Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
1.
Nutr Rev ; 80(7): 1739-1754, 2022 06 09.
Article in English | MEDLINE | ID: mdl-35178575

ABSTRACT

CONTEXT: Current dietary guidelines recommend eggs as a part of a healthy diet. However, whether egg consumption is associated with risk of mortality remains controversial. Moreover, the dose-response association of egg consumption with risk of mortality has not been determined. OBJECTIVE: To determine the potential dose-response association of egg consumption with risk of mortality in the general population. DATA SOURCES: The PubMed and Embase databases were searched for publications meeting eligibility criteria through November 2021. DATA EXTRACTION: Required data were extracted by 1 reviewer and then checked for accuracy by another reviewer. A random-effects dose-response meta-regression model was used to calculate the pooled risk estimates. A restricted cubic spline model was used to test nonlinearity. The certainty of evidence was assessed using the GRADE system. DATA ANALYSIS: Nineteen prospective cohort studies, involving 1 737 893 participants, were included. The pooled hazard ratios for an increase of 1 egg/d were 1.08 (95%CI, 1.01-1.15) for all-cause mortality, 1.07 (95%CI, 0.97-1.18) for cardiovascular disease-caused mortality, and 1.16 (95%CI, 1.04-1.30) for cancer-caused mortality. The certainty of evidence for these observations was rated as very low. Nonlinear dose-response associations were found for egg consumption and all-cause, cardiovascular disease-caused, and cancer-caused mortality. Moreover, the positive association between egg consumption and all-cause mortality was more pronounced in studies with adjustment for blood cholesterol-related covariates than those without (Pinteraction = 0.011). CONCLUSIONS: Greater amount of egg consumption confers higher risks of death from all causes, cardiovascular disease, and canc er in a nonlinear dose-response pattern. These findings should be treated with caution and need to be confirmed by future studies.


Subject(s)
Cardiovascular Diseases , Neoplasms , Cardiovascular Diseases/etiology , Cause of Death , Diet , Humans , Prospective Studies , Risk Factors
2.
Am J Epidemiol ; 191(3): 472-486, 2022 02 19.
Article in English | MEDLINE | ID: mdl-34729579

ABSTRACT

We aimed to examine whether type 2 diabetes-prevention diet, a dietary pattern previously developed for reducing type 2 diabetes risk, was associated with mortality in a US population. A population-based cohort of 86,633 subjects was identified from the Prostate, Lung, Colorectal, and Ovarian Cancer Screening Trial (1993-2015). Dietary information was collected with a food frequency questionnaire. A dietary diabetes risk-reduction score was calculated to reflect adherence to this dietary pattern, with higher scores representing better adherence. Hazard ratios (HRs) and absolute risk differences (ARDs) in mortality rates per 10,000 person-years were calculated. After a mean follow-up of 13.6 years, 17,532 all-cause deaths were observed. Participants with the highest versus the lowest quintiles of dietary diabetes risk-reduction score were observed to have decreased risks of death from all causes (HR = 0.76, 95% CI: 0.72, 0.80; ARD: -81.94, 95% CI: -93.76, -71.12), cardiovascular disease (HR = 0.73, 95% CI: 0.66, 0.81; ARD: -17.82, 95% CI: -24.81, -11.30), and cancer (HR = 0.85, 95% CI: 0.78, 0.94; ARD: -9.92, 95% CI: -15.86, -3.59), which were modified by sex, smoking status, or alcohol consumption in subgroup analyses (P for interaction < 0.05 for all). In conclusion, a type 2 diabetes-prevention diet confers reduced risks of death from all causes, cardiovascular disease, and cancer in this US population.


