1
|
He C, Ye P, Zhang X, Li Y, Li Q, Lü P, Cai C, Cai X. Sex differences in the benefit of tea consumption: A critical summation of the epidemiological evidence. FOOD BIOSCI 2024; 58:103716. [DOI: 10.1016/j.fbio.2024.103716] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
2
|
Rota M, Possenti I, Valsassina V, Santucci C, Bagnardi V, Corrao G, Bosetti C, Specchia C, Gallus S, Lugo A. Dose-response association between cigarette smoking and gastric cancer risk: a systematic review and meta-analysis. Gastric Cancer 2024; 27:197-209. [PMID: 38231449 DOI: 10.1007/s10120-023-01459-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Accepted: 12/16/2023] [Indexed: 01/18/2024]
Abstract
This study aims at providing an accurate and up-to-date quantification of the dose-response association between cigarette smoking and gastric cancer (GC) risk, overall and by subsite. We conducted a systematic review and meta-analysis of case-control and cohort studies on the association between cigarette smoking and GC risk published up to January 2023. We estimated pooled relative risks (RR) of GC and its subsites according to smoking status, intensity, duration, and time since quitting. Among 271 eligible articles, 205 original studies were included in this meta-analysis. Compared with never smokers, the pooled RR for GC was 1.53 (95% confidence interval; CI 1.44-1.62; n = 92) for current and 1.30 (95% CI 1.23-1.37; n = 82) for former smokers. The RR for current compared with never smokers was 2.08 (95% CI 1.66-2.61; n = 21) for gastric cardia and 1.48 (95% CI 1.33-1.66; n = 8) for distal stomach cancer. GC risk nonlinearly increased with smoking intensity up to 20 cigarettes/day (RR:1.69; 95% CI 1.55-1.84) and levelled thereafter. GC risk significantly increased linearly with increasing smoking duration (RR: 1.31; 95% CI 1.25-1.37 for 20 years) and significantly decreased linearly with increasing time since quitting (RR: 0.65; 95% CI 0.44-0.95 for 30 years since cessation). The present meta-analysis confirms that cigarette smoking is an independent risk factor for GC, particularly for gastric cardia. GC risk increases with a low number of cigarettes up to 20 cigarettes/day and increases in a dose-dependent manner with smoking duration.
Collapse
Affiliation(s)
- Matteo Rota
- Department of Molecular and Translational Medicine, Università Degli Studi Di Brescia, Brescia, Italy
| | - Irene Possenti
- Department of Medical Epidemiology, Istituto Di Ricerche Farmacologiche Mario Negri IRCCS, Via Mario Negri 2, 20156, Milan, Italy
| | - Valeria Valsassina
- Department of Medical Epidemiology, Istituto Di Ricerche Farmacologiche Mario Negri IRCCS, Via Mario Negri 2, 20156, Milan, Italy
| | - Claudia Santucci
- Department of Medical Epidemiology, Istituto Di Ricerche Farmacologiche Mario Negri IRCCS, Via Mario Negri 2, 20156, Milan, Italy
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| | - Vincenzo Bagnardi
- Department of Statistics and Quantitative Methods, Università Degli Studi Di Milano-Bicocca, Milan, Italy
| | - Giovanni Corrao
- Laboratory of Healthcare Research and Pharmacoepidemiology, Unit of Biostatistics, Epidemiology and Public Health, Department of Statistics and Quantitative Methods, University of Milano-Bicocca, Milan, Italy
- National Centre of Healthcare Research and Pharmacoepidemiology, University of Milano-Bicocca, Milan, Italy
| | - Cristina Bosetti
- Department of Medical Epidemiology, Istituto Di Ricerche Farmacologiche Mario Negri IRCCS, Via Mario Negri 2, 20156, Milan, Italy
| | - Claudia Specchia
- Department of Molecular and Translational Medicine, Università Degli Studi Di Brescia, Brescia, Italy
| | - Silvano Gallus
- Department of Medical Epidemiology, Istituto Di Ricerche Farmacologiche Mario Negri IRCCS, Via Mario Negri 2, 20156, Milan, Italy
| | - Alessandra Lugo
- Department of Medical Epidemiology, Istituto Di Ricerche Farmacologiche Mario Negri IRCCS, Via Mario Negri 2, 20156, Milan, Italy.
| |
Collapse
|
3
|
Martimianaki G, Alicandro G, Pelucchi C, Bonzi R, Rota M, Hu J, Johnson KC, Rabkin CS, Liao LM, Sinha R, Zhang ZF, Dalmartello M, Lunet N, Morais S, Palli D, Ferraroni M, Yu GP, Tsugane S, Hidaka A, Curado MP, Dias-Neto E, Zaridze D, Maximovitch D, Vioque J, Garcia de la Hera M, López-Carrillo L, Hernández-Ramírez RU, Hamada GS, Ward MH, Mu L, Malekzadeh R, Pourfarzi F, Trichopoulou A, Karakatsani A, Kurtz RC, Lagiou A, Lagiou P, Boccia S, Boffetta P, Camargo MC, Negri E, La Vecchia C. Tea consumption and gastric cancer: a pooled analysis from the Stomach cancer Pooling (StoP) Project consortium. Br J Cancer 2022; 127:726-734. [PMID: 35610368 PMCID: PMC9381730 DOI: 10.1038/s41416-022-01856-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Revised: 04/19/2022] [Accepted: 05/10/2022] [Indexed: 11/09/2022] Open
Abstract
BACKGROUND Evidence from epidemiological studies on the role of tea drinking in gastric cancer risk remains inconsistent. We aimed to investigate and quantify the relationship between tea consumption and gastric cancer in the Stomach cancer Pooling (StoP) Project consortium. METHODS A total of 9438 cases and 20,451 controls from 22 studies worldwide were included. Odds ratios (ORs) and the corresponding 95% confidence intervals (CIs) of gastric cancer for regular versus non-regular tea drinkers were estimated by one and two-stage modelling analyses, including terms for sex, age and the main recognised risk factors for gastric cancer. RESULTS Compared to non-regular drinkers, the estimated adjusted pooled OR for regular tea drinkers was 0.91 (95% CI: 0.85-0.97). When the amount of tea consumed was considered, the OR for consumption of 1-2 cups/day was 1.01 (95% CI: 0.94-1.09) and for >3 cups/day was 0.91 (95% CI: 0.80-1.03). Stronger inverse associations emerged among regular drinkers in China and Japan (OR: 0.67, 95% CI: 0.49-0.91) where green tea is consumed, in subjects with H. pylori infection (OR: 0.68, 95% CI: 0.58-0.80), and for gastric cardia cancer (OR: 0.64, 95% CI: 0.49-0.84). CONCLUSION Our results indicate a weak inverse association between tea consumption and gastric cancer.
Collapse
Affiliation(s)
- Georgia Martimianaki
- Department of Clinical Sciences and Community Health, Branch of Medical Statistics, Biometry and Epidemiology "G.A. Maccacaro", University of Milan, Milan, Italy.
