Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 863
Filtrar
1.
Occup Med (Lond) ; 74(6): 438-448, 2024 Sep 23.
Artigo em Inglês | MEDLINE | ID: mdl-39313244

RESUMO

BACKGROUND: Diesel exhaust exposure and cancer other than the lungs have been limitedly investigated. AIMS: To conduct a systematic review and meta-analysis on the association between occupational exposure to diesel exhaust and gastrointestinal cancers. METHODS: Two researchers performed a systematic literature review to identify all cohort studies on occupational exposure to diesel exhaust and risk of cancers other than lung. Of the 30 retained studies, 10 reported risk estimates for oesophageal, 18 on gastric, 15 on colon and 14 on rectal cancer. We performed random-effects meta-analyses to calculate summary relative risks (RRs) and 95% confidence intervals (CIs) for ever-exposure to diesel exhaust. RESULTS: We calculated summary RR = 1.08 (95% CI 0.97-1.21, P heterogeneity = 0.06) for oesophageal, 1.06 (95% CI 0.99-1.14, P < 0.001) for gastric, 0.98 (95% CI 0.96-1.00, P = 0.453) for colon, and RR = 1.04 (95% CI 0.97-1.11, P = 0.013) for rectal cancer. Drivers showed an association with oesophageal (RR = 1.26, 95% CI 0.99-1.62), gastric (RR = 1.20, 95% CI 0.91-1.59) and rectal cancer (RR = 1.41, 95% CI 1.13-1.75); machine operators with oesophageal (RR = 1.09, 95% CI 1.00-1.20) and gastric (RR = 1.15, 95% CI 1.10-1.20) and handlers with oesophageal cancer (RR = 1.95, 95% CI 1.23-3.09). Studies from Europe revealed an association with gastric cancer while those from North America did not (P < 0.05). No difference was found by quality score except for gastric cancer, where high-quality studies but not low-quality ones showed increased risk (P heterogeneity = 0.04). There was no evidence of publication bias. CONCLUSIONS: An increased but insignificant risk of oesophageal, gastric and rectal, but not colon cancer, was suggested in workers exposed to diesel exhaust. Residual confounding cannot be excluded.


Assuntos
Neoplasias Gastrointestinais , Doenças Profissionais , Exposição Ocupacional , Emissões de Veículos , Humanos , Exposição Ocupacional/efeitos adversos , Neoplasias Gastrointestinais/epidemiologia , Neoplasias Gastrointestinais/induzido quimicamente , Neoplasias Gastrointestinais/etiologia , Doenças Profissionais/epidemiologia , Doenças Profissionais/induzido quimicamente , Estudos de Coortes , Neoplasias Esofágicas/epidemiologia , Neoplasias Esofágicas/induzido quimicamente , Neoplasias Esofágicas/etiologia
2.
Eur J Cancer ; 210: 114258, 2024 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-39168001

RESUMO

BACKGROUND: Food biodiversity in human diets has potential co-benefits for both public health and sustainable food systems. However, current evidence on the potential relationship between food biodiversity and cancer risk, and particularly gastrointestinal cancers typically related to diet, remains limited. This study evaluated how dietary species richness (DSR) was associated with gastrointestinal cancer risk in a pan-European population. METHODS: Associations between DSR and subsequent gastrointestinal cancer risk were examined among 450,111 adults enrolled in the European Prospective Investigation into Cancer and Nutrition cohort (EPIC, initiated in 1992), free of cancer at baseline. Usual dietary intakes were assessed at recruitment with country-specific dietary questionnaires. DSR of an individual's yearly diet was calculated based on the absolute number of unique biological species in each food and drink item. Associations between DSR and cancer risk were assessed by multivariable Cox proportional hazards regression models. FINDINGS: During a median follow-up time of 14.1 years (SD=3.9), 10,705 participants were diagnosed with gastrointestinal cancer. Hazard ratios (HRs) and 95 % confidence intervals (CIs) comparing overall gastrointestinal cancer risk in the highest versus lowest quintiles of DSR indicated inverse associations in multivariable-adjusted models [HR (95 % CI): 0.77 (0.69-0.87); P-value < 0·0001] (Table 2). Specifically, inverse associations were observed between DSR and oesophageal squamous cell carcinoma, proximal colon, colorectal, and liver cancer risk (p-trend<0.05 for all cancer types). INTERPRETATION: Greater food biodiversity in the diet may lower the risk of certain gastrointestinal cancers. Further research is needed to replicate these novel findings and to understand potential mechanisms.