Subject(s)
Cardiovascular Diseases , Diabetes Mellitus, Type 2 , Neoplasms , Cardiovascular Diseases/epidemiology , Cause of Death , Diabetes Mellitus, Type 2/epidemiology , Diabetes Mellitus, Type 2/prevention & control , Diet , Humans , Male , Neoplasms/prevention & control , Prospective Studies , Risk Factors
3.
Aging (Albany NY) ; 13(5): 7147-7165, 2021 02 26.
Article in English | MEDLINE | ID: mdl-33658397

ABSTRACT

Whether tenofovir disoproxil fumarate (TDF) is superior to entecavir in reducing hepatocellular carcinoma (HCC) risk among treatment-naïve chronic hepatitis B (CHB) patients remains controversial. We aimed to clarify this controversy. Several databases, including PubMed and Embase, were retrieved through November 2020. Cohort studies comparing the effectiveness of TDF and entecavir in reducing HCC incidence among treatment-naïve CHB patients were included if they reported multivariable-adjusted or propensity-score-matched risk estimates. A random-effects model was used to pool hazard ratios (HRs). Thirteen cohort studies, involving 4097 HCC cases and 80202 CHB patients, were included. Multivariable-adjusted meta-analysis revealed no significant difference in HCC incidence between TDF and entecavir groups (HR 0.86, 95% confidence interval 0.72-1.04), which was consistent with propensity-score-matched meta-analysis (HR 0.83, 95% confidence interval 0.66-1.03). Subgroup analysis showed that the observed similarity of TDF to entecavir for HCC prevention persisted in studies with follow-up length of ≥4 years but not in those with follow-up length of <4 years (Pinteraction<0.01). In conclusion, TDF is similar to entecavir in reducing HCC incidence among treatment-naïve CHB patients. Heterogeneous results of included studies may result from their disparity in follow-up length. Our findings should be treated with caution and need to be further confirmed.


Subject(s)
Antiviral Agents/therapeutic use , Carcinoma, Hepatocellular/prevention & control , Guanine/analogs & derivatives , Hepatitis B, Chronic/drug therapy , Liver Neoplasms/prevention & control , Tenofovir/therapeutic use , Carcinoma, Hepatocellular/etiology , Guanine/therapeutic use , Humans , Liver Neoplasms/etiology , Risk Factors
4.
Int J Behav Nutr Phys Act ; 18(1): 21, 2021 02 03.
Article in English | MEDLINE | ID: mdl-33536027

ABSTRACT

BACKGROUND: Ultra-processed foods have now become dominant in the global food system. Whether their consumption is associated with cardiovascular mortality remains controversial. Moreover, data on ultra-processed foods and cardiovascular outcomes are scarce in the US population. We aimed to examine the association of ultra-processed food consumption with cardiovascular mortality in a US population. METHODS: A population-based cohort of 91,891 participants was identified from the Prostate, Lung, Colorectal, and Ovarian Cancer Screening Trial. Dietary data were collected through a validated 137-item food frequency questionnaire. Ultra-processed foods were defined by the NOVA classification. Cox regression was used to calculate hazard ratios (HRs) and 95% confidence intervals (CIs) for cardiovascular mortality. Restricted cubic spline regression was used to test nonlinearity. Subgroup analyses were conducted to identify the potential effect modifiers. RESULTS: After an average follow-up of 13.5 years (1,236,049.2 person-years), 5490 cardiovascular deaths were documented, including 3985 heart disease deaths and 1126 cerebrovascular deaths. In the fully adjusted model, participants in the highest vs. the lowest quintiles of ultra-processed food consumption had higher risks of death from cardiovascular disease (HRquintile 5 vs. 1, 1.50; 95% CI, 1.36-1.64) and heart disease (HRquintile 5 vs. 1, 1.68; 95% CI, 1.50-1.87) but not cerebrovascular disease (HRquintile 5 vs. 1, 0.94; 95% CI, 0.76-1.17). A nonlinear dose-response pattern was observed for overall cardiovascular and heart disease mortality (all Pnonlinearity < 0.05), with a threshold effect observed at ultra-processed food consumption of 2.4 servings/day and 2.3 servings/day, respectively; below the thresholds, no significant associations were observed for these two outcomes. Subgroup analyses showed that the increased risks of mortality from ultra-processed foods were significantly higher in women than in men (all Pinteraction < 0.05). CONCLUSIONS: High consumption of ultra-processed foods is associated with increased risks of overall cardiovascular and heart disease mortality. These harmful associations may be more pronounced in women. Our findings need to be confirmed in other populations and settings.


