smoking in particular.
Collapse
Affiliation(s)
- Giulia Collatuzzo
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - Gianfranco Alicandro
- Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Milano, Italy
- Cystic Fibrosis Center, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Paola Bertuccio
- Department of Clinical Sciences and Community Health, Branch of Medical Statistics, Biometry and Epidemiology "G.A. Maccacaro", Università degli Studi di Milano, Milan, Italy
- Department of Public Health, Experimental and Forensic Medicine, University of Pavia, Pavia, Italy
| | - Claudio Pelucchi
- Department of Clinical Sciences and Community Health, Branch of Medical Statistics, Biometry and Epidemiology "G.A. Maccacaro", Università degli Studi di Milano, Milan, Italy
| | - Rossella Bonzi
- Department of Clinical Sciences and Community Health, Branch of Medical Statistics, Biometry and Epidemiology "G.A. Maccacaro", Università degli Studi di Milano, Milan, Italy
| | - Domenico Palli
- Cancer Risk Factors and Life-Style Epidemiology Unit, Institute for Cancer Research, Prevention and Clinical Network, ISPRO, Florence, Italy
| | - Monica Ferraroni
- Department of Clinical Sciences and Community Health, Branch of Medical Statistics, Biometry and Epidemiology "G.A. Maccacaro", Università degli Studi di Milano, Milan, Italy
| | - Weimin Ye
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Amelie Plymoth
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - David Zaridze
- Department of Epidemiology and Prevention, Russian N.N. Blokhin Cancer Research Center, Moscow, Russia
| | - Dmitry Maximovich
- Department of Epidemiology and Prevention, Russian N.N. Blokhin Cancer Research Center, Moscow, Russia
| | - Nuria Aragones
- Consortium for Biomedical Research in Epidemiology and Public Health (CIBERESP), Madrid, Spain
- Cancer Epidemiology Section, Public Health Division, Department of Health of Madrid, Madrid, Spain
| | - Gemma Castaño-Vinyals
- Consortium for Biomedical Research in Epidemiology and Public Health (CIBERESP), Madrid, Spain
- Barcelona Institute for Global Health-ISGlobal, Barcelona, Spain
- IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain
- Universitat Pompeu Fabra (UPF), Barcelona, Spain
| | - Jesus Vioque
- Consortium for Biomedical Research in Epidemiology and Public Health (CIBERESP), Madrid, Spain
- Instituto de Investigación Sanitaria y Biomédica de Alicante, Universidad Miguel Hernandez (ISABIAL-UMH), Alicante, Spain
| | - Manoli Garcia de la Hera
- Consortium for Biomedical Research in Epidemiology and Public Health (CIBERESP), Madrid, Spain
- Instituto de Investigación Sanitaria y Biomédica de Alicante, Universidad Miguel Hernandez (ISABIAL-UMH), Alicante, Spain
| | - Zuo-Feng Zhang
- Department of Epidemiology, UCLA Fielding School of Public Health and Jonsson Comprehensive Cancer Center, Los Angeles, California, USA
| | - Jinfu Hu
- Harbin Medical University, Harbin, People's Republic of China
| | | | | | - Michela Dalmartello
- Department of Clinical Sciences and Community Health, Branch of Medical Statistics, Biometry and Epidemiology "G.A. Maccacaro", Università degli Studi di Milano, Milan, Italy
| | - Lina Mu
- Department of Epidemiology and Environmental Health, School of Public Health and Health Professions, University at Buffalo, Buffalo, NY, USA
| | - Mary H Ward
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Rockville, MD, USA
| | - Charles Rabkin
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Rockville, MD, USA
| | - Guo-Pei Yu
- Medical Informatics Center, Peking University, Peking, People's Republic of China
| | - M Constanza Camargo
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Rockville, MD, USA
| | - Maria Paula Curado
- Centro Internacional de Pesquisa, A. C. Camargo Cancer Center, São Paulo, Brazil
| | - Nuno Lunet
- EPIUnit - Instituto de Saúde Pública da Universidade do Porto, Porto, Portugal
- Laboratório para a Investigação Integrativa e Translacional em Saúde Populacional (ITR), Porto, Portugal
- Departamento de Ciências da Saúde Pública e Forenses e Educação Médica, Faculdade de Medicina da Universidade do Porto, Porto, Portugal
| | - Eva Negri
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
- Department of Clinical Sciences and Community Health, Branch of Medical Statistics, Biometry and Epidemiology "G.A. Maccacaro", Università degli Studi di Milano, Milan, Italy
| | - Carlo La Vecchia
- Department of Clinical Sciences and Community Health, Branch of Medical Statistics, Biometry and Epidemiology "G.A. Maccacaro", Università degli Studi di Milano, Milan, Italy
| | - Paolo Boffetta
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
- Stony Brook Cancer Center, Stony Brook University, Stony Brook, New York, USA
| |
Collapse
|
32
|
Wang W, Qiao L, Dong W, Ren J, Chang X, Zhan S, Du P, Xi Y, Wang S. Differences in the Association Between Modifiable Lifestyle Factors and Gastric Precancerous Lesions Among Mongolians and Han Chinese. Front Oncol 2022; 12:798829. [PMID: 35719924 PMCID: PMC9200956 DOI: 10.3389/fonc.2022.798829] [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: 10/20/2021] [Accepted: 05/06/2022] [Indexed: 12/24/2022] Open
Abstract
Background There has been a paucity of evidence examining whether preventable behavioral risk factors led to ethnic differences of gastric precancerous lesions (GPL). We aimed to investigate the ethnic disparity of associations between GPL and lifestyle factors in Mongolian and Han Chinese populations. Methods The study included participants aged 36-75 years enrolled in the Cancer Screening Program during 2016-2017 in Hohhot and Tongliao City, Inner Mongolia. GPL was defined as the gross cascading events (i.e., gastric ulcer, atrophic gastritis, intestinal metaplasia, and dysplasia) that preceded gastric cancer. Results A total of 61638 participants were included, of whom 6863(11·1%) were Mongolians. Alcohol consumption was positively associated with GPL risk in both ethnic groups, but the magnitude was greater in Mongolians (odds ratio (OR) 6·91, 95%CI 5·82-8·28) than in Han Chinese (OR 5·64, 95%CI 5·27-6·04), corresponding to a higher population attributable fraction (PAF) for Mongolians (53·18% vs 43·71%). Besides, the strength of the positive association between physical inactivity and GPL risk was greater among Mongolians (OR 2·02, 95%CI 1·70-2·41; OR 1·09, 95%CI 1·02-1·17 among Han Chinese) with a higher PAF. Smoking was strongly associated with GPL risk in both ethnic groups as well, but the association was more prominent among Han Chinese (OR 5·24 (1·70-2·41) for <10 cigarettes/d, 8·19 (7·48-8·97) for 11-20 cigarettes/d, 7·07 (6·40-7·81) for ≥21 cigarettes/d; the corresponding ORs were 2·96 (2·19-4·00), 6·22 (5·04-7·68), and 7·03 (5·45-9·08) among Mongolians). Lastly, our findings revealed that a significant correlation between insufficient fruits and vegetable consumption and GPL risk was only found among Mongolians (OR 1·27, 95%CI 1·04-1·56). Conclusions Our result suggested that high-risk lifestyle factors should be reduced, particularly in Mongolians. Further studies are needed to elucidate the underlying mechanisms and to reduce health disparities in underserved ethnic groups.