Assuntos
Biodiversidade , Dieta , Neoplasias Gastrointestinais , Humanos , Neoplasias Gastrointestinais/epidemiologia , Neoplasias Gastrointestinais/etiologia , Estudos Prospectivos , Europa (Continente)/epidemiologia , Masculino , Feminino , Pessoa de Meia-Idade , Fatores de Risco , Adulto , Dieta/efeitos adversos , Dieta/estatística & dados numéricos , Idoso
3.
J Dent Res ; 103(10): 962-972, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-39185624

RESUMO

The association between periodontal diseases and the risk of gastrointestinal cancers, especially site-specific gastrointestinal cancers, remains unclear. Here, we comprehensively searched PubMed, EMBASE, Web of Science, and Google Scholar from inception to April 2024 to identify relevant studies. The pooled hazard ratios (HRs) and 95% confidence intervals (CIs) were calculated with a random-effects model. Subgroup analyses and sensitivity analyses were conducted to confirm the robustness of the main findings in different populations. This study was reported according to PRISMA 2020 guidelines. In total, we identified 19 studies, including 16.6 million participants. Individuals with periodontal diseases had an increased risk of overall gastrointestinal cancers compared with those without periodontal diseases (HR 1.31, 95% CI 1.16-1.49). Periodontal diseases significantly increased the risk of esophageal cancer by 39% (HR 1.39, 95% CI 1.15-1.68), gastric cancer by 13% (HR 1.13, 95% CI 1.01-1.26), colorectal cancer by 21% (HR 1.21, 95% CI 1.05-1.39), pancreatic cancer by 35% (HR 1.35, 95% CI 1.00-1.82), and liver cancer by 9% (HR 1.09, 95% CI 1.04-1.13). The risk of gastrointestinal cancers was significantly increased by periodontitis (HR 1.45, 95% CI 1.14-1.85), gingivitis (HR 1.03, 95% CI 1.01-1.04), and periodontitis/gingivitis (HR 1.27, 95% CI 1.07-1.51). Furthermore, severe periodontal diseases showed a significantly increased risk of gastrointestinal cancer (HR 1.79, 95% CI 1.07-2.99). Results of sensitivity analyses for site-specific gastrointestinal cancers were robust with the main findings. In summary, periodontal diseases, especially severe periodontitis, increase the risk of overall and site-specific gastrointestinal cancers. Interventions to prevent and manage periodontal diseases may reduce the risk of developing gastrointestinal cancers.


Assuntos
Neoplasias Gastrointestinais , Doenças Periodontais , Humanos , Neoplasias Gastrointestinais/epidemiologia , Neoplasias Gastrointestinais/etiologia , Neoplasias Gastrointestinais/prevenção & controle , Doenças Periodontais/complicações , Doenças Periodontais/diagnóstico , Doenças Periodontais/epidemiologia , Doenças Periodontais/terapia , Fatores de Risco
4.
Metabolism ; 160: 156014, 2024 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-39182602

RESUMO

Metabolic dysfunction-associated steatotic liver disease (MASLD) poses a significant and ever-increasing health and economic burden worldwide. Substantial epidemiological evidence shows that MASLD is a multisystem disease that is associated not only with liver-related complications but is also associated with an increased risk of developing cardiometabolic comorbidities and extrahepatic cancers (principally gastrointestinal [GI] cancers). GI cancers account for a quarter of the global cancer incidence and a third of cancer-related deaths. In this narrative review, we provide an overview of the literature on (a) the epidemiological data on the risk of non-liver GI cancers in MASLD, (b) the putative mechanisms by which MASLD (and factors linked with MASLD) may increase this risk, and (c) the possible pharmacotherapies beneficially affecting both MASLD and extrahepatic GI cancer risk. There are multiple potential pathophysiological mechanisms by which MASLD may increase extrahepatic GI cancer risk. Although further studies are needed, the current evidence supports a possible extrahepatic carcinogenic role for MASLD, regardless of obesity and diabetes status, thus highlighting the potential role of tailoring cancer screening for individuals with MASLD. Although there are conflicting data in the literature, aspirin, statins and metformin appear to exert some chemo-preventive effects against GI cancer.


Assuntos
Fígado Gorduroso , Neoplasias Gastrointestinais , Humanos , Neoplasias Gastrointestinais/etiologia , Neoplasias Gastrointestinais/epidemiologia , Neoplasias Gastrointestinais/metabolismo , Neoplasias Gastrointestinais/complicações , Neoplasias Gastrointestinais/patologia , Fígado Gorduroso/complicações , Fígado Gorduroso/epidemiologia , Fígado Gorduroso/etiologia , Doenças Metabólicas/complicações , Doenças Metabólicas/epidemiologia , Doenças Metabólicas/etiologia , Fatores de Risco
5.
Nutrients ; 16(13)2024 Jun 23.
Artigo em Inglês | MEDLINE | ID: mdl-38999742