- Hellenic Health Foundation, Athens, Greece.
| | - Gianfranco Alicandro
- Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy
- Cystic Fibrosis Centre, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Claudio Pelucchi
- Department of Clinical Sciences and Community Health, Branch of Medical Statistics, Biometry and Epidemiology "G.A. Maccacaro", University of Milan, Milan, Italy
| | - Rossella Bonzi
- Department of Clinical Sciences and Community Health, Branch of Medical Statistics, Biometry and Epidemiology "G.A. Maccacaro", University of Milan, Milan, Italy
| | - Matteo Rota
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Jinfu Hu
- Harbin Medical University, Harbin, China
| | - Kenneth C Johnson
- School of Epidemiology and Public Health, Department of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Charles S Rabkin
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Rockville, MD, USA
| | - Linda M Liao
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Rockville, MD, USA
| | - Rashmi Sinha
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Rockville, MD, USA
| | - Zuo-Feng Zhang
- Department of Epidemiology, UCLA Fielding School of Public Health and Jonsson Comprehensive Cancer Center, Los Angeles, CA, USA
| | - Michela Dalmartello
- Department of Clinical Sciences and Community Health, Branch of Medical Statistics, Biometry and Epidemiology "G.A. Maccacaro", University of Milan, Milan, Italy
| | - Nuno Lunet
- 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
- 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
| | - 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", University of Milan, Milan, Italy
| | - Guo-Pei Yu
- Medical Informatics Center, Peking University, Peking, China
| | - 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
| | - Maria Paula Curado
- Centro Internacional de Pesquisas, A.C. Camargo Cancer Center, São Paulo, Brazil
| | - Emmanuel Dias-Neto
- Centro Internacional de Pesquisas, A.C. Camargo Cancer Center, São Paulo, Brazil
| | - 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
| | - 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
| | | | | | | | - Mary H Ward
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Rockville, MD, USA
| | - Lina Mu
- Department of Epidemiology and Environmental Health, School of Public Health and Health Professions, University at Buffalo, Buffalo, NY, USA
| | - Reza Malekzadeh
- Digestive Oncology Research Center, Digestive Disease Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Farhad Pourfarzi
- Digestive Disease Research Center, Ardabil University of Medical Sciences, Ardabil, Iran
| | | | - Anna Karakatsani
- Hellenic Health Foundation, Athens, Greece
- 2nd Pulmonary Medicine Department, National and Kapodistrian University of Athens, Medical School, "ATTIKON" University Hospital, Haidari, Greece
| | - Robert C Kurtz
- Department of Medicine, Memorial Sloan Kettering Cancer Centre, New York, NY, 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
| | - Stefania Boccia
- Section of Hygiene, University Department of Life Sciences and Public Health, Università Cattolica del Sacro Cuore, Roma, Italia
- Department of Woman and Child Health and Public Health-Public Health Area, Fondazione Policlinico Universitario A. Gemelli IRCCS, Roma, Italia
| | - Paolo Boffetta
- Stony Brook Cancer Center, Stony Brook University, Stony Brook, NY, USA
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - M Constanza Camargo
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Rockville, MD, USA
| | - Eva Negri
- Department of Clinical Sciences and Community Health, Branch of Medical Statistics, Biometry and Epidemiology "G.A. Maccacaro", University of Milan, Milan, Italy
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
- Pegaso Online University, Naples, Italy
| | - Carlo La Vecchia
- Department of Clinical Sciences and Community Health, Branch of Medical Statistics, Biometry and Epidemiology "G.A. Maccacaro", University of Milan, Milan, Italy
| |
Collapse
|
4
|
Upper Gastrointestinal Cancer and Liver Cirrhosis. Cancers (Basel) 2022; 14:cancers14092269. [PMID: 35565397 PMCID: PMC9105927 DOI: 10.3390/cancers14092269] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [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
|
5
|
Nguyen NLT, Dang NDT, Dang QH, Tran VC, Vo HL, Yamaguchi M, Ta TV. Polymorphism of MUC1 Gene in Vietnamese Gastric Cancer Patients: A Multicenter Case-Control Study. Front Oncol 2021; 11:694977. [PMID: 34532288 PMCID: PMC8439541 DOI: 10.3389/fonc.2021.694977] [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: 04/14/2021] [Accepted: 08/10/2021] [Indexed: 12/31/2022] Open
Abstract
Background A few studies revealed that the polymorphisms of Mucin 1 gene have a role and significance as a susceptible factor contributing to gastric cancer. To better understand the roles of two MUC1 genotype polymorphisms of rs4072037 and rs2070803 in the development of gastric cancer in Vietnamese population, a multicenter, large-sample, case-control study was conducted to investigate the potential association of these single-nucleotide polymorphisms (SNPs) of MUC1 gene with gastric cancer risk and to evaluate the combination factors in relation with these SNPs. Methods This case-control study included 302 gastric cancer patients and 304 controls at four national medical hospitals between 2016 and 2018. All participants were interviewed for sociodemographic characteristics, smoking and drinking status, and personal and family history of gastric diseases. Genotyping was done using polymerase chain reaction-restriction fragment length polymorphism analysis. The association of SNPs with gastric cancer was explored using logistic regression models. Results AA genotype for rs4072037 was significantly associated with increased gastric cancer. Those with AA genotype had higher gastric cancer risk than had patients with AG (OR: 2.09, 95% CI: 1.48-2.96) and a combination of AG+GG (OR: 1.85, 95% CI: 1.33-2.56). In rs2070803, GG genotype increased gastric cancer risk when compared with AG (OR: 1.97, 95% CI: 1.39-2.80) and AG+AA (OR: 1.71, 95% CI: 1.23-2.39). AG genotypes in both SNPs decreased gastric cancer risk when compared with homogenous genotype, more specifically AA (OR: 0.51, 95% CI: 0.35-0.72) and GG (OR: 0.58, 95% CI: 0.35-0.97). These genotypes in combination with above-60-year-old age, male gender, alcoholism, and personal history of gastric disease were also significantly elevated risk factors for gastric cancer. Conclusions rs4072037 and rs2070803 of Mucin 1 genes are two genotypic risk factors for gastric cancer. Those in combination with gender, family history, smoking, and drinking habits significantly increase the risk of gastric cancer.