Collapse
Affiliation(s)
- Weiwei Wang
- National Clinical Research Center for Mental Disorders and Beijing Key Laboratory of Mental Disorders, Beijing Anding Hospital, Capital Medical University, Beijing, China
| | - Liying Qiao
- The Inner Mongolia Autonomous Region Comprehensive Center for Disease Control and Prevention, Hohhot, China
| | - Weiqi Dong
- Baotou Medical College, School of Public Health, Baotou, China
| | - Jing Ren
- Baotou Medical College, School of Public Health, Baotou, China
| | - Xiaotian Chang
- Department of Psychology, University of Michigan-Ann Arbor, Ann Arbor, MI, United States
| | - Siyan Zhan
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing, China.,Research Center of Clinical Epidemiology, Peking University Third Hospital, Beijing, China.,Center for Intelligent Public Health, Institute for Artificial Intelligence, Peking University, Beijing, China
| | - Peng Du
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Urology, Peking University Cancer Hospital and Institute, Beijing, China
| | - Yunfeng Xi
- The Inner Mongolia Autonomous Region Comprehensive Center for Disease Control and Prevention, Hohhot, China
| | - Shengfeng Wang
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing, China
| |
Collapse
|
33
|
Shah D, Bentrem D. Environmental and genetic risk factors for gastric cancer. J Surg Oncol 2022; 125:1096-1103. [PMID: 35481919 PMCID: PMC9322002 DOI: 10.1002/jso.26869] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 03/10/2022] [Accepted: 03/12/2022] [Indexed: 12/11/2022]
Abstract
Gastric cancer is a heterogeneous and prevalent disease. The traditional environmental exposures associated with an elevated risk of gastric cancer are less prevalent in the United States 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.
Collapse
Affiliation(s)
- Dhavan Shah
- Department of Surgery, Surgical Outcome and Quality Improvement Center, Feinberg School of MedicineNorthwestern UniversityChicagoIllinoisUSA
| | - David Bentrem
- Department of Surgery, Feinberg School of MedicineNorthwestern UniversityChicagoIllinoisUSA
- Jesse Brown VA Medical CenterChicagoIllinoisUSA
| |
Collapse
|
34
|
LINC00922 promotes deterioration of gastric cancer. PLoS One 2022; 17:e0267798. [PMID: 35511773 PMCID: PMC9070913 DOI: 10.1371/journal.pone.0267798] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 04/12/2022] [Indexed: 12/24/2022] Open
Abstract
Several studies have demonstrated the association of lncRNAs with a variety of cancers. Here, we explored the role of LINC00922 in gastric cancer (GC) using bioinformatics approaches and in vitro experiments. We examined the expression of LINC00922 and the prognosis of GC patients based on data from The Cancer Genome Atlas (TCGA) and Gene Expression Profiling Interactive Analysis (GEPIA). LINC00922-related genes were identified by the Multi Experiment Matrix (MEM) database and The Atlas of Noncoding RNAs in Cancer (TANRIC), followed by Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG) and protein-protein interaction analysis. The significance of LINC00922 in cell proliferation, apoptosis, invasion and migration was assessed by MTT assay, flow cytometry, Transwell assay and wound-healing assay. The expression of LINC00922 was increased in GC tissues compared with adjacent non-tumor tissues, and increased LINC00922 expression was correlated with poor overall survival and disease-free survival. In addition, 336 overlapping genes were identified by the MEM database and TANRIC and found to be involved in GC-related biological processes, such as cell adhesion and migration, as well as TGF-β signaling. In the protein-protein interaction network, hub genes, such as FSTL3 and LAMC1, were identified. LINC00922 overexpression significantly promoted cell proliferation and invasion in vitro, whereas LINC00922 knockdown exerted opposite effects. In summary, our findings indicate that LINC00922 is overexpressed in GC tissues, suggesting that it might play a role in the development and progression of GC, and thus, it might serve as a prognostic indicator of GC.
Collapse
|
35
|
Upper Gastrointestinal Cancer and Liver Cirrhosis. Cancers (Basel) 2022; 14:cancers14092269. [PMID: 35565397 PMCID: PMC9105927 DOI: 10.3390/cancers14092269] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Revised: 04/26/2022] [Accepted: 04/29/2022] [Indexed: 02/01/2023] Open
Abstract
Simple Summary There is a higher incidence rate of upper gastrointestinal cancer in those with liver cirrhosis. The contributing factors include gastric ulcers, congestive gastropathy, zinc deficiency, alcohol drinking, tobacco use and gut microbiota. Most of the de novo malignancies that develop after liver transplantation for cirrhotic patients are upper gastrointestinal cancers. The surgical risk of upper gastrointestinal cancers in cirrhotic patients with advanced liver cirrhosis is higher. Abstract The extended scope of upper gastrointestinal cancer can include esophageal cancer, gastric cancer and pancreatic cancer. A higher incidence rate of gastric cancer and esophageal cancer in patients with liver cirrhosis has been reported. It is attributable to four possible causes which exist in cirrhotic patients, including a higher prevalence of gastric ulcers and congestive gastropathy, zinc deficiency, alcohol drinking and tobacco use and coexisting gut microbiota. Helicobacter pylori infection enhances the development of gastric cancer. In addition, Helicobacter pylori, Porphyromonas gingivalis and Aggregatibacter actinomycetemcomitans also contribute to the development of pancreatic cancer in cirrhotic patients. Cirrhotic patients (especially those with alcoholic liver cirrhosis) who undergo liver transplantation have a higher overall risk of developing de novo malignancies. Most de novo malignancies are upper gastrointestinal malignancies. The prognosis is usually poor. Considering the surgical risk of upper gastrointestinal cancer among those with liver cirrhosis, a radical gastrectomy with D1 or D2 lymph node dissection can be undertaken in Child class A patients. D1 lymph node dissection can be performed in Child class B patients. Endoscopic submucosal dissection for gastric cancer or esophageal cancer can be undertaken safely in selected cirrhotic patients. In Child class C patients, a radical gastrectomy is potentially fatal. Pancreatic radical surgery should be avoided in those with liver cirrhosis with Child class B or a MELD score over 15. The current review focuses on the recent reports on some factors in liver cirrhosis that contribute to the development of upper gastrointestinal cancer. Quitting alcohol drinking and tobacco use is important. How to decrease the risk of the development of gastrointestinal cancer in those with liver cirrhosis remains a challenging problem.