RESUMO

BACKGROUND: Ultra-Processed Foods (UPFs) are increasingly consumed worldwide, even in regions with strong dietary traditions like the Mediterranean and can play a crucial role in the development of chronic diseases, including cancer. This population-based prospective cohort study investigates the association between UPF consumption and gastrointestinal cancers and other causes of mortality in Southern Italy. METHODS: Data were collected from 4870 participants in the MICOL and NUTRIHEP cohorts. The EPIC questionnaire was used to elicit information on food and drink consumption and UPFs were categorized by degree of processing according to the NOVA classification. Cox proportional hazards regression and competing risk models were employed for statistical analysis. RESULTS: UPF consumption was positively associated with all-cause mortality: participants in the 3rd UFP quartile, as compared to the lowest, had a 27% higher risk of death (SHR 1.27 95% CI, 1.03; 1.57), while in the highest quartile as compared to the lowest, the risk was 34% higher (SHR 1.34 95% CI, 1.00; 1.79). Higher UPFs intake was also correlated with an increased gastrointestinal cancers mortality risk, especially the 2nd (SHR 1.65, 95% CI: 1.01; 2.71) and 4th quartile (SHR 3.14 95% CI: 1.56; 6.32), with a dose-dependent effect. For the other cancers, a SHR 1.61 (95% CI 1.03; 2.54) was observed for the 3rd quartile. CONCLUSIONS: Our results reinforce the link between UPF consumption and cancer risk, emphasizing the urgent need for interventions targeting dietary patterns.


Assuntos
Fast Foods , Neoplasias Gastrointestinais , Humanos , Itália/epidemiologia , Feminino , Masculino , Pessoa de Meia-Idade , Neoplasias Gastrointestinais/mortalidade , Neoplasias Gastrointestinais/etiologia , Neoplasias Gastrointestinais/epidemiologia , Fatores de Risco , Estudos Prospectivos , Fast Foods/efeitos adversos , Fast Foods/estatística & dados numéricos , Idoso , Adulto , Manipulação de Alimentos , Modelos de Riscos Proporcionais , Dieta/efeitos adversos , Dieta/estatística & dados numéricos , Inquéritos e Questionários , Alimento Processado
6.
PLoS One ; 19(6): e0305994, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38924054

RESUMO

BACKGROUND: The association between red meat, fish, and processed meat consumption and the risk of developing gastrointestinal (GI) cancers remains inconclusive despite several investigations. Therefore, we conducted a systematic review and meta-analysis of observational studies to update the existing scientific evidence. METHODS: We searched PubMed, Web of Science, and Scopus databases until May 20, 2023. We analyzed observational studies that examined the associations between red and processed meat and fish consumption and GI cancers. We assessed between-study heterogeneity using the χ2 and τ2 tests, as well as I2 statistics. We explored the likelihood of publication bias using Begg's and Egger's tests and trim-and-fill analysis. We reported the overall effect sizes as odds ratios (ORs) with a 95% confidence interval (CI) using a random-effects model. RESULTS: Of the 21,004 studies identified, 95 studies involving 5,794,219 participants were included in the meta-analysis. The consumption of high levels of red meat, as compared to low levels, was found to significantly increase the risk of developing esophageal, pancreatic, liver, colon, rectal, and colorectal cancers. Similarly, the consumption of high levels of processed meat, as compared to low levels, significantly increased the risk of pancreatic, colon, rectal, and colorectal cancers. In contrast, the consumption of high levels of fish, as compared to low levels, significantly reduced the risk of colon, rectal, and colorectal cancers. CONCLUSIONS: This meta-analysis provides updated evidence on the association between red meat, processed meat, and fish consumption and the risk of developing five major types of GI cancers.


Assuntos
Neoplasias Gastrointestinais , Estudos Observacionais como Assunto , Carne Vermelha , Humanos , Neoplasias Gastrointestinais/etiologia , Neoplasias Gastrointestinais/epidemiologia , Carne Vermelha/efeitos adversos , Animais , Peixes , Produtos da Carne/efeitos adversos , Fatores de Risco , Carne/efeitos adversos , Alimentos Marinhos , Dieta/efeitos adversos
7.
Am J Gastroenterol ; 119(6): 1056-1065, 2024 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-38832708

RESUMO

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


Assuntos
Fast Foods , Neoplasias Gastrointestinais , Humanos , Neoplasias Gastrointestinais/epidemiologia , Neoplasias Gastrointestinais/etiologia , Fast Foods/efeitos adversos , Fatores de Risco , Neoplasias Colorretais/epidemiologia , Neoplasias Colorretais/etiologia , Neoplasias Gástricas/epidemiologia , Neoplasias Gástricas/etiologia , Alimento Processado
8.
Hematol Oncol Clin North Am ; 38(4): 783-811, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-38760197

RESUMO

Gastrointestinal cancers are a leading cause of cancer morbidity and mortality. Many gastrointestinal cancers develop from cancer precursor lesions, which are commonly found in individuals with hereditary cancer syndromes. Hereditary cancer syndromes have advanced our understanding of cancer development and progression and have facilitated the evaluation of cancer prevention and interception efforts. Common gastrointestinal hereditary cancer syndromes, including their organ-specific cancer risk and surveillance recommendations, are reviewed in this article. The management of common gastroesophageal, pancreatic, and colonic precursor lesions is also discussed, regardless of their genetic background. Further research is needed to advance chemoprevention and immunoprevention strategies.