Subject(s)
Cardiovascular Diseases/mortality , Diet/statistics & numerical data , Fast Foods/statistics & numerical data , Humans , Prospective Studies , United States
5.
Carcinogenesis ; 42(5): 724-732, 2021 05 28.
Article in English | MEDLINE | ID: mdl-33480980

ABSTRACT

Low-carbohydrate diets have become a popular approach for weight loss in recent years. However, whether low-carbohydrate diets are associated with the risk of pancreatic cancer remains to be elucidated. Hence, we examined the association of low-carbohydrate diets with the risk of pancreatic cancer in a US population. A population-based cohort of 95 962 individuals was identified. A low-carbohydrate-diet score was calculated to quantify adherence to this dietary pattern, with higher scores indicating greater adherence. Cox regression was used to calculate risk estimate for the association of the low-carbohydrate-diet score with the risk of pancreatic cancer. Subgroup analysis was used to identify the potential effect modifiers. After an average follow-up of 8.87 years (875856.9 person-years), we documented a total of 351 pancreatic cancer cases. In the fully adjusted model, the highest versus the lowest quartiles of the overall low-carbohydrate-diet score were found to be associated with a reduced risk of pancreatic cancer (hazard ratioquartile 4 versus 1: 0.61; 95% confidence interval: 0.45, 0.82; Ptrend < 0.001). Subgroup analysis found that the inverse association of low-carbohydrate diets with the risk of pancreatic cancer was more pronounced in individuals aged ≥65 years than in those aged <65 years (Pinteraction = 0.015). Similar results were obtained for animal and vegetable low-carbohydrate-diet scores. In conclusion, low-carbohydrate diets, regardless of the type of protein and fat, are associated with a lower risk of pancreatic cancer in the US population, suggesting that adherence to low-carbohydrate diets may be beneficial for pancreatic cancer prevention. Future studies should validate our findings in other populations.


Subject(s)
Diet, Carbohydrate-Restricted/adverse effects , Dietary Carbohydrates/metabolism , Pancreatic Neoplasms/metabolism , Aged , Cohort Studies , Dietary Fats/metabolism , Female , Humans , Male , Middle Aged , Pancreatic Neoplasms/etiology , Pancreatic Neoplasms/pathology , Risk Factors , Weight Loss/physiology
6.
Cancer Med ; 9(18): 6843-6853, 2020 09.
Article in English | MEDLINE | ID: mdl-32716590

ABSTRACT

BACKGROUND: Whether adherence to the World Cancer Research Fund/American Institute for Cancer Research (WCRF/AICR) cancer prevention recommendations is associated with a reduced risk of pancreatic cancer remains controversial. Additionally, no study has investigated this association in the US population. Hence, we investigated the association of adherence to the 2018 WCRF/AICR cancer prevention recommendations with pancreatic cancer incidence and mortality in a US population. METHODS: A population-based cohort of 95 962 participants was identified. A score incorporating eight WCRF/AICR components was constructed to reflect adherence to the WCRF/AICR guidelines, with higher scores representing greater adherence to the guidelines. Cox and competing risk regression were used to calculate risk estimates for pancreatic cancer incidence and mortality, respectively. Restricted cubic spline functions were used to test nonlinearity. RESULTS: In the fully adjusted model, higher overall WCRF/AICR scores were shown to be associated with lower risks of developing pancreatic cancer (hazard ratiotertile 3 vs 1 :0.67; 95% confidence interval: 0.49, 0.90; Ptrend  = .0099) and mortality due to this cancer (subdistribution hazard ratiotertile 3 vs 1 0.65; 95% confidence interval: 0.47, 0.89; Ptrend  = .0108) in a linear dose-response manner (all Pnonlinearity  > .05). The component "be physically active" was shown to be a key contributor to the observed associations. No association of the diet-specific WCRF/AICR score with pancreatic incidence and mortality was found. CONCLUSIONS: Adherence to the 2018 WCRF/AICR guidelines, especially "be physically active," confers reduced risks of pancreatic cancer incidence and mortality in the US population; however, adherence to dietary components alone does not confer such beneficial effects.