Collapse
|
36
|
Lee HW, Huang D, Shin WK, de la Torre K, Song M, Shin A, Lee JK, Kang D. Frequent low dose alcohol intake increases gastric cancer risk: the Health Examinees-Gem (HEXA-G) study. Cancer Biol Med 2022; 19:j.issn.2095-3941.2021.0642. [PMID: 35484712 PMCID: PMC9425184 DOI: 10.20892/j.issn.2095-3941.2021.0642] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
OBJECTIVE Epidemiological studies indicate that alcohol increases gastric cancer (GC) risk, yet most studies have focused on heavy alcohol intake, leaving other factors understudied. A comprehensive investigation of the effects of the frequency and amount of alcohol intake may help elucidate the GC risk associated with drinking behavior. METHODS The Health Examinees-Gem (HEXA-G) study, a community-based large-scale prospective cohort study, enrolled Korean adults 40-69 years of age between the years 2004 and 2013. Incident GC cases were identified through linkage to Korea Central Cancer Registry data until December 31, 2017. Self-reported questionnaires were used to survey alcohol consumption-related factors (duration, frequency, amount, and type of alcoholic beverages). The frequency and amount of alcohol consumption were combined to explore GC risk according to 4 drinking patterns: "infrequent-light", "frequent-light", "infrequent-heavy", and "frequent-heavy". We used Cox proportional hazard models to estimate the adjusted hazard ratios (HRs) and 95% confidence intervals (CIs), and investigate the relationship between alcohol intake and GC incidence. RESULTS A total of 128,218 participants were included in the analysis. During an average follow-up period of 8.6 years, 462 men and 385 women were diagnosed with GC. In men, current drinkers showed a 31% greater risk of GC than non-drinkers (HR 1.31, 95% CI 1.03-1.66), whereas no significant association was observed in women. In men, GC risk was associated with a higher frequency (P trend 0.02) and dose of ethanol intake in grams (P trend 0.03). In men, the "frequent-light" (≥5 times/week and <40 g ethanol/day) drinking pattern was associated with a 46% greater risk of GC (HR 1.46, 95% CI 1.02-2.07) than the "infrequent-light" pattern (<5 times/week and <40 g ethanol/day). CONCLUSIONS This study suggests that frequent intake of alcohol, even in low quantities per session, increases GC risk. Further research is warranted to evaluate the relationship between alcohol and GC in detail.
Collapse
Affiliation(s)
- Hwi-Won Lee
- Department of Biomedical Sciences, Seoul National University Graduate School, Seoul 03080, Korea.,Department of Preventive Medicine, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Dan Huang
- Department of Preventive Medicine, Seoul National University College of Medicine, Seoul 03080, Korea.,Integrated Major in Innovative Medical Science, Seoul National University Graduate School, Seoul 03080, Korea
| | - Woo-Kyoung Shin
- Department of Preventive Medicine, Seoul National University College of Medicine, Seoul 03080, Korea.,Integrated Major in Innovative Medical Science, Seoul National University Graduate School, Seoul 03080, Korea
| | - Katherine de la Torre
- Department of Biomedical Sciences, Seoul National University Graduate School, Seoul 03080, Korea.,Department of Preventive Medicine, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Minkyo Song
- Department of Preventive Medicine, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Aesun Shin
- Department of Preventive Medicine, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Jong-Koo Lee
- Department of Family Medicine, Seoul National University Hospital, Seoul 03080, Korea
| | - Daehee Kang
- Department of Preventive Medicine, Seoul National University College of Medicine, Seoul 03080, Korea.,Integrated Major in Innovative Medical Science, Seoul National University Graduate School, Seoul 03080, Korea
| |
Collapse
|
37
|
Yang X, Zhang T, Zhang H, Sang S, Chen H, Zuo X. Temporal trend of gastric cancer burden along with its risk factors in China from 1990 to 2019, and projections until 2030: comparison with Japan, South Korea, and Mongolia. Biomark Res 2021; 9:84. [PMID: 34784961 PMCID: PMC8597246 DOI: 10.1186/s40364-021-00340-6] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Accepted: 10/21/2021] [Indexed: 12/25/2022] Open
Abstract
Background Identifying and projecting the epidemiological burden of gastric cancer (GC) can optimize the control strategies, especially in high-burden areas. Methods We collected incidence, deaths, disability-adjusted life-years (DALYs), age-standardized incidence rate (ASIR), age-standardized mortality rate (ASMR), age-standardized DALY rate (ASDR) of GC from 1990 to 2019 in China, Japan, South Korea, and Mongolia from the Global Burden of Disease Study 2019. The average annual percentage change (AAPC) was calculated to quantify the temporal trends, and the projection was estimated by applying the Bayesian age-period-cohort model. Results In China, the ASIR of GC declined slightly from 37.56/100000 in 1990 to 30.64/100000 in 2019 (AAPC of − 0.41), while the declines of ASMR and ASDR were pronounced (AAPC of − 1.68 and − 1.98, respectively), which were weaker than Japan and South Korea. Although the age-standardized rates of gastric cancer in most countries have declined overall in the past 30 years, the downward trend in the last 4 years has become flattened. Smoking remained one main contributor to DALYs of GC in China, Japan, South Korea, and Mongolia, with more than 24%. The contribution from high-sodium diet was similar between men and women, and kept relatively stable over the three decades. The predicted ASMRs among the four East Asian countries continued to decline until 2030, but the absolute deaths would still increase significantly, especially in South Korea and Mongolia. Conclusions Although the age-standardized rates of GC in most countries have declined, the absolute burden of GC in the world, especially in China and Mongolia, is on the rise gradually. Low socio-demographic index and aging along with Helicobacter pylori infection, smoking, and high-salt diet were the main risk factors of GC occurrence and should be paid more attention. Supplementary Information The online version contains supplementary material available at 10.1186/s40364-021-00340-6.
Collapse
Affiliation(s)
- Xiaorong Yang
- Department of Gastroenterology, Qilu Hospital, School of Medicine, Cheeloo College of Medicine, Shandong University, 107 Wenhuaxi Road, Jinan, 250012, Shandong, China. .,Clinical Epidemiology Unit, Qilu Hospital of Shandong University, Jinan, China. .,Laboratory of Translational Gastroenterology, Qilu Hospital of Shandong University, Jinan, China.
| | - Tongchao Zhang
- Clinical Epidemiology Unit, Qilu Hospital of Shandong University, Jinan, China
| | - Hong Zhang
- Department of Gastroenterology, Qilu Hospital, School of Medicine, Cheeloo College of Medicine, Shandong University, 107 Wenhuaxi Road, Jinan, 250012, Shandong, China.,Laboratory of Translational Gastroenterology, Qilu Hospital of Shandong University, Jinan, China
| | - Shaowei Sang
- Clinical Epidemiology Unit, Qilu Hospital of Shandong University, Jinan, China
| | - Hui Chen
- Clinical Epidemiology Unit, Qilu Hospital of Shandong University, Jinan, China
| | - Xiuli Zuo
- Department of Gastroenterology, Qilu Hospital, School of Medicine, Cheeloo College of Medicine, Shandong University, 107 Wenhuaxi Road, Jinan, 250012, Shandong, China. .,Laboratory of Translational Gastroenterology, Qilu Hospital of Shandong University, Jinan, China.
