Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 856
Filter
1.
Nutrients ; 16(13)2024 Jun 23.
Article in English | MEDLINE | ID: mdl-38999742

ABSTRACT

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.


Subject(s)
Fast Foods , Gastrointestinal Neoplasms , Humans , Italy/epidemiology , Female , Male , Middle Aged , Gastrointestinal Neoplasms/mortality , Gastrointestinal Neoplasms/etiology , Gastrointestinal Neoplasms/epidemiology , Risk Factors , Prospective Studies , Fast Foods/adverse effects , Fast Foods/statistics & numerical data , Aged , Adult , Food Handling , Proportional Hazards Models , Diet/adverse effects , Diet/statistics & numerical data , Surveys and Questionnaires , Food, Processed
2.
PLoS One ; 19(6): e0305994, 2024.
Article in English | MEDLINE | ID: mdl-38924054

ABSTRACT

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.


Subject(s)
Gastrointestinal Neoplasms , Observational Studies as Topic , Red Meat , Humans , Gastrointestinal Neoplasms/etiology , Gastrointestinal Neoplasms/epidemiology , Red Meat/adverse effects , Animals , Fishes , Meat Products/adverse effects , Risk Factors , Meat/adverse effects , Seafood , Diet/adverse effects
3.
Am J Gastroenterol ; 119(6): 1056-1065, 2024 06 01.
Article in English | MEDLINE | ID: mdl-38832708

ABSTRACT

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


Subject(s)
Fast Foods , Gastrointestinal Neoplasms , Humans , Gastrointestinal Neoplasms/epidemiology , Gastrointestinal Neoplasms/etiology , Fast Foods/adverse effects , Risk Factors , Colorectal Neoplasms/epidemiology , Colorectal Neoplasms/etiology , Stomach Neoplasms/epidemiology , Stomach Neoplasms/etiology , Food, Processed
4.
Hematol Oncol Clin North Am ; 38(4): 783-811, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38760197

ABSTRACT

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.


Subject(s)
Gastrointestinal Neoplasms , Humans , Gastrointestinal Neoplasms/therapy , Gastrointestinal Neoplasms/diagnosis , Gastrointestinal Neoplasms/genetics , Gastrointestinal Neoplasms/etiology , Precancerous Conditions/diagnosis , Precancerous Conditions/genetics , Precancerous Conditions/pathology , Precancerous Conditions/therapy , Genetic Predisposition to Disease , Early Detection of Cancer , Neoplastic Syndromes, Hereditary/diagnosis , Neoplastic Syndromes, Hereditary/genetics , Neoplastic Syndromes, Hereditary/therapy
5.
Int J Cancer ; 155(7): 1203-1211, 2024 Oct 01.
Article in English | MEDLINE | ID: mdl-38712628

ABSTRACT

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


Subject(s)
Esophageal Neoplasms , Helicobacter Infections , Helicobacter pylori , Stomach Neoplasms , Humans , Helicobacter Infections/complications , Helicobacter Infections/epidemiology , Middle Aged , Male , Female , Helicobacter pylori/isolation & purification , Adult , Stomach Neoplasms/microbiology , Stomach Neoplasms/epidemiology , Stomach Neoplasms/etiology , Stomach Neoplasms/pathology , Aged , Esophageal Neoplasms/epidemiology , Esophageal Neoplasms/microbiology , Esophageal Neoplasms/etiology , China/epidemiology , Cohort Studies , Risk Factors , Prevalence , Gastrointestinal Neoplasms/microbiology , Gastrointestinal Neoplasms/epidemiology , Gastrointestinal Neoplasms/etiology , Upper Gastrointestinal Tract/pathology , Upper Gastrointestinal Tract/microbiology
6.
Cancer Med ; 13(4): e7064, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38457256

ABSTRACT

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.


Subject(s)
Gastrointestinal Neoplasms , Lymphoma, B-Cell , Lymphoma , Humans , Immunotherapy, Adoptive/adverse effects , Receptors, Antigen, T-Cell , T-Lymphocytes , Ulcer/etiology , Lymphoma/therapy , Lymphoma, B-Cell/etiology , Gastrointestinal Neoplasms/therapy , Gastrointestinal Neoplasms/etiology , Cytokine Release Syndrome/etiology , Antigens, CD19 , Gastrointestinal Hemorrhage
7.
Cancer ; 130(10): 1807-1815, 2024 May 15.
Article in English | MEDLINE | ID: mdl-38198291

ABSTRACT

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.