Subject(s)
Healthy Lifestyle , Pancreatic Neoplasms/epidemiology , Pancreatic Neoplasms/prevention & control , Risk Reduction Behavior , Aged , Cause of Death , Diet, Healthy , Exercise , Female , Humans , Incidence , Male , Middle Aged , Pancreatic Neoplasms/diagnosis , Pancreatic Neoplasms/mortality , Prospective Studies , Protective Factors , Risk Assessment , Risk Factors , Smoking Cessation , Time Factors , United States/epidemiology
7.
Eur J Prev Cardiol ; 27(11): 1187-1203, 2020 07.
Article in English | MEDLINE | ID: mdl-32283957

ABSTRACT

OBJECTIVE: The association between high-density lipoprotein cholesterol (HDL-C) levels and mortality remains controversial. We aimed to investigate the potential dose-response associations between HDL-C levels and mortality from all causes, cardiovascular disease and cancer in the general population. METHODS: PubMed and Embase were searched through April 2019. Prospective cohort studies reporting risk estimates of HDL-C levels and mortality were included. Linear and non-linear dose-response analyses were conducted. A random-effects model was employed to calculate pooled hazard ratio. RESULTS: Thirty-seven studies, involving 3,524,505 participants and more than 612,027 deaths, were included. HDL-C level was found to be associated with mortality from all causes, cardiovascular disease and cancer in a J-shaped dose-response pattern, with the lowest risk observed at HDL-C levels of 54-58 mg/dL, 68-71 mg/dL and 64-68 mg/dL, respectively. Compared with HDL-C level of 56 mg/dL, the pooled hazard ratios for all-cause mortality were 1.03 (95% confidence interval (CI) 1.01, 1.05) and 1.10 (95% CI 1.09, 1.12) for each 10-mg/dL increase and decrease in HDL-C levels, respectively; furthermore, compared with the reference category, the pooled hazard ratios for all-cause mortality were 1.21 (95% CI 1.09, 1.36) and 1.36 (95% CI 1.21, 1.53) for the highest and the lowest categories of HDL-C levels, respectively. Similar results were obtained for cardiovascular and cancer mortality. CONCLUSIONS: In the general population, HDL-C level is associated with mortality from all causes, cardiovascular disease and cancer in a J-shaped dose-response manner; both extremely high and low HDL-C levels are associated with an increased risk of mortality.


Subject(s)
Cardiovascular Diseases/mortality , Cholesterol, HDL/blood , Neoplasms/complications , Biomarkers/blood , Cardiovascular Diseases/blood , Cardiovascular Diseases/etiology , Cause of Death/trends , Follow-Up Studies , Global Health , Humans , Neoplasms/blood , Neoplasms/mortality , Prognosis , Prospective Studies , Risk Factors , Survival Rate/trends
8.
Cancer Epidemiol Biomarkers Prev ; 29(5): 1019-1028, 2020 05.
Article in English | MEDLINE | ID: mdl-32051196