| |
Collapse
|
38
|
Histórico familiar de câncer gástrico em pacientes dispépticos indicados à triagem endoscópica. ACTA PAUL ENFERM 2021. [DOI: 10.37689/acta-ape/2021ao001985] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
|
39
|
Risk Prediction for Gastric Cancer Using GWAS-Identifie Polymorphisms, Helicobacter pylori Infection and Lifestyle-Related Risk Factors in a Japanese Population. Cancers (Basel) 2021; 13:cancers13215525. [PMID: 34771687 PMCID: PMC8583059 DOI: 10.3390/cancers13215525] [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: 09/01/2021] [Revised: 10/27/2021] [Accepted: 10/30/2021] [Indexed: 11/18/2022] Open
Abstract
Simple Summary Gastric cancer remains the major cancer in Japan and worldwide. It is expected that practical intervention strategies for prevention, such as personalized approaches based on genetic risk models, will be developed. Here, we developed and validated a risk prediction model for gastric cancer using genetic, biological, and lifestyle-related risk factors. Results showed that the combination of selected GWAS-identified SNP polymorphisms and other predictors provided high discriminatory accuracy and good calibration in both the derivation and validation studies; however, the contribution of genetic factors to risk prediction was limited. The greatest contributor to risk prediction was ABCD classification (Helicobacter pylori infection-related factor). Abstract Background: As part of our efforts to develop practical intervention applications for cancer prevention, we investigated a risk prediction model for gastric cancer based on genetic, biological, and lifestyle-related risk factors. Methods: We conducted two independent age- and sex-matched case–control studies, the first for model derivation (696 cases and 1392 controls) and the second (795 and 795) for external validation. Using the derivation study data, we developed a prediction model by fitting a conditional logistic regression model using the predictors age, ABCD classification defined by H. pylori infection and gastric atrophy, smoking, alcohol consumption, fruit and vegetable intake, and 3 GWAS-identified polymorphisms. Performance was assessed with regard to discrimination (area under the curve (AUC)) and calibration (calibration plots and Hosmer–Lemeshow test). Results: A combination of selected GWAS-identified polymorphisms and the other predictors provided high discriminatory accuracy and good calibration in both the derivation and validation studies, with AUCs of 0.77 (95% confidence intervals: 0.75–0.79) and 0.78 (0.77–0.81), respectively. The calibration plots of both studies stayed close to the ideal calibration line. In the validation study, the environmental model (nongenetic model) was significantly more discriminative than the inclusive model, with an AUC value of 0.80 (0.77–0.82). Conclusion: The contribution of genetic factors to risk prediction was limited, and the ABCD classification (H. pylori infection-related factor) contributes most to risk prediction of gastric cancer.
Collapse
|
40
|
Zhu Y, Jeong S, Wu M, Zhou JY, Jin ZY, Han RQ, Yang J, Zhang XF, Wang XS, Liu AM, Gu X, Su M, Hu X, Sun Z, Li G, Jung SY, Li L, Mu L, Lu QY, Vecchia CL, Zhao JK, Zhang ZF. Index-based dietary patterns and stomach cancer in a Chinese population. Eur J Cancer Prev 2021; 30:448-456. [PMID: 34292200 PMCID: PMC8487935 DOI: 10.1097/cej.0000000000000705] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
OBJECTIVES Dietary factors are of importance in the development of stomach cancer. This study aims to examine index-based dietary patterns associated with stomach cancer in a Chinese population. METHODS Using data from a population-based case-control study conducted in Jiangsu Province, China, we included a total of 8432 participants (1900 stomach cancer cases and 6532 controls). Dietary data collected by food frequency questionnaire was evaluated by modified Chinese Healthy Eating Index-2016 (mCHEI-2016) and the US Healthy Eating Index-2015 (HEI-2015). Multiple logistic regression analyses were applied to examine the association of mCHEI-2016 and HEI-2015 with stomach cancer while adjusting for potential confounders. The possible interactions between mCHEI-2016 or HEI-2015 and established risk factors were explored. RESULTS Among nonproxy interviews, after adjusting for potential confounding factors, a higher score of sodium, reflecting lower intake per day, was inversely associated with stomach cancer [odds ratio (OR), 0.95; 95% CI, 0.91-0.99 for mCHEI-2016; OR, 0.97; 95% CI, 0.94-0.99 for HEI-2015]. No clear associations with stomach cancer were identified for total scores of HEI-2015 (OR, 0.98; 95% CI, 0.87-1.10 with a 10-point increase, P trend = 0.98) and mCHEI-2016 (OR, 1.05; 95% CI, 0.94-1.17 with a 10-point increase, P trend = 0.22). However, the relation between stomach cancer and the mCHEI-2016 was modified by BMI, with a possible inverse association in normal-weight subjects. CONCLUSIONS Our findings highlight that reduced intake of dietary sodium would prevent the development of stomach cancer. The data indicate a heterogeneity between normal weight and overweight's dietary factors in relation to stomach cancer.
Collapse
Affiliation(s)
- Yuhui Zhu
- Department of Epidemiology, Fielding School of Public Health, University of California at Los Angeles (UCLA), Los Angeles, CA, USA
| | - Somee Jeong
- Department of Epidemiology, Fielding School of Public Health, University of California at Los Angeles (UCLA), Los Angeles, CA, USA
| | - Ming Wu
- Department of Non-communicable Chronic Disease Control, Jiangsu Provincial Center for Disease Control and Prevention, Nanjing, Jiangsu Province, China
| | - Jin-Yi Zhou
- Department of Non-communicable Chronic Disease Control, Jiangsu Provincial Center for Disease Control and Prevention, Nanjing, Jiangsu Province, China
| | - Zi-Yi Jin
- Department of Epidemiology, School of Public Health, Fudan University, Shanghai, China
| | - Ren-Qiang Han
- Department of Non-communicable Chronic Disease Control, Jiangsu Provincial Center for Disease Control and Prevention, Nanjing, Jiangsu Province, China
| | - Jie Yang
- Department of Non-communicable Chronic Disease Control, Jiangsu Provincial Center for Disease Control and Prevention, Nanjing, Jiangsu Province, China
| | - Xiao-Feng Zhang
- Ganyu County Center for Disease Control and Prevention, Ganyu, Jiangsu Province, China
| | - Xu-Shan Wang
- Ganyu County Center for Disease Control and Prevention, Ganyu, Jiangsu Province, China
| | - Ai-Ming Liu
- Dafeng County Center for Disease Control and Prevention, Dafeng, Jiangsu Province, China
| | - Xiaoping Gu
- Dafeng County Center for Disease Control and Prevention, Dafeng, Jiangsu Province, China
| | - Ming Su
- Chuzhou County Center for Disease Control and Prevention, Chuzhou, Jiangsu Province, China
| | - Xu Hu
- Chuzhou County Center for Disease Control and Prevention, Chuzhou, Jiangsu Province, China
| | - Zheng Sun
- Tongshan County Center for Disease Control and Prevention, Tongshan, Jiangsu Province, China
| | - Gang Li
- Tongshan County Center for Disease Control and Prevention, Tongshan, Jiangsu Province, China
| | - Su Yon Jung
- Jonsson Comprehensive Cancer Center, UCLA, Los Angeles, CA, USA
- School of Nursing, UCLA, Los Angeles, CA, USA