Subject(s)
Blood Glucose , Fasting , Gastrointestinal Neoplasms , Humans , Male , Female , Gastrointestinal Neoplasms/epidemiology , Gastrointestinal Neoplasms/etiology , Middle Aged , Fasting/blood , Blood Glucose/metabolism , Blood Glucose/analysis , Republic of Korea/epidemiology , Risk Factors , Adult , Aged , Cohort Studies
8.
Clin Gastroenterol Hepatol ; 22(3): 662-665.e1, 2024 Mar.
Article in English | MEDLINE | ID: mdl-37714395

ABSTRACT

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.


Subject(s)
Gastrointestinal Neoplasms , Li-Fraumeni Syndrome , Humans , Li-Fraumeni Syndrome/complications , Li-Fraumeni Syndrome/genetics , Tumor Suppressor Protein p53/genetics , Genetic Predisposition to Disease , Germ-Line Mutation , Gastrointestinal Neoplasms/epidemiology , Gastrointestinal Neoplasms/etiology
9.
Am J Clin Nutr ; 119(2): 406-416, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38042409

ABSTRACT

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.


Subject(s)
Gastrointestinal Neoplasms , Hypertension , Humans , Prospective Studies , Dietary Patterns , Risk Factors , Diet , Gastrointestinal Neoplasms/etiology , Gastrointestinal Neoplasms/genetics , Plants , Genetic Risk Score , Genetic Predisposition to Disease
11.
Cancer Res Treat ; 56(1): 238-246, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37536710

ABSTRACT

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.


Subject(s)
Gastrointestinal Neoplasms , Obesity, Metabolically Benign , Humans , Risk Factors , Obesity, Metabolically Benign/complications , Obesity, Metabolically Benign/epidemiology , Obesity, Metabolically Benign/pathology , Obesity/epidemiology , Body Mass Index , Gastrointestinal Neoplasms/etiology , Gastrointestinal Neoplasms/complications
12.
Oncology ; 102(2): 141-156, 2024.
Article in English | MEDLINE | ID: mdl-37651986

ABSTRACT

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.


Subject(s)
Colorectal Neoplasms , Gastrointestinal Neoplasms , Sugar-Sweetened Beverages , Humans , Sugars , Risk Factors , Gastrointestinal Neoplasms/epidemiology , Gastrointestinal Neoplasms/etiology , Observational Studies as Topic
13.
J Cancer Res Ther ; 19(5): 1385-1391, 2023.
Article in English | MEDLINE | ID: mdl-37787313

ABSTRACT

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.


Subject(s)
Diet , Gastrointestinal Neoplasms , Humans , Case-Control Studies , Diet/adverse effects , Risk Factors , Gastrointestinal Neoplasms/epidemiology , Gastrointestinal Neoplasms/etiology , Tea , Odds Ratio
14.
Eur J Gastroenterol Hepatol ; 35(11): 1244-1252, 2023 11 01.
Article in English | MEDLINE | ID: mdl-37724454

ABSTRACT

The systematic review aimed to assess the association between vegetarian diet and the risk of gastrointestinal tumorigenesis. PubMed, Embase, Cochrane Library, and Web of Science were searched from inception to August 2022 for observational studies on vegetarian diets and the risk of gastrointestinal tumorigenesis. The primary outcome was morbidity due to gastrointestinal cancer. The Newcastle-Ottawa Scale was used to assess the quality of included studies. Pooled effects were analyzed using a random-effects model. The study protocol was registered in PROSPERO (no. CRD42022310187). Eight original studies (seven cohorts and one case-control), involving 686 691 participants, were included. Meta-analysis showed a negative correlation between vegetarian diets and gastrointestinal tumorigenesis risk [relative risk (RR) equals 0.77, 95% confidence interval (CI) is (0.65-0.90)], compared with non-vegetarian diets. Subgroup analysis indicated that vegetarian diets were negatively correlated with the risks of gastric cancer [RR = 0.41, 95% CI (0.28-0.61)] and colorectal cancer [RR = 0.85, 95% CI (0.76-0.95)], but not with that of upper gastrointestinal cancer (excluding stomach) [RR = 0.93, 95% CI (0.61-1.42)]. Vegetarian diets were negatively correlated with the risk of gastrointestinal tumorigenesis in men [RR = 0.57, 95% CI (0.36-0.91)], but were uncorrelated in women [RR = 0.89, 95% CI (0.71-1.11)]. Vegetarian diets were negatively correlated with the risk of gastrointestinal tumorigenesis in North American [RR = 0.76, 95% CI (0.61-0.95)] and Asian populations [RR = 0.43, 95% CI (0.26-0.72)] and were uncorrelated in the European population [RR = 0.83, 95% CI (0.68-1.01)]. Adhering to vegetarian diets reduces the risk of gastrointestinal tumorigenesis. More data from well-conducted cohort and other studies are needed.