Collapse
Affiliation(s)
- Ngoc-Lan Thi Nguyen
- Biochemistry Department, Hanoi Medical University, Hanoi, Vietnam.,Clinical Laboratory, Hanoi Medical University Hospital, Hanoi Medical University, Hanoi, Vietnam
| | - Ngoc-Dzung Thi Dang
- Biochemistry Department, Hanoi Medical University, Hanoi, Vietnam.,Clinical Laboratory, Hanoi Medical University Hospital, Hanoi Medical University, Hanoi, Vietnam
| | - Quang-Huy Dang
- Department of Medical Laboratory Science, Faculty of Medical Technology, Hanoi Medical University, Hanoi, Vietnam
| | - Van-Chuc Tran
- Biochemistry Department, Hanoi Medical University, Hanoi, Vietnam
| | - Hoang-Long Vo
- Department of Scientific Research and International Cooperation, Hanoi Medical University Hospital, Hanoi Medical University, Hanoi, Vietnam
| | - Masamitsu Yamaguchi
- Department of Applied Biology, Advanced Insect Research Promotion Center, Kyoto Institute of Technology, Kyoto, Japan
| | - Thanh-Van Ta
- Biochemistry Department, Hanoi Medical University, Hanoi, Vietnam.,Clinical Laboratory, Hanoi Medical University Hospital, Hanoi Medical University, Hanoi, Vietnam
| |
Collapse
|
6
|
Filippini T, Malavolti M, Borrelli F, Izzo AA, Fairweather-Tait SJ, Horneber M, Vinceti M. Green tea (Camellia sinensis) for the prevention of cancer. Cochrane Database Syst Rev 2020; 3:CD005004. [PMID: 32118296 PMCID: PMC7059963 DOI: 10.1002/14651858.cd005004.pub3] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
BACKGROUND This review is an update of a previously published review in the Cochrane Database of Systematic Reviews (2009, Issue 3).Tea is one of the most commonly consumed beverages worldwide. Teas from the plant Camellia sinensis can be grouped into green, black and oolong tea, and drinking habits vary cross-culturally. C sinensis contains polyphenols, one subgroup being catechins. Catechins are powerful antioxidants, and laboratory studies have suggested that these compounds may inhibit cancer cell proliferation. Some experimental and nonexperimental epidemiological studies have suggested that green tea may have cancer-preventative effects. OBJECTIVES To assess possible associations between green tea consumption and the risk of cancer incidence and mortality as primary outcomes, and safety data and quality of life as secondary outcomes. SEARCH METHODS We searched eligible studies up to January 2019 in CENTRAL, MEDLINE, Embase, ClinicalTrials.gov, and reference lists of previous reviews and included studies. SELECTION CRITERIA We included all epidemiological studies, experimental (i.e. randomised controlled trials (RCTs)) and nonexperimental (non-randomised studies, i.e. observational studies with both cohort and case-control design) that investigated the association of green tea consumption with cancer risk or quality of life, or both. DATA COLLECTION AND ANALYSIS Two or more review authors independently applied the study criteria, extracted data and assessed methodological quality of studies. We summarised the results according to diagnosis of cancer type. MAIN RESULTS In this review update, we included in total 142 completed studies (11 experimental and 131 nonexperimental) and two ongoing studies. This is an additional 10 experimental and 85 nonexperimental studies from those included in the previous version of the review. Eleven experimental studies allocated a total of 1795 participants to either green tea extract or placebo, all demonstrating an overall high methodological quality based on 'Risk of bias' assessment. For incident prostate cancer, the summary risk ratio (RR) in the green tea-supplemented participants was 0.50 (95% confidence interval (CI) 0.18 to 1.36), based on three studies and involving 201 participants (low-certainty evidence). The summary RR for gynaecological cancer was 1.50 (95% CI 0.41 to 5.48; 2 studies, 1157 participants; low-certainty evidence). No evidence of effect of non-melanoma skin cancer emerged (summary RR 1.00, 95% CI 0.06 to 15.92; 1 study, 1075 participants; low-certainty evidence). In addition, adverse effects of green tea extract intake were reported, including gastrointestinal disorders, elevation of liver enzymes, and, more rarely, insomnia, raised blood pressure and skin/subcutaneous reactions. Consumption of green tea extracts induced a slight improvement in quality of life, compared with placebo, based on three experimental studies. In nonexperimental studies, we included over 1,100,000 participants from 46 cohort studies and 85 case-control studies, which were on average of intermediate to high methodological quality based on Newcastle-Ottawa Scale 'Risk of bias' assessment. When comparing the highest intake of green tea with the lowest, we found a lower overall cancer incidence (summary RR 0.83, 95% CI 0.65 to 1.07), based on three studies, involving 52,479 participants (low-certainty evidence). Conversely, we found no association between green tea consumption and cancer-related mortality (summary RR 0.99, 95% CI 0.91 to 1.07), based on eight studies and 504,366 participants (low-certainty evidence). For most of the site-specific cancers we observed a decreased RR in the highest category of green tea consumption compared with the lowest one. After stratifying the analysis according to study design, we found strongly conflicting results for some cancer sites: oesophageal, prostate and urinary tract cancer, and leukaemia showed an increased RR in cohort studies and a decreased RR or no difference in case-control studies. AUTHORS' CONCLUSIONS Overall, findings from experimental and nonexperimental epidemiological studies yielded inconsistent results, thus providing limited evidence for the beneficial effect of green tea consumption on the overall risk of cancer or on specific cancer sites. Some evidence of a beneficial effect of green tea at some cancer sites emerged from the RCTs and from case-control studies, but their methodological limitations, such as the low number and size of the studies, and the inconsistencies with the results of cohort studies, limit the interpretability of the RR estimates. The studies also indicated the occurrence of several side effects associated with high intakes of green tea. In addition, the majority of included studies were carried out in Asian populations characterised by a high intake of green tea, thus limiting the generalisability of the findings to other populations. Well conducted and adequately powered RCTs would be needed to draw conclusions on the possible beneficial effects of green tea consumption on cancer risk.
Collapse
Affiliation(s)
- Tommaso Filippini
- University of Modena and Reggio Emilia, Research Center in Environmental, Nutritional and Genetic Epidemiology (CREAGEN), Department of Biomedical, Metabolic and Neural Sciences, Via Campi 287, Modena, Italy, 41125
| | - Marcella Malavolti
- University of Modena and Reggio Emilia, Research Center in Environmental, Nutritional and Genetic Epidemiology (CREAGEN), Department of Biomedical, Metabolic and Neural Sciences, Via Campi 287, Modena, Italy, 41125
| | - Francesca Borrelli
- University of Naples 'Federico II', Department of Pharmacy, School of Medicine and Surgery, Via D Montesano 49, Naples, Italy, 80131
| | - Angelo A Izzo
- University of Naples 'Federico II', Department of Pharmacy, School of Medicine and Surgery, Via D Montesano 49, Naples, Italy, 80131
| | | | - Markus Horneber
- Paracelsus Medical University, Klinikum Nuremberg, Department of Internal Medicine, Division of Oncology and Hematology, Prof.-Ernst-Nathan-Str. 1, Nuremberg, Germany, D-90419
| | - Marco Vinceti
- University of Modena and Reggio Emilia, Research Center in Environmental, Nutritional and Genetic Epidemiology (CREAGEN), Department of Biomedical, Metabolic and Neural Sciences, Via Campi 287, Modena, Italy, 41125
- Boston University School of Public Health, Department of Epidemiology, 715 Albany Street, Boston, USA, MA 02118
| |
Collapse
|
7
|
Trujillo Rivera A, Sampieri CL, Morales Romero J, Montero H, Acosta Mesa HG, Cruz Ramírez N, Novoa Del Toro EM, León Córdoba K. Risk factors associated with gastric cancer in Mexico: education, breakfast and chili. REVISTA ESPANOLA DE ENFERMEDADES DIGESTIVAS 2018; 110:372-379. [PMID: 29843516 DOI: 10.17235/reed.2018.5042/2017] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