ABSTRACT

BACKGROUND: Total antioxidant capacity (TAC) reflects an individual's overall antioxidant intake. We sought to clarify whether higher TAC is associated with lower risks of pancreatic cancer incidence and mortality in the U.S. general population. METHODS: A total of 96,018 American adults were identified from the Prostate, Lung, Colorectal, and Ovarian Cancer Screening Trial. A ferric-reducing ability of plasma score was used to reflect an individual's TAC intake from diet and/or supplements. Cox regression was used to calculate hazard ratios (HR) for pancreatic cancer incidence, and competing risk regression was used to calculate subdistribution HRs for pancreatic cancer mortality. Restricted cubic spline regression was used to test nonlinearity. RESULTS: A total of 393 pancreatic cancer cases and 353 pancreatic cancer-related deaths were documented. Total (diet + supplements) TAC was found to be inversely associated with pancreatic cancer incidence (HR quartile 4 vs. quartile 1 = 0.53; 95% confidence interval, 0.39-0.72; P trend = 0.0002) and mortality (subdistribution HR quartile 4 vs. quartile 1 = 0.52; 95% confidence interval 0.38-0.72; P trend = 0.0003) in a nonlinear dose-response manner (all P nonlinearity < 0.01). Similar results were observed for dietary TAC. No association of supplemental TAC with pancreatic cancer incidence and mortality was found. CONCLUSIONS: In the U.S. general population, dietary but not supplemental TAC level is inversely associated with risks of pancreatic cancer incidence and mortality in a nonlinear dose-response pattern. IMPACT: This is the first prospective study indicating that a diet rich in antioxidants may be beneficial in decreasing pancreatic cancer incidence and mortality.


Subject(s)
Antioxidants/administration & dosage , Diet Surveys/statistics & numerical data , Feeding Behavior , Pancreatic Neoplasms/epidemiology , Aged , Dietary Supplements , Female , Humans , Incidence , Male , Middle Aged , Mortality , Multicenter Studies as Topic , Pancreatic Neoplasms/prevention & control , Proportional Hazards Models , Prospective Studies , Randomized Controlled Trials as Topic , Risk Assessment/methods , Risk Assessment/statistics & numerical data , Risk Factors , United States/epidemiology
9.
Int J Cancer ; 147(6): 1577-1586, 2020 09 15.
Article in English | MEDLINE | ID: mdl-32105342

ABSTRACT

Epidemiological studies on magnesium intake and primary liver cancer (PLC) are scarce, and no prospective studies have examined the associations of magnesium intake with PLC incidence and mortality. We sought to clarify whether higher magnesium intake from diet and supplements was associated with lower risks of PLC incidence and mortality in the US population. Magnesium intake from diet and supplements was evaluated through a food frequency questionnaire in a cohort of 104,025 participants. Cox regression was employed to calculate hazard ratios for PLC incidence and competing risk regression was employed to calculate subdistribution hazard ratios for PLC mortality. Restricted cubic spline regression was employed to test nonlinearity. We documented 116 PLC cases during 1,193,513.5 person-years of follow-up and 100 PLC deaths during 1,198,021.3 person-years of follow-up. Total (diet + supplements) magnesium intake was found to be inversely associated with risks of PLC incidence (hazard ratiotertile 3 vs. 1 : 0.44; 95% confidence interval: 0.24, 0.80; ptrend = 0.0065) and mortality (subdistribution hazard ratiotertile 3 vs. 1 : 0.37; 95% confidence interval: 0.19, 0.71; ptrend = 0.0008). Similar results were obtained for dietary magnesium intake. Nonlinear inverse dose-response associations with PLC incidence and mortality were observed for both total and dietary magnesium intakes (all pnonlinearity < 0.05). In summary, in the US population, a high magnesium intake is associated with decreased risks of PLC incidence and mortality in a nonlinear dose-response manner. These findings support that increasing the consumption of foods rich in magnesium may be beneficial in reducing PLC incidence and mortality.