Assuntos
Neoplasias Gastrointestinais , Humanos , Neoplasias Gastrointestinais/terapia , Neoplasias Gastrointestinais/diagnóstico , Neoplasias Gastrointestinais/genética , Neoplasias Gastrointestinais/etiologia , Lesões Pré-Cancerosas/diagnóstico , Lesões Pré-Cancerosas/genética , Lesões Pré-Cancerosas/patologia , Lesões Pré-Cancerosas/terapia , Predisposição Genética para Doença , Detecção Precoce de Câncer , Síndromes Neoplásicas Hereditárias/diagnóstico , Síndromes Neoplásicas Hereditárias/genética , Síndromes Neoplásicas Hereditárias/terapia
9.
Indian J Gastroenterol ; 43(4): 729-739, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-38767806

RESUMO

BACKGROUND: Poor oral health and oral dysbiosis were found to be associated with cancers, especially of the gastrointestinal (GI) system. But the cause-and-effect relationship and the effect of the risk are not yet known due to scarcity of literature. Understanding such risk relationship can contribute to an integrated multi-disciplinary approach for GI cancer prevention. AIM: The aim of the present systematic review and meta-analysis is to assess the role of oral dysbiosis on increasing the risk of digestive system cancers. OBJECTIVE: To evaluate the effect of poor oral health on increasing the risk of gastrointestinal cancers. METHODS: We conducted a systematic search following Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines in databases PubMed, Elsevier, Wiley's online library and Web of Science from inception to February 2023 to include recent cohort studies that assessed the association between poor oral health and the risk of cancer. We assessed bias using the New Castle Ottawa scale. We used inferential statistics to describe the effect of oral dysbiosis on gastrointestinal cancers. We performed a sub-group analysis to assess the effect of oral conditions on individual cancers. RESULTS: We included 10 longitudinal studies in the meta-analysis. The overall effect size of poor oral health and GI cancer risk was hazard's ratio (HR) =1.30 (95% CI: [1.14, 1.46]) (p<0.001) (I2 = 68.78). Sub-group analysis indicated that poor oral health increases the risk of esophageal cancer HR=1.61 (95% CI: [1.37, 1.85]), stomach cancer HR=1.33 (95% CI: [1.08, 1.58]), pancreatic cancer HR=1.90 (95% CI; [1.29, 2.50]) and colorectal and hepatocellular carcinoma HR=1.16 (95% CI: [1.08, 1.23]). CONCLUSION: The meta-analysis indicated that poor oral health was significantly associated with increasing the risk of GI cancers.


Assuntos
Disbiose , Neoplasias Gastrointestinais , Saúde Bucal , Humanos , Disbiose/complicações , Neoplasias Gastrointestinais/etiologia , Neoplasias Gastrointestinais/epidemiologia , Estudos Longitudinais , Fatores de Risco , Doenças da Boca/epidemiologia , Doenças da Boca/etiologia , Risco
10.
Int J Cancer ; 155(7): 1203-1211, 2024 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-38712628

RESUMO

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


Assuntos
Neoplasias Esofágicas , Infecções por Helicobacter , Helicobacter pylori , Neoplasias Gástricas , Humanos , Infecções por Helicobacter/complicações , Infecções por Helicobacter/epidemiologia , Pessoa de Meia-Idade , Masculino , Feminino , Helicobacter pylori/isolamento & purificação , Adulto , Neoplasias Gástricas/microbiologia , Neoplasias Gástricas/epidemiologia , Neoplasias Gástricas/etiologia , Neoplasias Gástricas/patologia , Idoso , Neoplasias Esofágicas/epidemiologia , Neoplasias Esofágicas/microbiologia , Neoplasias Esofágicas/etiologia , China/epidemiologia , Estudos de Coortes , Fatores de Risco , Prevalência , Neoplasias Gastrointestinais/microbiologia , Neoplasias Gastrointestinais/epidemiologia , Neoplasias Gastrointestinais/etiologia , Trato Gastrointestinal Superior/patologia , Trato Gastrointestinal Superior/microbiologia
11.
Cancer Med ; 13(4): e7064, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-38457256