BACKGROUND AND AIM the aim of the study was to use a validated questionnaire to identify factors associated with the development of gastric cancer (GC) in the Mexican population. METHODS the study included cases and controls that were paired by sex and ± 10 years of age at diagnosis. In relation to cases, 46 patients with a confirmed histopathological diagnosis of adenocarcinoma-type GC, as reported in the hospital records, were selected, and 46 blood bank donors from the same hospital were included as controls. The previously validated Questionnaire to Find Factors Associated with Gastric Cancer (QUFA-GC©) was used to collect data. Odds ratio (OR) and 95% confidence interval (IC) were estimated via univariate analysis (paired OR). Multivariate analysis was performed by logistic regression. A decision tree was constructed using the J48 algorithm. RESULTS an association was found by univariate analysis between GC risk and a lack of formal education, having smoked for ≥ 10 years, eating rapidly, consuming very hot food and drinks, a non-suitable breakfast within two hours of waking, pickled food and capsaicin. In contrast, a protective association against GC was found with taking recreational exercise and consuming fresh fruit and vegetables. No association was found between the development of GC and having an income that reflected poverty, using a refrigerator, perception of the omission of breakfast and time period of alcoholism. In the final multivariate analysis model, having no formal education (OR = 17.47, 95% CI = 5.17-76.69), consuming a non-suitable breakfast within two hours of waking (OR = 8.99, 95% CI = 2.85-35.50) and the consumption of capsaicin ˃ 29.9 mg capsaicin per day (OR = 3.77, 95% CI = 1.21-13.11) were factors associated with GC. CONCLUSIONS an association was found by multivariate analysis between the presence of GC and education, type of breakfast and the consumption of capsaicin. These variables are susceptible to intervention and can be identified via the QUFA-GC
Collapse
Affiliation(s)
| | - Clara Luz Sampieri
- Cáncer gástrico, Instituto de Salud Pública de la Universidad Veracruzana, México
| | | | - Hilda Montero
- Instituto de Salud Pública de la Universidad Veracruzana, México
| | | | - Nicandro Cruz Ramírez
- 2Centro de Investigación en Inteligencia Artificial. Universidad Veracruzana, México
| | | | - Kenneth León Córdoba
- Gastroenterología, Centro Estatal de Cancerología "Dr. Miguel Dorantes Mesa", México
| |
Collapse
|
8
|
Xia Y, Yan Z, Wan Y, Wei S, Bi Y, Zhao J, Liu J, Liao DJ, Huang H. Knockdown of long noncoding RNA GHET1 inhibits cell‑cycle progression and invasion of gastric cancer cells. Mol Med Rep 2018; 18:3375-3381. [PMID: 30066922 PMCID: PMC6102745 DOI: 10.3892/mmr.2018.9332] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2018] [Accepted: 07/19/2018] [Indexed: 12/17/2022] Open
Abstract
GHET1 is an oncogenic long noncoding RNA (lncRNA) that promotes the proliferation and invasion of many malignant cell types. However, the function and underlying mechanisms of lncRNA GHET1 in gastric cancer are not fully understood. In this study, the expression of GHET1 was investigated in gastric cancer and it was determined whether GHET1 may potentially be used as a biomarker for the disease. The gastric cancer cell lines MGC‑803 and AGS were transfected with GHET1‑directed small interfering RNA (siRNA) and the changes in phenotype and cell‑cycle‑related molecules were assessed. The downregulation of GHET1 induced G0/G1‑phase arrest in gastric cancer cells and inhibited their proliferation, migration, and invasion. DNA synthesis and the expression of proliferating cell nuclear antigen (PCNA) decreased, which was consistent with the results of the CCK‑8 assay. The levels of specific cell‑cycle regulators were determined and the expression and activities of positive cell‑cycle regulators (cyclin D, CDK4, CDK6, cyclin E, CDK2) were reduced, whereas those of a negative regulator (P21) were increased in GHET1‑knockdown cells. Taken together, the present findings show that the downregulation of GHET1 not only inhibits the migration and invasion of gastric cancer cells, but also inhibits their proliferation, at least in part by upregulating P21 expression and downregulating cyclin and CDK expression to inhibit the G0/G1 to S phase transition. The present findings may provide a potential therapeutic target for gastric cancer.
Collapse
Affiliation(s)
- Ying Xia
- Department of Clinical Biochemistry, School of Clinical Laboratory Science, Guizhou Medical University, Guiyang, Guizhou 550004, P.R. China
| | - Zhiqiang Yan
- Department of Gastrointestinal Surgery, Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou 550004, P.R. China
| | - Ying Wan
- Department of Clinical Biochemistry, School of Clinical Laboratory Science, Guizhou Medical University, Guiyang, Guizhou 550004, P.R. China
| | - Sixi Wei
- Department of Clinical Biochemistry, School of Clinical Laboratory Science, Guizhou Medical University, Guiyang, Guizhou 550004, P.R. China
| | - Ying Bi
- Department of Clinical Biochemistry, School of Clinical Laboratory Science, Guizhou Medical University, Guiyang, Guizhou 550004, P.R. China
| | - Juanjuan Zhao
- Department of Clinical Biochemistry, School of Clinical Laboratory Science, Guizhou Medical University, Guiyang, Guizhou 550004, P.R. China
| | - Juanjuan Liu
- Department of Clinical Biochemistry, School of Clinical Laboratory Science, Guizhou Medical University, Guiyang, Guizhou 550004, P.R. China
| | - Dezhong Joshua Liao
- Department of Pathology, Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou 550004, P.R. China
| | - Hai Huang
- Department of Clinical Biochemistry, School of Clinical Laboratory Science, Guizhou Medical University, Guiyang, Guizhou 550004, P.R. China
| |
Collapse
|
9
|
Ferro A, Morais S, Rota M, Pelucchi C, Bertuccio P, Bonzi R, Galeone C, Zhang ZF, Matsuo K, Ito H, Hu J, Johnson KC, Yu GP, Palli D, Ferraroni M, Muscat J, Malekzadeh R, Ye W, Song H, Zaridze D, Maximovitch D, Fernández de Larrea N, Kogevinas M, Vioque J, Navarrete-Muñoz EM, Pakseresht M, Pourfarzi F, Wolk A, Orsini N, Bellavia A, Håkansson N, Mu L, Pastorino R, Kurtz RC, Derakhshan MH, Lagiou A, Lagiou P, Boffetta P, Boccia S, Negri E, La Vecchia C, Peleteiro B, Lunet N. Alcohol intake and gastric cancer: Meta-analyses of published data versus individual participant data pooled analyses (StoP Project). Cancer Epidemiol 2018; 54:125-132. [PMID: 29727805 DOI: 10.1016/j.canep.2018.04.009] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2017] [Revised: 04/16/2018] [Accepted: 04/17/2018] [Indexed: 12/22/2022]
Abstract
BACKGROUND Individual participant data pooled analyses allow access to non-published data and statistical reanalyses based on more homogeneous criteria than meta-analyses based on systematic reviews. We quantified the impact of publication-related biases and heterogeneity in data analysis and presentation in summary estimates of the association between alcohol drinking and gastric cancer. METHODS We compared estimates obtained from conventional meta-analyses, using only data available in published reports from studies that take part in the Stomach Cancer Pooling (StoP) Project, with individual participant data pooled analyses including the same studies. RESULTS A total of 22 studies from the StoP Project assessed the relation between alcohol intake and gastric cancer, 19 had specific data for levels of consumption and 18 according to cancer location; published reports addressing these associations were available from 18, 5 and 5 studies, respectively. The summary odds ratios [OR, (95%CI)] estimate obtained with published data for drinkers vs. non-drinkers was 10% higher than the one obtained with individual StoP data [18 vs. 22 studies: 1.21 (1.07-1.36) vs. 1.10 (0.99-1.23)] and more heterogeneous (I2: 63.6% vs 54.4%). In general, published data yielded less precise summary estimates (standard errors up to 2.6 times higher). Funnel plot analysis suggested publication bias. CONCLUSION Meta-analyses of the association between alcohol drinking and gastric cancer tended to overestimate the magnitude of the effects, possibly due to publication bias. Additionally, individual participant data pooled analyses yielded more precise estimates for different levels of exposure or cancer subtypes.