Subject(s)
Diet Surveys/statistics & numerical data , Feeding Behavior , Liver Neoplasms/epidemiology , Magnesium Deficiency/diet therapy , Magnesium/administration & dosage , Aged , Dietary Supplements , Early Detection of Cancer/statistics & numerical data , Female , Follow-Up Studies , Humans , Incidence , Liver Neoplasms/diagnosis , Liver Neoplasms/etiology , Liver Neoplasms/prevention & control , Magnesium Deficiency/complications , Male , Middle Aged , Multicenter Studies as Topic , Prospective Studies , Randomized Controlled Trials as Topic , Risk Factors , Survival Analysis
10.
Gastrointest Endosc ; 91(3): 684-697.e15, 2020 03.
Article in English | MEDLINE | ID: mdl-31790657

ABSTRACT

BACKGROUND AND AIMS: The fecal immunochemical test (FIT) and colonoscopy are the most commonly used strategies for colorectal cancer (CRC) screening worldwide. We aimed to compare their efficacy and cost-effectiveness in CRC screening in an average-risk population. METHODS: PubMed, Embase, and National Health Services Economic Evaluation Database were searched. Risk ratio (RR) was used to evaluate the differences in detection rates of colorectal neoplasia between FIT and colonoscopy groups. A random-effects model was used to pool RRs. Incremental cost-effectiveness ratios (ICERs) were calculated to evaluate the cost-effectiveness of FIT versus colonoscopy. RESULTS: Six randomized controlled trials and 17 cost-effectiveness studies were included. The participation rate in the FIT group was higher than that in the colonoscopy group (41.6% vs 21.9%). In the intention-to-treat analysis, FIT had a detection rate of CRC comparable with colonoscopy (RR, .73; 95% confidence interval, .37-1.42) and lower detection rates of any adenoma and advanced adenoma than 1-time colonoscopy. Most included cost-effectiveness studies showed that annual (13/15) or biennial (5/6) FIT was cost-saving (ICER < $0) or very cost-effective ($0 < ICER ≤ $25000/quality-adjusted life-year) compared with colonoscopy every 10 years. CONCLUSIONS: FIT may be similar to 1-time colonoscopy in the detection rate of CRC, although it has lower detection rates of any adenoma and advanced adenoma than 1-time colonoscopy. Furthermore, annual or biennial FIT appears to be very cost-effective or cost-saving compared with colonoscopy every 10 years. These findings indicate, at least partly, that FIT is noninferior to colonoscopy in CRC screening in an average-risk population. Our findings should be treated with caution and need to be further confirmed.


Subject(s)
Adenoma/diagnosis , Colonoscopy , Colorectal Neoplasms , Early Detection of Cancer , Feces/chemistry , Adenoma/pathology , Colonoscopy/economics , Colorectal Neoplasms/diagnosis , Colorectal Neoplasms/pathology , Cost-Benefit Analysis , Early Detection of Cancer/economics , Humans , Immunochemistry/economics , Mass Screening/economics , Occult Blood , Randomized Controlled Trials as Topic , Treatment Outcome
11.
PLoS One ; 14(12): e0225792, 2019.
Article in English | MEDLINE | ID: mdl-31800606

ABSTRACT

BACKGROUND: ABO blood group has been associated with cardiovascular disease and cancer. However, whether ABO blood group is associated with nonalcoholic fatty liver disease (NAFLD) remains unknown. The present study aimed to clarify this issue. METHODS: A hospital-based case-control study was performed in southwestern China. A total of 583 newly ultrasound-diagnosed NAFLD cases and 2068 controls were included. The adjusted odds ratios (ORs) and 95% confidence intervals (CIs) of developing NAFLD were calculated by multivariate logistic regression. A propensity score was developed for adjustment and matching. RESULTS: The proportions of blood groups A, B, AB and O were 31%, 26%, 8% and 35%, respectively. Non-O blood groups were found to be significantly associated with an increased risk of NAFLD (the fully adjusted OR = 1.51, 95% CI: 1.19, 1.91); moreover, compared with blood group O, the fully adjusted ORs of developing NAFLD were 1.50 (95% CI: 1.13, 1.99) for blood group A, 1.59 (95% CI: 1.19, 2.14) for blood group B, and 1.37 (95% CI: 0.86, 2.18) for blood group AB. Similar results were obtained in both propensity-score-adjusted and propensity-score-matched analyses. No evidence of significant effect modification for the association of ABO blood group with the risk of NAFLD was found (all Pinteraction>0.05). CONCLUSIONS: Non-O blood groups are significantly associated with an increased risk of NAFLD. Our findings provide some epidemiological evidence for a possible role of ABO glycosyltransferase in the pathogenesis of NAFLD. However, these findings need to be validated by future studies.