| | - Liming Li
- Department of Epidemiology and Biostatistics, Peking University School of Public Health, Beijing, China
| | - Lina Mu
- Department of Social and Preventive Medicine, State University of New York at Buffalo, Buffalo, New York, USA
| | - Qing-Yi Lu
- Center for Human Nutrition, Department of Medicine, UCLA David Geffen School of Medicine, University of California at Los Angeles (UCLA), Los Angeles, CA, USA
| | - Carlo La Vecchia
- Department of Clinical Sciences and Community Health, University of Milan, 20133, Milan, Italy
| | - Jin-Kou Zhao
- Department of Non-communicable Chronic Disease Control, Jiangsu Provincial Center for Disease Control and Prevention, Nanjing, Jiangsu Province, China
| | - Zuo-Feng Zhang
- Department of Epidemiology, Fielding School of Public Health, University of California at Los Angeles (UCLA), Los Angeles, CA, USA
- Jonsson Comprehensive Cancer Center, UCLA, Los Angeles, CA, USA
- Center for Human Nutrition, Department of Medicine, UCLA David Geffen School of Medicine, University of California at Los Angeles (UCLA), Los Angeles, CA, USA
| |
Collapse
|
41
|
Collatuzzo G, Pelucchi C, Negri E, López-Carrillo L, Tsugane S, Hidaka A, Shigueaki Hamada G, Hernández-Ramírez RU, López-Cervantes M, Malekzadeh R, Pourfarzi F, Mu L, Zhang ZF, Lunet N, La Vecchia C, Boffetta P. Exploring the interactions between Helicobacter pylori (Hp) infection and other risk factors of gastric cancer: A pooled analysis in the Stomach cancer Pooling (StoP) Project. Int J Cancer 2021; 149:1228-1238. [PMID: 33990950 DOI: 10.1002/ijc.33678] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 04/12/2021] [Accepted: 04/13/2021] [Indexed: 12/12/2022]
Abstract
Helicobacter pylori (Hp) is crucial in gastric carcinogenesis, but infection alone is not a sufficient cause, and the interaction between Hp infection and other risk factors has not been adequately studied. We conducted a pooled analysis of seven case-control studies from the Stomach cancer Pooling (StoP) Project, comprising 1377 cases and 2470 controls, to explore the interaction among Hp infection and tobacco smoking, alcohol drinking, socioeconomic status (SES) and dietary salt intake on the risk of gastric cancer. We estimated summary odds ratios (ORs) and the corresponding 95% confidence intervals (CIs) by multivariate unconditional logistic regression. The analysis showed no consistent interaction between Hp infection and cigarette smoking, while interaction was more than multiplicative for alcohol drinking (OR = 1.38, 95% CI: 1.07-1.77, P-interaction 0.02) and high intake of salt (OR = 2.62, 95% CI: 1.88-3.65, P-interaction = 0.04). The interaction with SES followed the multiplicative model (P = 0.49), resulting in a weakening among infected individuals of the protective effect of high SES among observed Hp-negative individuals. The interactions found were more pronounced in subjects with history of peptic ulcer. The interactions with Hp infection were stronger for cigarette smoking and dietary salt in the case of noncardia cancer, and for alcohol and SES in the case of cardia cancer. No differences were found when stratifying for histologic type. This large-scale study aimed to quantify the interaction between Hp infection and other modifiable risk factors of gastric cancer revealed that the benefit of combined Hp eradication and lifestyle modification on gastric cancer prevention may be larger than commonly appreciated.
Collapse
Affiliation(s)
- Giulia Collatuzzo
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - Claudio Pelucchi
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| | - Eva Negri
- Department of Biomedical and Clinical Sciences L. Sacco, University of Milan, Milan, Italy
| | | | - Shoichiro Tsugane
- Epidemiology and Prevention Group, Center for Public Health Sciences, National Cancer Center, Tokyo, Japan
| | - Akihisa Hidaka
- Epidemiology and Prevention Group, Center for Public Health Sciences, National Cancer Center, Tokyo, Japan
| | | | | | | | - Reza Malekzadeh
- Digestive Oncology Research Center, Digestive Disease Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Farhad Pourfarzi
- Digestive Oncology Research Center, Digestive Disease Research Institute, Tehran University of Medical Sciences, Tehran, Iran
- Digestive Disease Research Center, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Lina Mu
- Department of Epidemiology and Environmental Health, School of Public Health and Health Professions, University at Buffalo, Buffalo, New York, USA
| | - Zuo-Feng Zhang
- Department of Epidemiology, UCLA Fielding School of Public Health and Jonsson Comprehensive Cancer Center, Los Angeles, California, USA
| | - Nuno Lunet
- Department of Epidemiology, EPIUnit-Instituto de Saúde Pública da Universidade do Porto, Porto, Portugal
- Departamento de Ciências da Saúde Pública e Forenses e Educação Médica, Faculdade de Medicina da Universidade do Porto, Porto, Portugal
| | - Carlo La Vecchia
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| | - Paolo Boffetta
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
- Stony Brook Cancer Center, Stony Brook University, Stony Brook, New York, USA
| |
Collapse
|
42
|
Duan F, Song C, Shi J, Wang P, Ye H, Dai L, Zhang J, Wang K. Identification and epidemiological evaluation of gastric cancer risk factors: based on a field synopsis and meta-analysis in Chinese population. Aging (Albany NY) 2021; 13:21451-21469. [PMID: 34491229 PMCID: PMC8457565 DOI: 10.18632/aging.203484] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2021] [Accepted: 08/11/2021] [Indexed: 12/16/2022]
Abstract
To summarize and assess the credibility and strength of non-genetic factors and genetic variation on gastric cancer risk, we performed a field synopsis and meta-analysis to identify the risk of gastric cancer in Chinese population. Cumulative evidence was graded according to the Venice criteria, and attributable risk percentage (ARP) and population attributable risk percentage (PARP) were used to evaluate the epidemiological effect. A total of 956 studies included non-genetic (404 studies) and genetic factors (552 studies) were quantified, and data on 1161 single nucleotide polymorphisms (SNPs) were available. We identified 14 non-genetic factors were significantly associated with gastric cancer risk. For the analysis of time trends, H. pylori infection rate in gastric cancer and population showed a downward trend. Meanwhile 22 variants were identified significantly associated with gastric cancer: 3 (PLCE1 rs2274223, PSCA rs2976392, MUC1 rs4072037) were high and 19 SNPs were intermediate level of summary evidence, respectively. For non-genetic factors, the top three for ARP were 54.75% (pickled food), 65.87% (stomach disease), and 49.75% (smoked and frying). For PARP were 34.22% (pickled food), 34.24% (edible hot food) and 23.66%(H. pylori infection). On the basis of ARP and PARP associated with SNPs of gastric cancer, the top three for ARP were 53.91% (NAT2, rs1799929),53.05% (NAT2 phenotype), and 42.85% (IL-10, rs1800896). For PARP (Chinese Han in Beijing) were 36.96% (VDR, rs731236), 25.58% (TGFBR2, rs3773651) and 20.56% (MUC1, rs4072037). Our study identified non-genetic risk factors and high-quality biomarkers of gastric cancer susceptibility and their contribution to gastric cancer.