RESUMO

INTRODUCTION: Although anti-CD19 chimeric antigen receptor (CAR) T cell therapy was approved as a very effective salvage strategy in relapsed/refractory (R/R) B cell lymphoma, the experience in R/R gastrointestinal (GI) lymphoma is still insufficient. METHODS: We summarized the efficacy and side effects of anti-CD19 CAR T-cell therapy in 12 patients with R/R GI lymphoma. Based on literature, the R/R GI lymphoma patients were divided into subgroups with different characteristics: Bulky/No bulky disease, Gastric/Gastrointestinal involvement, Gastrointestinal/Combined extra-gastrointestinal lesions, Ulcer/Lumps or nodules type, With/without gastrointestinal bleeding. RESULTS: The objective response rate (ORR) was 66.67% in these 12 patients. The ORR was 83.33% in no bulky disease group, 80.00% in gastric involvement group, 100.00% in ulcer type group, and 80.00% in no gastrointestinal bleeding group. The CR rate was 33.33% in these 12 patients. The CR was 50.0% in no bulky disease group, 60.00% in gastric involvement group, and 80.00% in ulcer type group. The PFS and OS rate of the 12 patients at 6 months after infusion were 54.55% and 58.33%, respectively. The overall survival (OS) at 6 months was higher in no bulky disease group. There was no difference of the OS or the progression free survival (PFS) at 6 months between the other groups. The mean peak of CAR-T cells and Cytokine Release Syndrome (CRS) grade were higher in gastrointestinal lesions group. The mean peak of IFN-γ and CRS grade were higher in gastrointestinal bleeding group. Four out of six patients in group of gastrointestinal lesions group were patient with high tumor burden. Patients with gastrointestinal involvement only were at higher risk for gastrointestinal bleeding. CONCLUSIONS: The ORR and CR of high tumor load, gastrointestinal involvement, lumps or nodules type and gastrointestinal bleeding group were lower. The CRS grade was higher in gastrointestinal lesions group and in gastrointestinal bleeding group. Patients with gastrointestinal involvement only were at higher risk for gastrointestinal bleeding.


Assuntos
Neoplasias Gastrointestinais , Linfoma de Células B , Linfoma , Humanos , Imunoterapia Adotiva/efeitos adversos , Receptores de Antígenos de Linfócitos T , Linfócitos T , Úlcera/etiologia , Linfoma/terapia , Linfoma de Células B/etiologia , Neoplasias Gastrointestinais/terapia , Neoplasias Gastrointestinais/etiologia , Síndrome da Liberação de Citocina/etiologia , Antígenos CD19 , Hemorragia Gastrointestinal
12.
Int Arch Occup Environ Health ; 97(3): 231-251, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38356028

RESUMO

BACKGROUND: Although silica is a proven lung carcinogen, there is no convincing evidence linking crystalline silica to gastrointestinal malignancies. METHODS: We detailedly searched studies on the link between gastrointestinal malignancies and occupational silica exposure. Studies published between 1987 and 2023 were found by searching PubMed, Scopus, Cochrane Library, and Web of Science databases. Further studies were included from reference searching. We conducted a meta-analysis of the incidence and mortality of gastrointestinal malignancies and occupational silica exposure. We computed pooled-risk estimates using random effects models. Egger's regression asymmetry test and a funnel plot were used to identify publication bias. Moreover, sensitivity analysis and subgroup analysis were out. RESULTS: We identified 40 research with individuals from 13 different countries. The results indicate that occupational silica exposure raises the risk of gastric and esophageal cancer incidence, with pooled standardized incidence ratio of 1.35 (95% CI 1.21-1.51, p < 0.001), 1.31 (95% CI 1.04-1.65, p = 0.023), respectively, but there was a lack of statistically significant relationship between standardized mortality ratio. In addition, we found that silica exposure did not increase the risk of colorectal and pancreatic cancers. Occupational silica exposure was found to increase the risk of liver cancer, with pooled SIR and SMR of 1.19 (95% CI 1.04-1.35, p = 0.009), 1.24 (95% CI 1.03-1.49, p = 0.026), respectively. CONCLUSIONS: We discovered a link between occupational silica exposure and gastrointestinal malignancies, with cancers of the liver, stomach, and esophagus being the most prevalent. Colorectal and pancreatic cancer were not linked to occupational silica exposure.


Assuntos
Neoplasias Gastrointestinais , Exposição Ocupacional , Dióxido de Silício , Humanos , Dióxido de Silício/efeitos adversos , Exposição Ocupacional/efeitos adversos , Neoplasias Gastrointestinais/epidemiologia , Neoplasias Gastrointestinais/etiologia , Doenças Profissionais/epidemiologia , Incidência , Estudos de Coortes , Neoplasias Esofágicas/epidemiologia , Fatores de Risco
13.
J Gastroenterol Hepatol ; 39(4): 630-641, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38230882