Collapse
Affiliation(s)
- Ana Ferro
- EPIUnit - Instituto de Saúde Pública, Universidade do Porto, Rua das Taipas, nº 135, 4050-600, Porto, Portugal
| | - Samantha Morais
- EPIUnit - Instituto de Saúde Pública, Universidade do Porto, Rua das Taipas, nº 135, 4050-600, Porto, Portugal
| | - Matteo Rota
- Department of Biomedical and Clinical Sciences, University of Milan, Milan, Italy; Department of Clinical Sciences and Community Health (DISCCO), University of Milan, Milan, Italy
| | - Claudio Pelucchi
- Department of Clinical Sciences and Community Health (DISCCO), University of Milan, Milan, Italy
| | - Paola Bertuccio
- Department of Clinical Sciences and Community Health (DISCCO), University of Milan, Milan, Italy
| | - Rossella Bonzi
- Department of Clinical Sciences and Community Health (DISCCO), University of Milan, Milan, Italy
| | - Carlotta Galeone
- Department of Clinical Sciences and Community Health (DISCCO), University of Milan, Milan, Italy
| | - Zuo-Feng Zhang
- Department of Epidemiology, UCLA Fielding School of Public Health and Jonsson Comprehensive Cancer Center, Los Angeles, CA, USA
| | - Keitaro Matsuo
- Division of Molecular Medicine, Aichi Cancer Center Research Institute, Nagoya, Japan
| | - Hidemi Ito
- Division of Molecular Medicine, Aichi Cancer Center Research Institute, Nagoya, Japan
| | - Jinfu Hu
- Department of Epidemiology, Harbin Medical University, Harbin, China
| | - Kenneth C Johnson
- School of Epidemiology, Public Health and Preventive Medicine, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Guo-Pei Yu
- Medical Informatics Center, Peking University, Peking, China
| | - Domenico Palli
- Molecular and Nutritional Epidemiology Unit, Cancer Research and Prevention Institute - Istituto per lo Studio e la Prevenzione Oncologica (ISPO), Florence, Italy
| | - Monica Ferraroni
- Department of Clinical Sciences and Community Health (DISCCO), University of Milan, Milan, Italy
| | - Joshua Muscat
- Department of Public Health Sciences, The Pennsylvania State University College of Medicine, Penn State Hershey Medical Center, Hershey, PA, USA
| | - Reza Malekzadeh
- Digestive Oncology Research Center, Digestive Disease Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Weimin Ye
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Huan Song
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden; Center of Public Health Sciences, Faculty of Medicine, University of Iceland, Reykjavík, Iceland
| | - David Zaridze
- Department of Epidemiology and Prevention, Russian N.N. Blokhin Cancer Research Center, Moscow, Russia
| | - Dmitry Maximovitch
- Department of Epidemiology and Prevention, Russian N.N. Blokhin Cancer Research Center, Moscow, Russia
| | - Nerea Fernández de Larrea
- Environmental and Cancer Epidemiology Unit, National Center of Epidemiology, Instituto de Salud Carlos III, Madrid, Spain; CIBER Epidemiología y Salud Pública (CIBERESP), Madrid, Spain
| | - Manolis Kogevinas
- CIBER Epidemiología y Salud Pública (CIBERESP), Madrid, Spain; ISGlobal, Centre for Research in Environmental Epidemiology (CREAL), Barcelona, Spain; IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain; Universitat Pompeu Fabra (UPF), Barcelona, Spain
| | - Jesus Vioque
- Miguel Hernandez University and ISABIAL-FISABIO Foundation, Campus San Juan, Alicante, Spain
| | - Eva M Navarrete-Muñoz
- Miguel Hernandez University and ISABIAL-FISABIO Foundation, Campus San Juan, Alicante, Spain
| | - 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
| | - Farhad Pourfarzi
- Digestive Oncology Research Center, Digestive Disease Research Institute, Tehran University of Medical Sciences, Tehran, Iran; Digestive Diseases Research Center, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Alicja Wolk
- Unit of Nutritional Epidemiology, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden; Department of Surgical Sciences, Uppsala University, Uppsala Sweden
| | - Nicola Orsini
- Unit of Nutritional Epidemiology, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Andrea Bellavia
- Unit of Nutritional Epidemiology, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Niclas Håkansson
- Unit of Nutritional Epidemiology, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Lina Mu
- Department of Epidemiology and Environmental Health, School of Public Health and Health Professions, University at Buffalo, Buffalo, NY, USA
| | - Roberta Pastorino
- Section of Hygiene - Institute of Public Health; Università Cattolica del Sacro Cuore, IRCCS Fondazione Policlinico "Agostino Gemelli", L.go F. Vito, 1 - 00168, Rome, Italy
| | - Robert C Kurtz
- Department of Medicine, Memorial Sloan Kettering Cancer Centre, New York, NY, USA
| | - Mohammad H Derakhshan
- Institute of Cardiovascular & Medical Sciences, University of Glasgow, Glasgow, United Kingdom; Digestive Oncology Research Center, Digestive Disease Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Areti Lagiou
- Department of Public Health and Community Health, School of Health Professions, Athens Technological Educational Institute, 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
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Stefania Boccia
- Section of Hygiene - Institute of Public Health; Università Cattolica del Sacro Cuore, IRCCS Fondazione Policlinico "Agostino Gemelli", L.go F. Vito, 1 - 00168, Rome, Italy
| | - Eva Negri
- Department of Biomedical and Clinical Sciences, University of Milan, Milan, Italy
| | - Carlo La Vecchia
- Department of Clinical Sciences and Community Health (DISCCO), University of Milan, Milan, Italy
| | - Bárbara Peleteiro
- EPIUnit - Instituto de Saúde Pública, Universidade do Porto, Rua das Taipas, nº 135, 4050-600, Porto, Portugal; Departamento de Ciências da Saúde Pública e Forenses e Educação Médica, Faculdade de Medicina, Universidade do Porto, Al. Prof. Hernâni Monteiro, 4200-319, Porto, Portugal
| | - Nuno Lunet
- EPIUnit - Instituto de Saúde Pública, Universidade do Porto, Rua das Taipas, nº 135, 4050-600, Porto, Portugal; Departamento de Ciências da Saúde Pública e Forenses e Educação Médica, Faculdade de Medicina, Universidade do Porto, Al. Prof. Hernâni Monteiro, 4200-319, Porto, Portugal.
| |
Collapse
|
10
|
Xu G, Feng F, Liu S, Wang F, Zheng G, Wang Q, Cai L, Guo M, Lian X, Zhang H. Clinicopathological features and prognosis in elderly gastric cancer patients: a retrospective cohort study. Onco Targets Ther 2018; 11:1353-1362. [PMID: 29559793 PMCID: PMC5856060 DOI: 10.2147/ott.s152471] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Background Little is known about the clinicopathological features and prognosis in elderly gastric cancer (GC) patients aged 65–79 years. The aim of this study was to evaluate clinicopathological features and prognosis in elderly GC patients. Patients and methods From May 2008 to December 2014, a total of 5,282 GC patients were enrolled in our present study. Patients were divided into elderly and middle-aged groups. The clinicopathological features and clinical outcomes were analyzed. Results The proportion of dysphagia was significantly higher in elderly patients than that in middle-aged patients (P=0.002), whereas the proportion of abdominal pain and heartburn was significantly lower in elderly patients than that in middle-aged patients (P<0.001 vs P=0.038, respectively). The proportion of patients with carbohydrate antigen (CA) 19-9 was significantly higher in elderly patients than that in middle-aged patients (P=0.009). There was no significant difference in clinicopathological features between elderly and middle-aged patients with D2 gastrectomy (all P>0.05). Age, tumor size, histological type, tumor depth, lymph node metastasis, carcinoembryonic antigen, alpha fetoprotein, CA19-9, and CA125 were independent risk factors for the prognosis of GC patients in univariate and multivariate analyses. Overall survival in elderly patients was significantly reduced compared with middle-aged patients (P=0.001), especially in patients with tumor size >5 cm (P=0.002), poorly differentiated tumor (P<0.000), stage III tumor (P=0.002), or normal levels of carcinoembryonic antigen (P=0.009), alpha fetoprotein (P=0.002), CA19-9 (P=0.002), and CA125 (P=0.004). Conclusion The clinicopathological features of elderly patients were different to those of middle-aged patients. The prognosis for elderly GC patients was significantly worse than for middle-aged patients.