Collapse
Affiliation(s)
- Fujiao Duan
- College of Public Health, Zhengzhou University, Zhengzhou, Henan Province, China
- Key Laboratory of Tumor Epidemiology of Henan Province, Zhengzhou, Henan Province, China
- Medical Research Office, Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
| | - Chunhua Song
- College of Public Health, Zhengzhou University, Zhengzhou, Henan Province, China
- Key Laboratory of Tumor Epidemiology of Henan Province, Zhengzhou, Henan Province, China
| | - Jiachen Shi
- Department of Internal Medicine, The First Affiliated Hospital of Zhengzhou University,Zhengzhou, Henan Province, China
| | - Peng Wang
- College of Public Health, Zhengzhou University, Zhengzhou, Henan Province, China
- Key Laboratory of Tumor Epidemiology of Henan Province, Zhengzhou, Henan Province, China
| | - Hua Ye
- College of Public Health, Zhengzhou University, Zhengzhou, Henan Province, China
- Key Laboratory of Tumor Epidemiology of Henan Province, Zhengzhou, Henan Province, China
| | - Liping Dai
- College of Public Health, Zhengzhou University, Zhengzhou, Henan Province, China
- Key Laboratory of Tumor Epidemiology of Henan Province, Zhengzhou, Henan Province, China
| | - Jianying Zhang
- College of Public Health, Zhengzhou University, Zhengzhou, Henan Province, China
- Key Laboratory of Tumor Epidemiology of Henan Province, Zhengzhou, Henan Province, China
| | - Kaijuan Wang
- College of Public Health, Zhengzhou University, Zhengzhou, Henan Province, China
- Key Laboratory of Tumor Epidemiology of Henan Province, Zhengzhou, Henan Province, China
| |
Collapse
|
43
|
Abstract
In the United States, the incidence of gastric cancer has decreased over the past five decades. However, despite overall decreasing trends in incidence rates of gastric cancer, rates of noncardia gastric cancer among adults aged less than 50 years in the United States are increasing, and most cases of gastric cancer still present with advanced disease and poor resultant survival. Epidemiologic studies have identified the main risk factors for gastric cancer, including increasing age, male sex, non-White race, Helicobacter pylori infection, and smoking. This article summarizes the current epidemiologic evidence with implications for primary and secondary prevention of gastric cancer.
Collapse
Affiliation(s)
- Aaron P Thrift
- Section of Epidemiology and Population Sciences, Department of Medicine, Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA.
| | - Theresa H Nguyen
- Baylor Clinic, 6620 Main Street, MS: BCM620, Room 110D, Houston, TX, 77030, USA
| |
Collapse
|
44
|
Ohnishi I, Iwashita Y, Matsushita Y, Ohtsuka S, Yamashita T, Inaba K, Fukazawa A, Ochiai H, Matsumoto K, Kurono N, Matsushima Y, Mori H, Suzuki S, Suzuki S, Tanioka F, Sugimura H. Mass spectrometric profiling of DNA adducts in the human stomach associated with damage from environmental factors. Genes Environ 2021; 43:12. [PMID: 33836837 PMCID: PMC8034090 DOI: 10.1186/s41021-021-00186-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Accepted: 03/27/2021] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND A comprehensive understanding of DNA adducts, one of the most plausible origins of cancer mutations, is still elusive, especially in human tissues in clinical settings. Recent technological developments have facilitated the identification of multiple DNA adducts in a single experiment. Only a few attempts toward this "DNA adductome approach" in human tissues have been reported. Geospatial information on DNA adducts in human organs has been scarce. AIM Mass spectrometry of human gastric mucosal DNA was performed to identify DNA adducts associated with environmental factors. MATERIALS AND METHODS From 59 subjects who had received gastrectomy for gastric cancer, 306 samples of nontumor tissues and 15 samples of tumors (14 cases) were taken for DNA adductome analysis. Gastric nontumor tissue from autopsies of 7 subjects without gastric cancer (urothelial cancer, hepatocellular carcinoma, lung cancer each; the other four cases were without any cancers) was also investigated. Briefly, DNA was extracted from each sample with antioxidants, digested into nucleosides, separated by liquid chromatography, and then electrospray-ionized. Specific DNA adducts were identified by mass/charge number and column retention time compared to standards. Information on lifestyle factors such as tobacco smoking and alcohol drinking was taken from the clinical records of each subject. RESULTS Seven DNA adducts, including modified bases, C5-methyl-2'-deoxycytidine, 2'-deoxyinosine, C5-hydroxymethyl-2'-deoxycytidine, N6-methyl-2'-deoxyadenosine, 1,N6-etheno-2'-deoxyadenosine, N6-hydroxymethyl-2'-deoxyadenosine, and C8-oxo-2'-deoxyguanosine, were identified in the human stomach and characterized. Intraindividual differences according to the multiple sites of these adducts were noted but were less substantial than interindividual differences. N6-hydroxymethyl-2'-deoxyadenosine was identified in the human stomach for the first time. The amount of C5-hydroxymethyl-2'-deoxycytidine was higher in the stomachs of subjects without gastric cancer than in the nontumor and tumor portions of the stomach in gastric cancer patients. Higher levels of 1,N6-etheno-2'-deoxyadenosine were detected in the subjects who reported both smoking and drinking than in those without these habits. These DNA adducts showed considerable correlations with each other. CONCLUSIONS We characterized 7 DNA adducts in the nontumor portion of the human stomach in both gastric cancer subjects and nongastric cancer subjects. A reduction in C5-hydroxymethyl-dC even in the nontumor mucosa of patients with gastric cancer was observed. Smoking and drinking habits significantly influenced the quantity of one of the lipid peroxidation-derived adducts, etheno-dA. A more expansive DNA adductome profile would provide a comprehensive understanding of the origin of human cancer in the future.
Collapse
Affiliation(s)
- Ippei Ohnishi
- Department of Tumor Pathology, Hamamatsu University School of Medicine, 1-20-1 Handayama, Higashi-ku, Hamamatsu, Shizuoka, 431-3192, Japan
- Pathology Division, Iwata City Hospital, 512-3 Ohkubo, Iwata, Shizuoka, 438-8550, Japan
| | - Yuji Iwashita
- Department of Tumor Pathology, Hamamatsu University School of Medicine, 1-20-1 Handayama, Higashi-ku, Hamamatsu, Shizuoka, 431-3192, Japan.
| | - Yuto Matsushita
- Department of Tumor Pathology, Hamamatsu University School of Medicine, 1-20-1 Handayama, Higashi-ku, Hamamatsu, Shizuoka, 431-3192, Japan
- Department of Urology, Hamamatsu University School of Medicine, 1-20-1 Handayama, Higashi-ku, Hamamatsu, Shizuoka, 431-3192, Japan
| | - Shunsuke Ohtsuka
- Department of Tumor Pathology, Hamamatsu University School of Medicine, 1-20-1 Handayama, Higashi-ku, Hamamatsu, Shizuoka, 431-3192, Japan
- Hamamatsu Medical Center, 328 Tomitsuka-cho, Naka-ku, Hamamatsu, Shizuoka, 432-8580, Japan
| | - Takashi Yamashita
- Department of Tumor Pathology, Hamamatsu University School of Medicine, 1-20-1 Handayama, Higashi-ku, Hamamatsu, Shizuoka, 431-3192, Japan
- First Department of Surgery, Hamamatsu University School of Medicine, 1-20-1 Handayama, Higashi-ku, Hamamatsu, Shizuoka, 431-3192, Japan
| | - Keisuke Inaba
- Surgery Division, Iwata City Hospital, 512-3 Ohkubo, Iwata, Shizuoka, 438-8550, Japan
| | - Atsuko Fukazawa
- Surgery Division, Iwata City Hospital, 512-3 Ohkubo, Iwata, Shizuoka, 438-8550, Japan
| | - Hideto Ochiai
- Surgery Division, Iwata City Hospital, 512-3 Ohkubo, Iwata, Shizuoka, 438-8550, Japan
| | - Keigo Matsumoto
- Surgery Division, Iwata City Hospital, 512-3 Ohkubo, Iwata, Shizuoka, 438-8550, Japan
| | - Nobuhito Kurono
- Department of Chemistry, Hamamatsu University School of Medicine, 1-20-1 Handayama, Higashi-ku, Hamamatsu, Shizuoka, 431-3192, Japan
| | - Yoshitaka Matsushima
- Department of Agricultural Chemistry, Tokyo University of Agriculture, 1-1-1 Sakuragaoka, Setagaya-ku, Tokyo, 156-8502, Japan
| | - Hiroki Mori
- Hamamatsu Medical Center, 328 Tomitsuka-cho, Naka-ku, Hamamatsu, Shizuoka, 432-8580, Japan
| | - Shioto Suzuki
- Pathology Division, Iwata City Hospital, 512-3 Ohkubo, Iwata, Shizuoka, 438-8550, Japan
| | - Shohachi Suzuki
- Surgery Division, Iwata City Hospital, 512-3 Ohkubo, Iwata, Shizuoka, 438-8550, Japan
| | - Fumihiko Tanioka
- Pathology Division, Iwata City Hospital, 512-3 Ohkubo, Iwata, Shizuoka, 438-8550, Japan
| | - Haruhiko Sugimura
- Department of Tumor Pathology, Hamamatsu University School of Medicine, 1-20-1 Handayama, Higashi-ku, Hamamatsu, Shizuoka, 431-3192, Japan.