Subject(s)
ABO Blood-Group System , Asian People , Non-alcoholic Fatty Liver Disease/blood , Non-alcoholic Fatty Liver Disease/epidemiology , Adult , Aged , Case-Control Studies , China/epidemiology , Comorbidity , Disease Susceptibility , Female , Humans , Male , Middle Aged , Non-alcoholic Fatty Liver Disease/diagnosis , Odds Ratio , Propensity Score , Risk Assessment
12.
Sci Rep ; 9(1): 11124, 2019 07 31.
Article in English | MEDLINE | ID: mdl-31366982

ABSTRACT

Whether nonalcoholic fatty liver disease (NAFLD) is associated with an increased risk of mortality remains controversial. The present study aimed to clarify this issue. A systematic search of PubMed and Embase was conducted through October 2018. Studies providing risk estimates of NAFLD and mortality were included. A random-effects model was employed to calculate summary risk estimates. Subgroup analyses were performed to identify potential effect modifiers. Fourteen studies, involving 498501 subjects and 24234 deaths, were included. Patients with NAFLD were found to be at an elevated risk of all-cause mortality compared with those without [hazard ratio (HR) = 1.34; 95% confidence interval (CI) 1.17-1.54)]. The significantly positive association between NAFLD and all-cause mortality could not be modified by age, sex, follow-up duration, and adjustment for body mass index, diabetes, smoking or hypertension (all Pinteraction > 0.05), and remained in sensitivity analyses. No significant associations of NAFLD with CVD (HR = 1.13; 95% CI 0.92-1.38) and cancer (HR = 1.05; 95% CI 0.89-1.25) mortality were found. In conclusion, NAFLD is a predictor of increased all-cause mortality but not CVD and cancer mortality. These findings have important implications for decision making in public health and clinical practice, and highlight the urgency of developing effective treatments for NAFLD.


Subject(s)
Cardiovascular Diseases/mortality , Neoplasms/mortality , Non-alcoholic Fatty Liver Disease/mortality , Adult , Aged , Aged, 80 and over , Female , Follow-Up Studies , Humans , Male , Middle Aged , Observational Studies as Topic , Risk Factors , Young Adult
13.
Hum Reprod Update ; 23(1): 126-138, 2016 12.
Article in English | MEDLINE | ID: mdl-27655589