Collapse
Affiliation(s)
- Guanghui Xu
- Division of Digestive Surgery, Xijing Hospital of Digestive Diseases, The Fourth Military Medical University, Xi'an, Shaanxi
| | - Fan Feng
- Division of Digestive Surgery, Xijing Hospital of Digestive Diseases, The Fourth Military Medical University, Xi'an, Shaanxi
| | - Shushang Liu
- Division of Digestive Surgery, Xijing Hospital of Digestive Diseases, The Fourth Military Medical University, Xi'an, Shaanxi
| | - Fei Wang
- Division of Digestive Surgery, Xijing Hospital of Digestive Diseases, The Fourth Military Medical University, Xi'an, Shaanxi.,Department of General Surgery, No 534 Hospital of PLA, Luoyang
| | - Gaozan Zheng
- Division of Digestive Surgery, Xijing Hospital of Digestive Diseases, The Fourth Military Medical University, Xi'an, Shaanxi
| | - Qiao Wang
- Division of Digestive Surgery, Xijing Hospital of Digestive Diseases, The Fourth Military Medical University, Xi'an, Shaanxi.,Department of Surgery, No 91 Center Hospital of PLA, Jiaozuo, Henan, China
| | - Lei Cai
- Division of Digestive Surgery, Xijing Hospital of Digestive Diseases, The Fourth Military Medical University, Xi'an, Shaanxi
| | - Man Guo
- Division of Digestive Surgery, Xijing Hospital of Digestive Diseases, The Fourth Military Medical University, Xi'an, Shaanxi
| | - Xiao Lian
- Division of Digestive Surgery, Xijing Hospital of Digestive Diseases, The Fourth Military Medical University, Xi'an, Shaanxi
| | - Hongwei Zhang
- Division of Digestive Surgery, Xijing Hospital of Digestive Diseases, The Fourth Military Medical University, Xi'an, Shaanxi
| |
Collapse
|
11
|
Wang PL, Xiao FT, Gong BC, Liu FN. Alcohol drinking and gastric cancer risk: a meta-analysis of observational studies. Oncotarget 2017; 8:99013-99023. [PMID: 29228746 PMCID: PMC5716786 DOI: 10.18632/oncotarget.20918] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Accepted: 08/28/2017] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Many studies investigated the association between alcohol drinking and gastric cancer risk, but the results were controversial. We performed a meta-analysis of observational studies to explore the association. MATERIALS AND METHODS We searched PubMed to identify the relevant studies that reported the association between alcohol drinking and gastric cancer risk up to December 31, 2016. We pooled relative risks (RRs) in random effects model and performed dose-response analysis to quantify the association. Cochran Q test and I2 analyses were used to evaluate the heterogeneity. Meta-regression, subgroup, sensitivity and publication bias analyses were also performed. RESULTS 75 studies were included in our study. The pooled RR of high vs low total alcohol drinking was 1.25 (95% CI, 1.15-1.37, P < 0.001), and a nonlinear association was further observed. Subgroup analysis showed that alcohol drinking significantly associated with the risk of gastric noncardia cancer (RR, 1.19; 95% CI, 1.01-1.40, P = 0.033), but not with the risk of gastric cardia cancer (RR, 1.16; 95% CI, 0.98-1.39, P = 0.087). Notably, the pooled RRs of high vs low analyses were 1.13 (95% CI, 1.03-1.24, P = 0.012) for beer drinking, 1.22 (95% CI, 1.06-1.40, P = 0.005) for liquor drinking, and 0.99 (95% CI, 0.84-1.16, P = 0.857) for wine drinking. CONCLUSIONS Our meta-analysis found a nonlinear association between alcohol drinking and gastric cancer risk, and heavy drinking level was strongly related to gastric cancer risk. Beer and liquor had significant positive associations with gastric cancer risk, while wine drinking would not increase gastric cancer risk. These results need to be verified in future research.
Collapse
Affiliation(s)
- Peng-Liang Wang
- Department of Surgical Oncology, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Fang-Tao Xiao
- Department of Surgical Oncology, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Bao-Cheng Gong
- Department of Surgical Oncology, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Fu-Nan Liu
- Department of Surgical Oncology, The First Affiliated Hospital of China Medical University, Shenyang, China
| |
Collapse
|
12
|
Na HK, Lee JY. Molecular Basis of Alcohol-Related Gastric and Colon Cancer. Int J Mol Sci 2017; 18:E1116. [PMID: 28538665 PMCID: PMC5485940 DOI: 10.3390/ijms18061116] [Citation(s) in RCA: 100] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2017] [Revised: 05/04/2017] [Accepted: 05/06/2017] [Indexed: 02/06/2023] Open
Abstract
Many meta-analysis, large cohort studies, and experimental studies suggest that chronic alcohol consumption increases the risk of gastric and colon cancer. Ethanol is metabolized by alcohol dehydrogenases (ADH), catalase or cytochrome P450 2E1 (CYP2E1) to acetaldehyde, which is then further oxidized to acetate by aldehyde dehydrogenase (ALDH). Acetaldehyde has been classified by the International Agency for Research on Cancer (IARC) as a Group 1 carcinogen to humans. The acetaldehyde level in the stomach and colon is locally influenced by gastric colonization by Helicobacter pylori or colonic microbes, as well as polymorphisms in the genes encoding tissue alcohol metabolizing enzymes, especially ALDH2. Alcohol stimulates the uptake of carcinogens and their metabolism and also changes the composition of enteric microbes in a way to enhance the aldehyde level. Alcohol also undergoes chemical coupling to membrane phospholipids and disrupts organization of tight junctions, leading to nuclear translocation of β-catenin and ZONAB, which may contributes to regulation of genes involved in proliferation, invasion and metastasis. Alcohol also generates reactive oxygen species (ROS) by suppressing the expression of antioxidant and cytoprotective enzymes and inducing expression of CYP2E1 which contribute to the metabolic activation of chemical carcinogens. Besides exerting genotoxic effects by directly damaging DNA, ROS can activates signaling molecules involved in inflammation, metastasis and angiogenesis. In addition, alcohol consumption induces folate deficiency, which may result in aberrant DNA methylation profiles, thereby influencing cancer-related gene expression.