Subject(s)
Gastrointestinal Neoplasms , Stomach Neoplasms , Male , Humans , Female , Diet, Vegetarian , Gastrointestinal Neoplasms/epidemiology , Gastrointestinal Neoplasms/etiology , Carcinogenesis
15.
BMC Cancer ; 23(1): 782, 2023 Aug 23.
Article in English | MEDLINE | ID: mdl-37612616

ABSTRACT

BACKGROUND: The association between gastrointestinal cancer and types of meat consumption, including red meat, processed meat, or a combination of both, remains disputable. Therefore, we performed a systematic review and meta-analysis of prospective cohort studies to estimate the association between meat consumption and gastrointestinal cancer risk. METHODS: PubMed, EmBase, and the Cochrane library databases were searched systematically for eligible studies that investigated the relation between meat consumption and the risk of developing gastrointestinal cancers, including esophageal cancer (EC), gastric cancer (GC), colorectal cancer (CRC), colon cancer (CC), rectal cancer (RC), pancreatic cancer (PC), and hepatocellular carcinoma (HCC) throughout February, 2023. The pooled relative risk (RR) with 95% confidence interval (CI) was assigned as an effect estimate and calculated using a random-effects model with inverse variance weighting. RESULTS: Forty cohorts comprising 3,780,590 individuals were selected for the final quantitative analysis. The summary results indicated that a higher red meat consumption was associated with an increased risk of CRC (RR: 1.09; 95% CI: 1.02-1.16; P = 0.007) and CC (RR: 1.13; 95% CI: 1.03-1.25; P = 0.011). Moreover, a higher processed meat consumption was associated with an increased risk of CRC (RR: 1.19; 95% CI: 1.13-1.26; P < 0.001), CC (RR: 1.24; 95% CI: 1.13-1.26; P < 0.001), and RC (RR: 1.24; 95% CI: 1.08-1.42; P = 0.002). Furthermore, a higher total consumption of red and processed meat was associated with an increased risk of CRC (RR: 1.13; 95% CI: 1.06-1.20; P < 0.001), CC (RR: 1.17; 95% CI: 1.04-1.33; P = 0.012), and RC (RR: 1.20; 95% CI: 1.04-1.39; P = 0.016). Finally, the strength of higher consumption of total red and processed meat with the risk of GC, and higher consumption of red meat with the risk of RC in subgroup of high adjusted level was lower than subgroup of moderate adjusted level, while the strength of higher consumption of processed meat with the risk of RC and HCC in subgroup of follow-up ≥ 10.0 years was higher than subgroup of follow-up < 10.0 years. CONCLUSIONS: This study found that meat consumption was associated with an increased risk of CRC, CC, and RC, and dietary intervention could be considered an effective strategy in preventing CRC.


Subject(s)
Carcinoma, Hepatocellular , Colonic Neoplasms , Gastrointestinal Neoplasms , Liver Neoplasms , Stomach Neoplasms , Humans , Prospective Studies , Gastrointestinal Neoplasms/epidemiology , Gastrointestinal Neoplasms/etiology , Meat/adverse effects
16.
Clin Sci (Lond) ; 137(15): 1095-1114, 2023 08 14.
Article in English | MEDLINE | ID: mdl-37530554

ABSTRACT

The increasing prevalence of obesity brings forward its importance as a risk factor for cancer development, particularly in the gastrointestinal tract. Obesity may trigger cancer development through several mechanisms, where metabolic deregulation of adipokines can modulate multiple oncogenic molecular pathways. Leptin and adiponectin are the most well-studied adipokines, and their imbalance can trigger different tumorigenic responses. Both epidemiologic and experimental studies have associated leptin with increased cancer risk and cell responsiveness in carcinogenesis and tumor invasion. On the other hand, adiponectin is reported to elicit the opposite effect. In addition to circulating or tissue adipokine levels, adiponectin, and leptin receptors or genetic polymorphisms may also play a role in cancer development. Moreover, adiponectin and leptin modulation offer valuable therapeutic approaches. We will review the links underpinning obesity and cancer development and focus on discussing the pathophysiological roles of leptin and adiponectin.