| |
Collapse
|
45
|
Im PK, Millwood IY, Kartsonaki C, Chen Y, Guo Y, Du H, Bian Z, Lan J, Feng S, Yu C, Lv J, Walters RG, Li L, Yang L, Chen Z. Alcohol drinking and risks of total and site-specific cancers in China: A 10-year prospective study of 0.5 million adults. Int J Cancer 2021; 149:522-534. [PMID: 33634874 PMCID: PMC8359462 DOI: 10.1002/ijc.33538] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Revised: 02/08/2021] [Accepted: 02/09/2021] [Indexed: 12/15/2022]
Abstract
Alcohol drinking is associated with increased risks of several site‐specific cancers, but its role in many other cancers remains inconclusive. Evidence is more limited from China, where cancer rates, drinking patterns and alcohol tolerability differ importantly from Western populations. The prospective China Kadoorie Biobank recruited >512 000 adults aged 30 to 79 years from 10 diverse areas during 2004 to 2008, recording alcohol consumption patterns by a standardised questionnaire. Self‐reported alcohol consumption was estimated as grams of pure alcohol per week based on beverage type, amount consumed per occasion and drinking frequency. After 10 years of follow‐up, 26 961 individuals developed cancer. Cox regression was used to estimate adjusted hazard ratios (HRs) and 95% confidence intervals (CIs) relating alcohol consumption to incidence of site‐specific cancers. Overall, 33% (n = 69 734) of men drank alcohol regularly (ie, ≥weekly) at baseline. Among male current regular drinkers, alcohol intake showed positive dose‐response associations with risks of cancers in the oesophagus (655 events; HR = 1.98 [95%CI 1.79‐2.18], per 280 g/wk), mouth and throat (236; 1.74 [1.48‐2.05]), liver (573; 1.52 [1.31‐1.76]), colon‐rectum (575; 1.19 [1.00‐1.43]), gallbladder (107; 1.60 [1.16‐2.22]) and lung (1017; 1.25 [1.10‐1.42]), similarly among never‐ and ever‐regular smokers. After adjustment for total alcohol intake, there were greater risks of oesophageal cancer in daily drinkers than nondaily drinkers and of liver cancer when drinking without meals. The risks of oesophageal cancer and lung cancer were greater in men reporting flushing after drinking than not. In this male population, alcohol drinking accounted for 7% of cancer cases. Among women, only 2% drank regularly, with no clear associations between alcohol consumption and cancer risk. Among Chinese men, alcohol drinking is associated with increased risks of cancer at multiple sites, with certain drinking patterns (eg, daily, drinking without meals) and low alcohol tolerance further exacerbating the risks.
What's new?
A comprehensive assessment of the role of alcohol in cancer aetiology is needed in China, where cancer rates, drinking patterns, and alcohol tolerability differ from those in the West. In this large prospective study, regular alcohol drinkers had increased risks of cancers in several sites previously considered to be alcohol‐related (i.e., oesophagus, mouth and throat, liver and colon‐rectum) as well as in the lung and gallbladder. Certain drinking patterns (e.g., drinking daily or without meals) and low alcohol tolerance further exacerbated the risks. The findings suggest that lowering population‐levels of alcohol consumption is an important strategy for cancer prevention.
Collapse
Affiliation(s)
- Pek Kei Im
- Clinical Trial Service Unit and Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Iona Y Millwood
- Clinical Trial Service Unit and Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, Oxford, UK.,Medical Research Council Population Health Research Unit (MRC PHRU), Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Christiana Kartsonaki
- Clinical Trial Service Unit and Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, Oxford, UK.,Medical Research Council Population Health Research Unit (MRC PHRU), Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Yiping Chen
- Clinical Trial Service Unit and Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, Oxford, UK.,Medical Research Council Population Health Research Unit (MRC PHRU), Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Yu Guo
- Chinese Academy of Medical Sciences, Beijing, China
| | - Huaidong Du
- Clinical Trial Service Unit and Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, Oxford, UK.,Medical Research Council Population Health Research Unit (MRC PHRU), Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Zheng Bian
- Chinese Academy of Medical Sciences, Beijing, China
| | - Jian Lan
- NCDs Prevention and Control Department, Liuzhou CDC, Liuzhou, China
| | - Shixian Feng
- NCDs Prevention and Control Department, Henan CDC, Zhengzhou, China
| | - Canqing Yu
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing, China
| | - Jun Lv
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing, China
| | - Robin G Walters
- Clinical Trial Service Unit and Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, Oxford, UK.,Medical Research Council Population Health Research Unit (MRC PHRU), Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Liming Li
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing, China
| | - Ling Yang
- Clinical Trial Service Unit and Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, Oxford, UK.,Medical Research Council Population Health Research Unit (MRC PHRU), Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Zhengming Chen
- Clinical Trial Service Unit and Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, Oxford, UK.,Medical Research Council Population Health Research Unit (MRC PHRU), Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | | |
Collapse
|
46
|
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: 18] [Impact Index Per Article: 4.5] [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
|
47
|
Sarhadi V, Mathew B, Kokkola A, Karla T, Tikkanen M, Rautelin H, Lahti L, Puolakkainen P, Knuutila S. Gut microbiota of patients with different subtypes of gastric cancer and gastrointestinal stromal tumors. Gut Pathog 2021; 13:11. [PMID: 33596997 PMCID: PMC7888145 DOI: 10.1186/s13099-021-00403-x] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Accepted: 01/28/2021] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Gastric adenocarcinoma is associated with H. pylori infection and inflammation that can result in the dysbiosis of gastric microbiota. The association of intestinal microbiota with gastric adenocarcinoma subtypes or with gastric gastrointestinal stromal tumors (GIST) is however not well known. Therefore, we performed 16S rRNA gene sequencing on DNA isolated from stool samples of Finnish patients and controls to study differences in microbiota among different histological subtypes of gastric adenocarcinoma, gastric GIST and healthy controls. RESULTS We found that gut microbiota alpha diversity was lowest in diffuse adenocarcinoma patients, followed by intestinal type and GIST patients, although the differences were not significant compared to controls. Beta-diversity analysis however showed significant differences in microbiota composition for all subtypes compared to controls. Significantly higher abundance of Enterobacteriaceae was observed in both adenocarcinoma subtypes, whereas lower abundance of Bifidobacteriaceae was seen only in diffuse adenocarcinoma and of Oscillibacter in intestinal adenocarcinoma. Both GIST and adenocarcinoma patients had higher abundance of Enterobacteriaceae and lower abundance of Lactobacillaceae and Oscillibacter while lower abundance of Lachnoclostridium, Bifidobacterium, Parabacteroides and Barnesiella was seen only in the adenocarcinoma patients. CONCLUSIONS Our analysis shows association of higher Enterobacteriaceae abundance with all types of gastric tumors. Therefore it could be potentially useful as a marker of gastric malignancies. Lower gut microbiota diversity might be indicative of poorly differentiated, invasive, advanced or aggressive tumors and could possibly be a prognostic marker for gastric tumors.