Collapse
Affiliation(s)
- Hye-Kyung Na
- Department of Food Science and Biotechnology, College of Knowledge-Based Services Engineering, Sungshin Women's University, Seoul 01133, Korea.
| | - Ja Young Lee
- Department of Food Science and Biotechnology, College of Knowledge-Based Services Engineering, Sungshin Women's University, Seoul 01133, Korea.
| |
Collapse
|
13
|
Huang Q, Sun Q, Fan XS, Zhou D, Zou XP. Recent advances in proximal gastric carcinoma. J Dig Dis 2016; 17:421-32. [PMID: 27129018 DOI: 10.1111/1751-2980.12355] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2015] [Revised: 04/19/2016] [Accepted: 04/24/2016] [Indexed: 12/11/2022]
Abstract
The American Joint Committee on Cancer (AJCC) staging scheme requires staging proximal gastric carcinoma (PGC) as esophageal adenocarcinoma (EAC), which has been shown to be controversial by recent research results. To update the current research findings on PGC, we systematically reviewed and analyzed the scientific evidence on key arguments related to PGC. The data of high-quality research articles showed that PGC arised in the cardiac mucosa in the proximal stomach within 3 cm below the gastroesophageal junction. Its incidence is rising in East Asian countries, but decreasing in the West, and plateaued at a low level in the United States. PGC is a slowly progressive cancer with unknown independent risk factors and the mechanisms of pathogenesis. This carcinoma exhibits a wide histopathological spectrum and heterogeneous post-resection patient survival characteristics, and cannot be adequately staged for prognotic stratification by the current AJCC staging classification. The results on PGC genomics reveal unique genetic profiles, especially in East Asian populations. In conclusion, mounting evidence defies a simple placement of PGC in a single category of EAC for disease classification; further investigations on the mechanisms of PGC pathogenesis are urgently needed.
Collapse
Affiliation(s)
- Qin Huang
- Department of Pathology, Nanjing Drum Tower Hospital, Nanjing, Jiangsu Province, China. .,Department of Pathology and Laboratory Medicine, Veterans Affairs Boston Healthcare System and Harvard Medical School, West Roxbury, MA, USA.
| | - Qi Sun
- Department of Pathology, Nanjing Drum Tower Hospital, Nanjing, Jiangsu Province, China
| | - Xiang Shan Fan
- Department of Pathology, Nanjing Drum Tower Hospital, Nanjing, Jiangsu Province, China
| | - Dan Zhou
- Department of Pathology and Laboratory Medicine, Veterans Affairs Boston Healthcare System and Harvard Medical School, West Roxbury, MA, USA
| | - Xiao Ping Zou
- Department of Gastroenterology, Nanjing Drum Tower Hospital, Nanjing, Jiangsu Province, China
| |
Collapse
|
14
|
Aguiar Junior PN, Ribas C, Forones NM. Individualized Chemotherapy for Metastatic Gastric Cancer: Retrospective Data from a University Hospital in Brazil. Asian Pac J Cancer Prev 2016. [PMID: 26225668 DOI: 10.7314/apjcp.2015.16.13.5289] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Despite the decreased incidence, gastric cancer is still a frequent cause of cancer related death. The 1st line 2 or 3 drugs regimen is still a debatable issue. HER2 targeted therapy has emerged as the standard of care, but it is unavailable in the Brazilian Public Health System. The end-point of this trial was overall survival (OS) in patients with metastatic gastric cancer treated in a public university hospital in Brazil. The secondary end-points were efficacy and safety of regimens with 2 (F+P) or 3 (EOX) drugs to develop an institutional guideline to facilitate optimal treatments. MATERIALS AND METHODS In this retrospective study, 1st line regimens were evaluated for OS and PFS stratified by age and ECOG using Cox regression. RESULTS 47 patients were treated over the last 3 years. In 1st line, 29 were treated with F+P (mean 59.3 years, 34.5% ECOG 2 and a mean of 5.69 cycles) and 16 with EOX (mean 47 years, 18.8% ECOG 2 and a mean of 5.44 cycles). The median OS was 13.8 months (95%CI 10.7-16.9). Response was evaluated in 40 cases and was 64.3% for EOX and 37.5% for F+P (p=0.25). The median PFS was 9.5 months for EOX and 5.6 months for F+P (HR 0.85, 95%CI 0.41-1.74). However, among patients with ECOG 2 mPFS was 3.70 vs 5.40 months, respectively (p=0.86). Regimens showed similar manageable adverse events. A total of 34 patients suffered progression and 14 received 2nd line therapy. Diffuse histology (HR 1.89, 95%CI 1.22-2.88), achieving 2nd line (HR: 0.25, 95%CI 0.11-0.58) and treatment response (HR 0.23, 95%CI 0.12-0.47) were OS prognostic factors. CONCLUSIONS Patients treated in our hospital had outcomes compatible with the literature. The regimen choice should be related to patient features. Second line treatment should be considered.
Collapse
Affiliation(s)
- Pedro Nazareth Aguiar Junior
- Oncology Division, Department of Oncology and Hematology, Federal University of Sao Paulo - Escola Paulista de Medicina, San Paulo, Brazil E-mail :
| | | | | |
Collapse
|
15
|
Ma Y, Liu L, Yan F, Wei W, Deng J, Sun J. Enhanced expression of long non-coding RNA NEAT1 is associated with the progression of gastric adenocarcinomas. World J Surg Oncol 2016; 14:41. [PMID: 26911892 PMCID: PMC4765228 DOI: 10.1186/s12957-016-0799-3] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2015] [Accepted: 02/16/2016] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Long non-coding RNAs (lncRNAs) are emerging as new players in the cancer. The aim of this study was to examine the abnormalities of NEAT1 (nuclear paraspeckle assembly transcript 1, also known as MENε/β) in gastric adenocarcinomas (GACs). METHODS One hundred thirty-one GAC tissues and matched adjacent normal tissues (ANTs) were collected from patients who undergone surgery. Differences in of NEAT1 expression were examined via quantitative reverse transcriptase PCR (qRT-PCR). WST-1 assay and transwell assay were carried out in vitro to investigate the proliferation and migration of GAC cells with alteration in NEAT1 long non-coding RNA (lncRNA) expression. RESULTS The expression levels of lncRNA NEAT1 were significantly elevated in GAC tissues (P<0.001) compared with ANTs. There was also a statistical difference in NEAT1 expression between early and advanced GACs (P=0.0111). GACs with lymph node metastasis (LNM) expressed higher levels of NEAT1 lncRNA compared with those without LNM (P=0.004). In the in vitro experiments, the proliferation but not migration of GAC cells was attenuated after NEAT1 knockdown by RNA interference. CONCLUSIONS Expression of NEAT1 lncRNA was enhanced in GACs; and NEAT1 may influence GAC progression by promoting tumor growth.
Collapse
Affiliation(s)
- Yanling Ma
- Department of Oncology of Zhongshan Hospital, Wuhan University, No. 26 Zhongshan Road, Wuhan, Hubei, 430033, China.
| | - Li Liu
- Department of Oncology of Zhongshan Hospital, Wuhan University, No. 26 Zhongshan Road, Wuhan, Hubei, 430033, China.
| | - Fei Yan
- Department of Oncology of Zhongshan Hospital, Wuhan University, No. 26 Zhongshan Road, Wuhan, Hubei, 430033, China.
| | - Wujie Wei
- Department of Oncology of Zhongshan Hospital, Wuhan University, No. 26 Zhongshan Road, Wuhan, Hubei, 430033, China.
| | - Jie Deng
- Department of Oncology of Zhongshan Hospital, Wuhan University, No. 26 Zhongshan Road, Wuhan, Hubei, 430033, China.
| | - Jianhai Sun
- Department of Oncology of Zhongshan Hospital, Wuhan University, No. 26 Zhongshan Road, Wuhan, Hubei, 430033, China.