Subject(s)
Gastrointestinal Neoplasms , Leptin , Humans , Leptin/metabolism , Adiponectin/metabolism , Obesity/metabolism , Adipokines/metabolism , Gastrointestinal Neoplasms/etiology , Carcinogenesis
17.
Contemp Clin Trials ; 131: 107273, 2023 08.
Article in English | MEDLINE | ID: mdl-37380021

ABSTRACT

BACKGROUND: Oxaliplatin is a key chemotherapeutic agent in the treatment of local and metastatic gastrointestinal (GI) malignancies. Dose density and treatment adherence can be limited by chemotherapy-induced peripheral neuropathy (CIPN). Early research suggests CIPN incidence and severity may be mitigated by acupuncture, but rigorous data in GI oncology patients is limited. Here, we describe the protocol of a randomized, waitlist-controlled pilot study testing the use of preemptive of acupuncture plus acupressure to decrease CIPN and chemotherapy-related toxicities. METHODS: Patients with a GI malignancy (n = 56) with planned 5-fluorouracil (5-FU) and oxaliplatin IV (FOLFOX, FOLFIRINOX) every 2 weeks are being recruited. Additional concurrent anti-neoplastic agents may be used. Enrolled patients are randomized 1:1 to a 3-month intervention of Arm A: acupuncture with acupressure and standard-of-care treatment, or Arm B: standard-of-care alone. In Arm A, on days 1 and 3 of each chemotherapy cycle a standardized acupuncture protocol is administered and patients are taught self-acupressure to perform daily between chemotherapy treatments. Patients in both arms are given standard-of-care oral and peripheral (hands/feet) ice chip cryotherapy during oxaliplatin administration. CIPN and other symptoms are assessed at baseline, 6 weeks, and 3 months from registration. The primary endpoint is CIPN severity at 3 months (EORTC-CIPN 20). Additional endpoints evaluate CIPN incidence (CTCAE, Neuropen, tuning fork); incidence of pain, fatigue, nausea, oral dysesthesia, and anxiety; and feasibility (recruitment, retention, adherence, acceptability). If warranted, trial results will inform the design of a multi-center trial to expand testing of the intervention to a larger patient cohort.


Subject(s)
Acupressure , Acupuncture Therapy , Antineoplastic Agents , Gastrointestinal Neoplasms , Pancreatic Neoplasms , Peripheral Nervous System Diseases , Humans , Oxaliplatin/adverse effects , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Feasibility Studies , Antineoplastic Agents/adverse effects , Peripheral Nervous System Diseases/chemically induced , Peripheral Nervous System Diseases/prevention & control , Acupuncture Therapy/adverse effects , Acupuncture Therapy/methods , Gastrointestinal Neoplasms/drug therapy , Gastrointestinal Neoplasms/etiology , Cryotherapy , Randomized Controlled Trials as Topic , Multicenter Studies as Topic
18.
J Cancer Res Clin Oncol ; 149(13): 11369-11378, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37378673

ABSTRACT

PURPOSE: Sleep disorders are among the most common health problems worldwide and are linked to a variety of physical and mental health problems. Recently, there has been increasing evidence of an association between sleep disorders and cancer risk. We aimed to investigate this association specifically for cancers of the gastrointestinal (GI) tract. METHODS: Using the DA database (IQVIA), adult patients diagnosed with GI cancer between January 2010 and December 2022 were retrospectively compared to a 1:1 propensity score-matched cohort of patients without cancer. The outcome of the study was the association between sleep disorders and subsequent diagnosis of GI cancer. To determine whether sleep disorders were more common in patients with GI cancer than in patients without GI cancer, logistic regression models were used to estimate odds ratios (ORs) with 95% confidence intervals (95% CI). RESULTS: After matching, 37,161 cases with GI cancer and 37,161 controls without cancer were available for analysis. No association with cancer was found for sleep disorders in the overall history before the index date (OR 1.04; 95% CI 0.96-1.12), but considering sleep disorders documented within 1 year before the index date showed a positive association with GI cancer overall (OR 1.20; 95% CI 1.08-1.34). Stratified analyses by cancer site revealed higher odds of sleep disorders prior to diagnosis of gastric, pancreatic, and colorectal cancer. CONCLUSION: Our findings suggest that sleep disorders might be indicative of short-term health outcomes, including GI cancer, suggesting a role for sleep disorder screening in the context of cancer prevention efforts.