RESUMO

BACKGROUND AND AIM: Cohort studies have linked metabolic syndrome (MetS) to gastrointestinal (GI) cancer risk. We aimed to evaluate the associations between MetS, its components, and combinations of MetS components with eight GI cancers risk. METHODS: We conducted a systematic search of prospective cohort studies and performed a meta-analysis. Subgroup analyses regarding diagnostic criteria, sex, cancer sites, histological subtypes, ethnic groups, and studies adjusted for alcohol consumption were carried out. Mendelian randomization (MR) was employed to evaluate the causality between 17 MetS-related traits and eight GI cancers among Europeans and Asians separately. RESULTS: Meta-analyses of 31 prospective studies indicated that MetS was significantly associated with an increased risk of colorectal cancer (CRC) (RR [95% CI] = 1.13 [1.12-1.15]), esophageal cancer (EC) (RR [95% CI] = 1.17 [1.03-1.32]), gallbladder cancer (GBC) (RR [95% CI] = 1.37[1.10-1.71]), liver cancer (LC) (RR [95% CI] = 1.46 [1.29-1.64]), and pancreatic cancer (PaC) (RR [95% CI] = 1.25 [1.20-1.30]), but not gastric cancer (GC) (RR [95% CI] = 1.11 [0.96-1.28]). Regarding the associations between MetS components and GI cancers risk, the following associations showed statistical significance: obesity-CRC/LC/EC/, hypertriglyceridemia-LC/PaC, reduced high-density lipoprotein (HDL)-CRC/LC/GC/PaC, hyperglycemia-CRC/LC/PaC, and hypertension-CRC/LC/EC/PaC. Sex-specific associations were observed between individual MetS components on GI cancers risk. Among the top three common combinations in both sexes, obesity + HTN + hyperglycemia had the strongest association with CRC risk (RR [95% CI] = 1.54 [1.49-1.61] for males and 1.27 [1.21-1.33] for females). MR analyses revealed causality in 16 exposure-outcome pairs: waist-to-hip ratio/BMI/HbA1c-CRC; BMI/childhood obesity/waist circumference/T2DM/glucose-EC; BMI/waist circumference/cholesterol-LC; cholesterol/childhood obesity/waist circumference/HbA1c-PaC; and HbA1c-GBC. These results were robust against sensitivity analyses. CONCLUSIONS: Since MetS is reversible, lifestyle changes or medical interventions targeting MetS patients might be potential prevention strategies for GI cancers.


Assuntos
Neoplasias Gastrointestinais , Análise da Randomização Mendeliana , Síndrome Metabólica , Humanos , Síndrome Metabólica/genética , Síndrome Metabólica/epidemiologia , Neoplasias Gastrointestinais/genética , Neoplasias Gastrointestinais/etiologia , Neoplasias Gastrointestinais/epidemiologia , Estudos Prospectivos , Masculino , Fatores de Risco , Feminino , Risco , Estudos de Coortes , Obesidade/complicações
14.
Cancer ; 130(10): 1807-1815, 2024 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-38198291

RESUMO

BACKGROUND: Impaired fasting glucose (IFG) is associated with the risk of various cancers, but the cumulative effect of IFG on gastrointestinal cancer risk remains unclear. This study evaluated the association between the cumulative exposure to IFG and gastrointestinal cancer risk. METHODS: The authors extracted data from the Korean National Health Insurance Service and health examination data sets. Among individuals ≥40 years old who were free of diabetes or cancer, 1,430,054 who underwent national health examinations over 4 consecutive years from 2009 to 2012 were selected and followed up until gastrointestinal cancer diagnosis, death, or December 31, 2019. The IFG exposure score (range, 0-4) was based on the number of IFG diagnoses over 4 years. RESULTS: The median follow-up duration was 6.4 years. Consistent normoglycemia for 4 years was found in 44.3% of the population, whereas 5.0% had persistent IFG and 50.7% had intermittent IFG. Compared to the group with an IFG exposure score of 0, groups with IFG exposure scores of 1, 2, 3, and 4 had a 5%, 8%, 9%, and 12% increased risk of gastrointestinal cancer, respectively (score 1: adjusted hazard ratio [aHR], 1.05; 95% confidence interval [CI], 1.01-1.08; score 2: aHR, 1.08; 95% CI, 1.04-1.12; score 3: aHR, 1.09; 95% CI, 1.05-1.14; score 4: aHR, 1.12; 95% CI, 1.06-1.19). Persistent IFG exposure was also associated with higher risks of individual cancer types (colorectum, stomach, pancreas, biliary tract, and esophagus). CONCLUSIONS: Cumulative exposure to IFG is associated with an increased risk of developing gastrointestinal cancer, in a dose-dependent manner. PLAIN LANGUAGE SUMMARY: Hyperglycemia, including both diabetes and prediabetes, has been associated with an increased risk of various cancers. However, the cumulative effect of impaired fasting glucose on the risk of developing gastrointestinal cancer remains unclear. A frequent diagnosis of impaired fasting glucose was dose-dependently associated with a higher risk of developing overall gastrointestinal cancer. Furthermore, risks of individual cancer types increased with persistent impaired fasting glucose. Early detection of hyperglycemia and strict glycemic control can lower the risk of gastrointestinal cancer by reducing hyperglycemic burden. Additionally, for some individuals, lifestyle changes such as managing metabolic syndrome or abstaining from alcohol may also be helpful.