| |
Collapse
|
16
|
Basaran H, Koca T, Cerkesli AK, Arslan D, Karaca S. Treatment outcomes and survival study of gastric cancer patients: a retrospective analysis in an endemic region. Asian Pac J Cancer Prev 2015; 16:2055-60. [PMID: 25773850 DOI: 10.7314/apjcp.2015.16.5.2055] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
PURPOSE To present information about prognostic factors of gastric cancer patients treated in our Erzurum center including age, gender, tumour location, pathological grade, stage and the effect of treatment on survival. MATERIALS AND METHODS This retrospective study was performed on patients who applied to our clinic and diagnosed as gastric cancer. Age and gender of the patients, primary location, histopathological characteristics, TNM stage of the gastric cancers (GCs), treatment applied, oncological treatment modalities and survival outcomes were studied. A univariate analysis of potential prognostic factors was performed with the log-rank test for categorical factors and parameters with a p value < 0.05 at the univariate step were included in the multivariate regression. RESULTS A total of 228 patients with a confirmed diagnosis of gastric cancer were included in the study with a male/female ratio of 1.47. Median follow-up period was estimated as 22.3 (range, 3 to 96) months. When diagnosis of the patients at admission was analysed, stage III patients were most frequently encountered (n=147; 64.5%). One hundred and twenty-six (55.3%) underwent surgical treatment, while 117 (51.3%) were given adjuvant chemotherapy. Median overall survival time was 18.0 (± 1.19) months. Mean overall survival rates for 1, 2, 3 and 5 years were 68 ± 0.031%, 36 ± 0.033%, 24 ± 0.031% and 15.5 ± 0.036%, respectively. Univariate variables found to be significant for median OS in the multivariate analysis were evaluated with Cox regression analysis. A significant difference was found among TNM stage groups, location of the tumour and postoperative adjuvant treatment receivers (p values were 0.011, 0.025 and 0.001, respectively). CONCLUSIONS This study revealed that it is possible to achieve long-term survival of gastric cancer with early diagnosis. Besides, in locally advanced GC patients, curative resection followed by adjuvant concomitant chemoradiotherapy based on the McDonald regimen was an independent prognostic factor for survival.
Collapse
Affiliation(s)
- Hamit Basaran
- Department of Radiation Oncology, Erzurum Regional Training and Research Hospital, Erzurum, Turkey E-mail :
| | | | | | | | | |
Collapse
|
17
|
Alebouyeh F, Bidgoli SA, Ziarati P, Heshmati M, Qomi M. Mutagenicity Assessment of Drinking Water in Combination with Flavored Black Tea Bags: a Cross Sectional Study in Tehran. Asian Pac J Cancer Prev 2015; 16:7479-84. [PMID: 26625748 DOI: 10.7314/apjcp.2015.16.17.7479] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/07/2022] Open
Abstract
Diseases related to water impurities may present as major public health burdens. The present study aimed to assess the mutagenicity of drinking water from different zones of Tehran, and evaluate possible health risks through making tea with tea bags, by Ames mutagenicity test using TA 100, TA 98 and YG1029 strains. For this purpose, 450 water samples were collected over the period of July to December 2014 from 5 different zones of Tehran. Except for one sample, no mutagenic potential was detected during these two seasons and the MI scores were almost normal (≤ 1-1.6) in TA 100, TA 98 and YG1029 strains. Although no mutagenic effects were considered in TA 98 and TA 100 in the test samples of our three evaluated tea bag brands, one sample from a local company showed mutagenic effects in the YG1029 strain (MI=1.7-1.9 and 2) after prolonged (10-15 min.) steeping. Despite the mild mutagenic effect discovered for one of the brand, this cross sectional study showed relative safety of water samples and black tea bags in Tehran. According to the sensitivity of YG1029 to the mutagenic potential of water and black tea, even without metabolic activation by s9 fraction, this metabolizer strain could be considered as sensitive and applicable to food samples for quantitative analysis of mutagens.
Collapse
Affiliation(s)
- Farzaneh Alebouyeh
- Pharmaceutical Sciences Research Center, Islamic Azad University, Pharmaceutical Sciences Branch (IAUPS), Tehran, Iran E-mail :
| | | | | | | | | |
Collapse
|
18
|
Hudler P. Challenges of deciphering gastric cancer heterogeneity. World J Gastroenterol 2015; 21:10510-10527. [PMID: 26457012 PMCID: PMC4588074 DOI: 10.3748/wjg.v21.i37.10510] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2015] [Revised: 06/19/2015] [Accepted: 08/31/2015] [Indexed: 02/06/2023] Open
Abstract
Gastric cancer is in decline in most developed countries; however, it still accounts for a notable fraction of global mortality and morbidity related to cancer. High-throughput methods are rapidly changing our view and understanding of the molecular basis of gastric carcinogenesis. Today, it is widely accepted that the molecular complexity and heterogeneity, both inter- and intra-tumour, of gastric adenocarcinomas present significant obstacles in elucidating specific biomarkers for early detection of the disease. Although genome-wide sequencing and gene expression studies have revealed the intricate nature of the molecular changes that occur in tumour landscapes, the collected data and results are complex and sometimes contradictory. Several aberrant molecules have already been tested in clinical trials, although their diagnostic and prognostic utilities have not been confirmed thus far. The gold standard for the detection of sporadic gastric cancer is still the gastric endoscopy, which is considered invasive. In addition, genome-wide association studies have confirmed that genetic variations are important contributors to increased cancer risk and could participate in the initiation of malignant transformation. This hypothesis could in part explain the late onset of sporadic gastric cancers. The elaborate interplay of polymorphic low penetrance genes and lifestyle and environmental risk factors requires additional research to decipher their relative impacts on tumorigenesis. The purpose of this article is to present details of the molecular heterogeneity of sporadic gastric cancers at the DNA, RNA, and proteome levels and to discuss issues relevant to the translation of basic research data to clinically valuable tools. The focus of this work is the identification of relevant molecular changes that could be detected non-invasively.
Collapse
|
19
|
Mansoori AA, Jain SK. Molecular Links between Alcohol and Tobacco Induced DNA Damage, Gene Polymorphisms and Patho-physiological Consequences: A Systematic Review of Hepatic Carcinogenesis. Asian Pac J Cancer Prev 2015; 16:4803-12. [PMID: 26163595 DOI: 10.7314/apjcp.2015.16.12.4803] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Chronic alcohol and tobacco abuse plays a crucial role in the development of different liver associated disorders. Intake promotes the generation of reactive oxygen species within hepatic cells exposing their DNA to continuous oxidative stress which finally leads to DNA damage. However in response to such damage an entangled protective repair machinery comprising different repair proteins like ATM, ATR, H2AX, MRN complex becomes activated. Under abnormal conditions the excessive reactive oxygen species generation results in genetic predisposition of various genes (as ADH, ALDH, CYP2E1, GSTT1, GSTP1 and GSTM1) involved in xenobiotic metabolic pathways, associated with susceptibility to different liver related diseases such as fibrosis, cirrhosis and hepatocellular carcinoma. There is increasing evidence that the inflammatory process is inherently associated with many different cancer types, including hepatocellular carcinomas. The generated reactive oxygen species can also activate or repress epigenetic elements such as chromatin remodeling, non-coding RNAs (micro-RNAs), DNA (de) methylation and histone modification that affect gene expression, hence leading to various disorders. The present review provides comprehensive knowledge of different molecular mechanisms involved in gene polymorphism and their possible association with alcohol and tobacco consumption. The article also showcases the necessity of identifying novel diagnostic biomarkers for early cancer risk assessment among alcohol and tobacco users.
Collapse
Affiliation(s)
- Abdul Anvesh Mansoori
- Molecular Biology Laboratory, Department of Biotechnology, Dr. Hari Singh Gour Central University, Sagar, M.P. India E-mail :
| | | |
Collapse
|