Collapse
Affiliation(s)
- Virinder Sarhadi
- Faculty of Medicine, Department of Pathology, University of Helsinki, 00014, Helsinki, Finland
| | - Binu Mathew
- Department of Computing, University of Turku, Turku, Finland
| | - Arto Kokkola
- The HUCH Gastrointestinal Clinic, University Central Hospital of Helsinki, Helsinki, Finland
| | | | | | - Hilpi Rautelin
- Department of Medical Sciences, Clinical Microbiology, Uppsala University, Uppsala, Sweden
| | - Leo Lahti
- Department of Computing, University of Turku, Turku, Finland
| | - Pauli Puolakkainen
- The HUCH Gastrointestinal Clinic, University Central Hospital of Helsinki, Helsinki, Finland
| | - Sakari Knuutila
- Faculty of Medicine, Department of Pathology, University of Helsinki, 00014, Helsinki, Finland.
| |
Collapse
|
48
|
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
|
49
|
Deng W, Jin L, Zhuo H, Vasiliou V, Zhang Y. Alcohol consumption and risk of stomach cancer: A meta-analysis. Chem Biol Interact 2021; 336:109365. [PMID: 33412155 DOI: 10.1016/j.cbi.2021.109365] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2020] [Revised: 11/27/2020] [Accepted: 12/30/2020] [Indexed: 12/16/2022]
Abstract
Stomach cancer is one of the most common cancers in the world. The relationship between alcohol consumption and the risk of stomach cancer remains unclear. Epidemiology studies investigating this relationship have shown inconsistent findings. A meta-analysis was performed to explore the association between alcohol consumption and increased stomach cancer risk. Eighty-one epidemiology studies, including 68 case-control studies and 13 cohort studies, were included in this study. A significant association was found between alcohol consumption and increased risk of stomach cancer (OR = 1.20, 95% CI 1.12-1.27). To explore the source of the significant heterogeneity (p < 0.05, I2 = 86%), analysis was stratified by study type (case-control study and cohort study), control type (hospital-based control and population-based control), gender (male, female, and mix), race (White and Asian), region (United States, Sweden, China, Japan), subsite of stomach cancer, and type of alcohol. The stratified analyses found that region and cancer subsite are major sources of the high heterogeneity. The inconsistent results in different regions and different subsites might be related to smoking rates, Helicobacter pylori infection, obesity, and potential genetic susceptibility. The positive association between drinking and increased risk of stomach cancer is consistent in stratified analyses. The dose-response analysis showed a clear trend that a higher daily intake of alcohol is associated with a higher risk of stomach cancer.
Collapse
Affiliation(s)
- Wenting Deng
- Department of Environmental Health Sciences, Yale School of Public Health, New Haven, CT, USA
| | - Lan Jin
- Section of Surgical Outcomes and Epidemiology, Department of Surgery, Yale School of Medicine, New Haven, CT, USA
| | - Haoran Zhuo
- Department of Environmental Health Sciences, Yale School of Public Health, New Haven, CT, USA
| | - Vasilis Vasiliou
- Department of Environmental Health Sciences, Yale School of Public Health, New Haven, CT, USA; Yale Cancer Center, New Haven, CT, USA
| | - Yawei Zhang
- National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| |
Collapse
|
50
|
Xu L, Ma Y, Wang S, Feng J, Liu L, Wang J, Liu G, Xiu D, Fu W, Zhan S, Sun T, Gao P. Incidence of gastrointestinal stromal tumor in Chinese urban population: A national population-based study. Cancer Med 2021; 10:737-744. [PMID: 33320439 PMCID: PMC7877389 DOI: 10.1002/cam4.3644] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 11/07/2020] [Accepted: 11/24/2020] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Information on incidence of gastrointestinal stromal tumor (GIST), the most common type of mesenchymal tumor in gastrointestinal tract, was limited in China. This study aimed to estimate the incidence of GIST in urban population from mainland China in 2016. METHODS Urban Employee Basic Medical Insurance (UEBMI) and Urban Residence Basic Medical Insurance (URBMI) in China were used. The denominator of incidence was the total person-years of insured individuals in 2016 in the database, covering approximately 0.43 billion individuals. The numerator was the number of incident GIST cases in 2016. RESULTS The crude incidence in 2016 was 0.40 per 100,000 person-years (95% CI, 0.06-1.03). Male incidence was higher than female incidence (0.44 vs. 0.36, rate ratio: 1.22, p < 0.001). The mean age at diagnosis was 55.20 years (SD = 14.26) and the incidence among those aged 50 years or older was 2.63 times (0.84 vs. 0.32, p < 0.001) higher than those aged under 50. The highest incidence was observed in East China (2.29, 95% CI: 0.46-5.54). CONCLUSIONS The incidence of GIST in mainland China was lower than Europe, North America and Korea. The mean age at diagnosis of GIST in China was younger than that of Europe and Canada. This study provides useful information to further research, policy formulating and management of GIST.
Collapse
Affiliation(s)
- Lu Xu
- Department of Epidemiology and BiostatisticsSchool of Public HealthPeking UniversityBeijingChina
| | - Yanpeng Ma
- Department of General SurgeryPeking University Third HospitalBeijingChina
| | - Shengfeng Wang
- Department of Epidemiology and BiostatisticsSchool of Public HealthPeking UniversityBeijingChina
| | - Jingnan Feng
- Department of Epidemiology and BiostatisticsSchool of Public HealthPeking UniversityBeijingChina
| | - Lili Liu
- Department of Epidemiology and BiostatisticsSchool of Public HealthPeking UniversityBeijingChina
| | - Jinxi Wang
- Shanghai Songsheng Business Consulting Co. LtdBeijingChina
| | - Guozhen Liu
- Peking University Health Information Technology Co. LtdBeijingChina
| | - Dianrong Xiu
- Department of General SurgeryPeking University Third HospitalBeijingChina
| | - Wei Fu
- Department of General SurgeryPeking University Third HospitalBeijingChina
| | - Siyan Zhan
- Department of Epidemiology and BiostatisticsSchool of Public HealthPeking UniversityBeijingChina
- Research Center of Clinical EpidemiologyPeking University Third HospitalBeijingChina
- Center for Intelligent Public HealthInstitute for Artificial IntelligencePeking UniversityBeijingChina
| | - Tao Sun
- Department of General SurgeryPeking University Third HospitalBeijingChina
| | - Pei Gao
- Department of Epidemiology and BiostatisticsSchool of Public HealthPeking UniversityBeijingChina
| |
Collapse
|