Assuntos
Glicemia , Jejum , Neoplasias Gastrointestinais , Humanos , Masculino , Feminino , Neoplasias Gastrointestinais/epidemiologia , Neoplasias Gastrointestinais/etiologia , Pessoa de Meia-Idade , Jejum/sangue , Glicemia/metabolismo , Glicemia/análise , República da Coreia/epidemiologia , Fatores de Risco , Adulto , Idoso , Estudos de Coortes
15.
Oncology ; 102(2): 141-156, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-37651986

RESUMO

INTRODUCTION: Previous observational studies have reported inconsistent findings on the association between consumption of sugar-sweetened soft drinks (SSSDs) and the risk of gastrointestinal (GI) cancer. This study investigated the associations between SSSD consumption and the risk of GI cancer using a systematic review and meta-analysis. METHODS: Observational epidemiological studies were searched from the PubMed and EMBASE databases until June 2021. We conducted a meta-analysis of all included studies and subgroup meta-analyses based on various factors. RESULTS: In a meta-analysis of 27 studies with nine case-control studies and 18 cohort studies, the consumption of SSSDs was modestly associated with an increased risk of GI cancer (odds ratio [OR]/relative risk [RR]: 1.08; 95% confidence interval [CI]: 1.01-1.16), with a significant positive dose-response relationship. In the subgroup meta-analysis by study design, there was a significant positive association between the consumption of SSSDs and GI cancer in cohort studies (RR: 1.11; 95% CI: 1.03-1.20; n = 18), but not in case-control studies. In the subgroup meta-analysis by type of cancer, consumption of SSSDs was significantly associated with an increased risk of colorectal cancer (OR/RR: 1.13; 95% CI: 1.07-1.19). CONCLUSIONS: This meta-analysis suggests that SSSD consumption significantly increases the risk of GI cancer, specifically colorectal cancer.


Assuntos
Neoplasias Colorretais , Neoplasias Gastrointestinais , Bebidas Adoçadas com Açúcar , Humanos , Açúcares , Fatores de Risco , Neoplasias Gastrointestinais/epidemiologia , Neoplasias Gastrointestinais/etiologia , Estudos Observacionais como Assunto
16.
Clin Gastroenterol Hepatol ; 22(3): 662-665.e1, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-37714395

RESUMO

Li-Fraumeni syndrome (LFS), linked to heterozygous germline pathogenic/likely pathogenic variants in TP53, confers exceptionally high cancer risk, including core cancers (sarcoma, breast, adrenocortical, and brain cancer) among many other cancer types.1 Colorectal cancer (CRC) is most common after the core and hematologic cancers, accounting for ∼2.8% of diagnoses. Stomach and esophageal cancers constitute another 1.3% (TP53 Database; R20, July 2019: https://tp53.isb-cgc.org).2.


Assuntos
Neoplasias Gastrointestinais , Síndrome de Li-Fraumeni , Humanos , Síndrome de Li-Fraumeni/complicações , Síndrome de Li-Fraumeni/genética , Proteína Supressora de Tumor p53/genética , Predisposição Genética para Doença , Mutação em Linhagem Germinativa , Neoplasias Gastrointestinais/epidemiologia , Neoplasias Gastrointestinais/etiologia
17.
Cancer Res Treat ; 56(1): 238-246, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-37536710

RESUMO

PURPOSE: Although obesity is associated with numerous diseases, the risks of disease may depend on metabolically healthy status. Nevertheless, it is unclear to whether metabolically healthy status affects risk of gastrointestinal (GI) cancer in general Chinese population. MATERIALS AND METHODS: A total of 114,995 participants who met the criteria were included from the Kailuan Study. The study participants were divided into four groups according to body mass index (BMI)/waist circumference (WC) and metabolic status. Incident of GI cancer (esophageal cancer, gastric cancer, liver cancer, biliary cancer, pancreatic cancer, and colorectal cancer) during 2006-2020 were confirmed by review of medical records. The Cox proportional hazard regression models were used to assess the association metabolically healthy status with the risk of GI cancer by calculating the hazard ratios (HR) and 95% confidence interval (CI). RESULTS: During a mean 13.76 years of follow-up, we documented 2,311 GI cancers. Multivariate Cox regression analysis showed that compared with the metabolically healthy normal-weight group, metabolically healthy obese (MHO) participants demonstrated an increased risk of developing GI cancer (HR, 1.54; 95% CI, 1.11 to 2.13) by BMI categories. However, such associations were not found for WC category. These associations were moderated by age, sex, and anatomical site of the tumor. Individuals with metabolic unhealthy normal-weight or metabolic unhealthy obesity phenotype also have an increased risk of GI cancer. CONCLUSION: MHO phenotype was associated with increased risk of GI cancer. Moreover, individuals who complicated by metabolic unhealthy status have an increased risk of developing GI cancer. Hence, clinicians should consider the risk of incident GI cancer in people with abnormal metabolically healthy status and counsel them about metabolic fitness and weight control.