ABSTRACT

BACKGROUND: A striking gender disparity in the incidence and outcome of primary liver cancer (PLC) has been well recognized. Mounting evidence from basic research suggests that hormonal factors may be involved in the gender disparity of PLC. Whether hormonal exposures in human subjects are associated with PLC risk is largely unknown. OBJECTIVE AND RATIONALE: Whether reproductive factors and use of menopausal hormone therapies (MHTs) in women are associated with PLC risk remains controversial. We conducted this study to clarify this issue. SEARCH METHODS: PubMed and EMBASE were searched to July, 2016 for studies published in English or Chinese. Observational studies (cohort, nested case-control and case-control) that provided risk estimates of reproductive factors, MHTs and PLC risk were eligible. The quality of included studies was determined based on the Newcastle-Ottawa quality assessment scale. Summary risk ratios (RRs) were calculated using a random-effects model. Dose-response analysis was conducted where possible. OUTCOMES: Fifteen peer-reviewed studies, involving 1795 PLC cases and 2 256 686 women, were included. Overall meta-analyses on parity and PLC risk did not find any significant associations; however, when restricting to studies with PLC cases ≥100, increasing parity was found to be significantly associated with a decreased risk of PLC [RR for the highest versus lowest parity 0.67, 95% CI 0.52, 0.88; RR for parous versus nulliparous 0.71, 95% CI 0.53, 0.94; RR per one live birth increase 0.93, 95% CI 0.88, 0.99]. A J-shaped relationship between parity and PLC risk was identified (Pnon-linearity < 0.01). Compared with never users, the pooled RRs of PLC were 0.60 (95% CI 0.37, 0.96) for ever users of MHT, 0.73 (95% CI 0.46, 1.17) for ever users of estrogen-only therapy (ET) and 0.67 (95% CI 0.45, 1.02) for ever users of estrogen-progestin therapy (EPT). The pooled RR of PLC for the oldest versus youngest category of menarcheal age was 0.50 (95% CI 0.32, 0.79). Oophorectomy was significantly associated with an increased risk of PLC (RR 2.23, 95% CI 1.46, 3.41). No significant association of age at first birth, and spontaneous or induced abortion with PLC risk was found. No meta-analysis was performed for the association of age at menopause, breastfeeding, hysterectomy, menopausal status and stillbirth with PLC risk owing to huge methodological heterogeneity and/or very limited studies. WIDER IMPLICATIONS: Parity is associated with PLC risk in a J-shaped dose-response pattern. Late age at menarche and ever use of MHT are associated with a reduced risk of PLC, whereas there is no association of ever use of ET and EPT, age at first birth, or spontaneous and induced abortion with PLC risk. Compared to women with no history of oophorectomy, those with a history of oophorectomy are at an increased risk of PLC. Our findings provide some epidemiological support for a role of hormonal exposures in the development of PLC in women. However, these findings should be interpreted with much caution because of the limited number of studies and potential biases, and need to be validated by studies with good design and large sample size.


Subject(s)
Estrogens/adverse effects , Hormone Replacement Therapy/adverse effects , Liver Neoplasms/chemically induced , Progestins/adverse effects , Reproductive History , Contraceptives, Oral/adverse effects , Dose-Response Relationship, Drug , Female , Humans , Incidence , Menarche , Observational Studies as Topic , Odds Ratio , Parity , Pregnancy , Risk Factors , Sex Factors
14.
Sci Rep ; 6: 26256, 2016 05 20.
Article in English | MEDLINE | ID: mdl-27198922

ABSTRACT

A protective effect of statins on primary liver cancer (PLC) risk has been suggested. However, issues about the dose-response relationship, the protective effect of individual statins, and PLC risk reduction among at-risk populations remain unsolved. Therefore, a meta-analysis was conducted. PubMed and EMBASE were searched for studies providing the risk ratio (RR) on statins and PLC risk. Summary RRs were calculated using a random-effects model. Twenty-five studies were identified. Stain use was significantly associated with a reduced risk of PLC (RR = 0.60, 95% confidence interval (CI) = 0.53-0.69). The summary RR for every additional 50 cumulative defined daily doses per year was 0.87 (95% CI = 0.83-0.91). Evidence of a non-linear dose-response relationship between statins and PLC risk was found (Pnon-linearity < 0.01). All individual statins significantly reduced PLC risk, and the risk reduction was more evident with rosuvastatin. The inverse association between statins and PLC risk remained among populations with common risk factors. Subgroup analyses revealed more significant reduction in PLC risk by statins in high- versus non-high-risk populations (Pinteraction = 0.02). Overall, these findings add to our understanding of the association between statins and PLC risk. Whether statin use is causally associated with a reduced risk of PLC should be further studied.


Subject(s)
Hydroxymethylglutaryl-CoA Reductase Inhibitors/therapeutic use , Liver Neoplasms/prevention & control , Dose-Response Relationship, Drug , Humans , Odds Ratio
SELECTION OF CITATIONS
SEARCH DETAIL
...