Subject(s)
Gastrointestinal Neoplasms , Sleep Wake Disorders , Adult , Humans , Case-Control Studies , Retrospective Studies , Risk Factors , Gastrointestinal Neoplasms/epidemiology , Gastrointestinal Neoplasms/etiology , Sleep Wake Disorders/complications , Sleep Wake Disorders/epidemiology
19.
Dis Mon ; 69(12): 101592, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37308362

ABSTRACT

Obesity has been recognized to be increasing globally and is designated a disease with adverse consequences requiring early detection and appropriate care. In addition to being related to metabolic syndrome disorders such as type 2 diabetes, hypertension, stroke, and premature coronary artery disease. Obesity is also etiologically linked to several cancers. The non-gastrointestinal cancers are breast, uterus, kidneys, ovaries, thyroid, meningioma, and thyroid. Gastrointestinal (GI) cancers are adenocarcinoma of the esophagus, liver, pancreas, gallbladder, and colorectal. The brighter side of the problem is that being overweight and obese and cigarette smoking are mostly preventable causes of cancers. Epidemiology and clinical studies have revealed that obesity is heterogeneous in clinical manifestations. In clinical practice, BMI is calculated by dividing a person's weight in kilograms by the square of the person's height in square meters (kg/m2). A BMI above 30 kg/m2 (defining obesity in many guidelines) is considered obesity. However, obesity is heterogeneous. There are subdivisions for obesity, and not all obesities are equally pathogenic. Adipose tissue, in particular, visceral adipose tissue (VAT), is endocrine and abdominal obesity (a surrogate for VAT) is evaluated by waist-hip measurements or just waist measures. Visceral Obesity, through several hormonal mechanisms, induces a low-grade chronic inflammatory state, insulin resistance, components of metabolic syndrome, and cancers. Metabolically obese, normal-weight (MONW) individuals in several Asian countries may have BMI below normal levels to diagnose obesity but suffer from many obesity-related complications. Conversely, some people have high BMI but are generally healthy with no features of metabolic syndrome. Many clinicians advise weight loss by dieting and exercise to metabolically healthy obese with large body habitus than to individuals with metabolic obesity but normal BMI. The GI cancers (esophagus, pancreas, gallbladder, liver, and colorectal) are individually discussed, emphasizing the incidence, possible pathogenesis, and preventive measures. From 2005 to 2014, most cancers associated with overweight and Obesity increased in the United States, while cancers related to other factors decreased. The standard recommendation is to offer or refer adults with a body mass index (BMI) of 30 or more to intensive, multicomponent behavioral interventions. However, the clinicians have to go beyond. They should critically evaluate BMI with due consideration for ethnicity, body habitus, and other factors that influence the type of obesity and obesity-related risks. In 2001, the Surgeon General's ``Call to Action to Prevent and Decrease Overweight and Obesity'' identified obesity as a critical public health priority for the United States. At government levels reducing obesity requires policy changes that improve the food and physical activity for all. However, implementing some policies with the most significant potential benefit to public health is politically tricky. The primary care physician, as well as subspecialists, should identify overweight and Obesity based on all the variable factors in the diagnosis. The medical community should address the prevention of overweight and Obesity as an essential part of medical care as much as vaccination in preventing infectious diseases at all levels- from childhood, to adolescence, and adults.


Subject(s)
Colorectal Neoplasms , Diabetes Mellitus, Type 2 , Gastrointestinal Neoplasms , Metabolic Syndrome , Adult , Female , Humans , United States , Child , Metabolic Syndrome/complications , Metabolic Syndrome/epidemiology , Overweight/complications , Overweight/epidemiology , Obesity/complications , Obesity/epidemiology , Gastrointestinal Neoplasms/diagnosis , Gastrointestinal Neoplasms/epidemiology , Gastrointestinal Neoplasms/etiology
SELECTION OF CITATIONS
SEARCH DETAIL