Assuntos
Neoplasias Gastrointestinais , Obesidade Metabolicamente Benigna , Humanos , Fatores de Risco , Obesidade Metabolicamente Benigna/complicações , Obesidade Metabolicamente Benigna/epidemiologia , Obesidade Metabolicamente Benigna/patologia , Obesidade/epidemiologia , Índice de Massa Corporal , Neoplasias Gastrointestinais/etiologia , Neoplasias Gastrointestinais/complicações
19.
Am J Clin Nutr ; 119(2): 406-416, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-38042409

RESUMO

BACKGROUND: Dietary patterns have been associated with several cancers, especially gastrointestinal cancer (GIC). However, whether a healthy dietary pattern could modify the risk of GIC among people with different genetic backgrounds is not clear. OBJECTIVE: The objective of the study was to investigate how dietary patterns and genetic susceptibility contribute to the risk of GIC independently and jointly. METHODS: This large-scale prospective cohort study included 105,463 participants in UK Biobank who were aged 40-72 y and cancer-free at baseline. Dietary intake (Oxford WebQ) was used to calculate dietary pattern scores including dietary approach to stop hypertension (DASH) score and healthful plant-based diet index (hPDI). Genetic risk was quantified by a polygenic risk score (PRS) comprising 129 known GIC-associated loci. Cox proportional hazards regression was performed to estimate the associations of dietary patterns and PRS with GIC incidence after adjusting for potential confounders. RESULTS: Over a median follow-up of 11.70 y, 1,661 participants were diagnosed with GIC. DASH and hPDI were associated with 20% and 36% reductions, respectively, in GIC risk. Low PRS was associated with a 30 % decrease in GIC risk (HR: 0.70; 95% CI: 0.62, 0.79). Participants with healthy dietary scores at high-genetic risk had a lower GIC risk with HR of 0.77 (95% CI: 0.60, 0.98) for DASH and 0.66 (95% CI: 0.52, 0.84) for hPDI than those with unhealthy dietary score. Participants with both high-dietary score and low-genetic risk showed the lowest risk of GIC, with HR of 0.58 (95% CI: 0.45, 0.75) for DASH and 0.45 (95% CI: 0.34, 0.58) for hPDI. CONCLUSIONS: Adherence to DASH and hPDI were associated with a lower risk of some gastrointestinal cancers, and these 2 dietary patterns may partly compensate for genetic predispositions to cancer. Our results advance the development of precision medicine strategies that consider both dietary patterns and genetics to improve gastrointestinal health.


Assuntos
Neoplasias Gastrointestinais , Hipertensão , Humanos , Estudos Prospectivos , Padrões Dietéticos , Fatores de Risco , Dieta , Neoplasias Gastrointestinais/etiologia , Neoplasias Gastrointestinais/genética , Plantas , Estratificação de Risco Genético , Predisposição Genética para Doença
20.
J Cancer Res Ther ; 19(5): 1385-1391, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37787313

RESUMO

Background: One-third of all cancer deaths are preventable by alterations in diet. Methods: A case control study was conducted in a Regional Cancer Center in North India to evaluate the relationship of diet with selected gastrointestinal cancers. A total of 171 cases, 151 hospital controls, and 167 healthy controls were interviewed using food frequency questionnaire. Data was analyzed using odds ratio with 95% confidence interval and Chi-square test. Results: Two to three times increased risk of GI cancers was observed with hot and salted tea. Alcohol [OR 2.30 (1.32-4)] and smoking [OR (2.77 (1.77-4.33)] emerged as risk factors in healthy controls among whom freshly prepared food had significant protective effect [OR 0.57 (0.37-0.88)]. Sweet tea showed protective effect in hospital and healthy controls (OR 0.33 and 0.26, respectively). NSAIDS was associated with significantly higher risk of GI cancers. Consumption of dietary fibers decreased risk, which was significant for wheat and pulses but insignificant for rice. Vegetables and fruits showed significant protective effect ranging from 20 to 80% while intake of non-vegetarian foods showed significantly higher odds among controls (OR 2.37-13.4). Odds of GI cancer cases having consumed chutneys and pickles were significantly higher in comparison to healthy controls while consumption of dairy products showed protection. Low and medium intake of mixed spices inclusive of curcumin showed protection (OR 0.13 and 0.39, respectively) while intake of red chillies was associated with 2-30 times significantly higher odds. Conclusions: We have been able to generate baseline evidence of association between diet and selected GI cancers to encourage prevention and further research.


Assuntos
Dieta , Neoplasias Gastrointestinais , Humanos , Estudos de Casos e Controles , Dieta/efeitos adversos , Fatores de Risco , Neoplasias Gastrointestinais/epidemiologia , Neoplasias Gastrointestinais/etiologia , Chá , Razão de Chances
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA