Skip to main content
Hepatology Communications logoLink to Hepatology Communications
. 2025 Jan 16;9(2):e0627. doi: 10.1097/HC9.0000000000000627

Environmental exposures and the risk of hepatocellular carcinoma

Divya Rayapati 1, Katherine A McGlynn 2, John D Groopman 3,, Amy K Kim 1
PMCID: PMC11737496  PMID: 39813595

Abstract

The global epidemiology of HCC is shifting due to changes in both established and emerging risk factors. This transformation is marked by an emerging prevalence of metabolic dysfunction–associated steatotic liver disease (MASLD) and type 2 diabetes, alongside traditional risks such as viral hepatitis (HBV and HCV), and exposure to chemical agents like aflatoxin, alcohol, tobacco, and air pollution. This review examines how environmental exposures and evolving liver pathology, exacerbated by lifestyle and metabolic conditions, are contributing to the rising worldwide incidence of HCC. Effective prevention strategies must not only address traditional risk factors through vaccination and therapeutic measures but also confront metabolic and socioeconomic disparities through comprehensive public health efforts. As the burden of liver cancer continues to grow, particularly in resource-limited settings, an expansive and inclusive approach is vital for mitigating its impact across diverse populations.

Keywords: aflatoxin, environmental exposure, liver cancer, metabolic dysfunction-associated steatotic liver disease, viral hepatitis

INTRODUCTION

An epidemiologic transition is underway in the etiology of liver cancer, particularly HCC, that impacts future cancer therapeutics, prevention, and control interventions for this often fatal disease. The rising proportion of people across the globe with metabolic syndrome, obesity, diabetes, and metabolic dysfunction–associated steatotic liver disease (MASLD) is accelerating the risk for liver cancer in people exposed to established hepatocarcinogens. The established carcinogens implicated in this context include viruses such as HBV and HCV, individual chemical agents like aflatoxin B1 (AFB1) and alcohol, as well as complex mixtures such as tobacco smoke. Additionally, emerging exposures to other complex mixtures, notably air pollution, may also play a significant role in contributing to future cases of HCC. Each of these risk factors varies in their global prevalence, OR for developing HCC, and population attributable fraction both in the United States and globally (Table 1).

TABLE 1.

Risk factors with epidemiologic metrics

Risk Factors Prevalence (%) OR of HCC PAF in United States (%)1 PAF globally (%)2
HBV 3.33 234 4.3-6.3 44
HCV 0.73 174 20.5-35.4 21
Aflatoxin 555 5.56 4.6-28.2
Air pollution 997
Alcohol consumption 178 3-109 13.4-26 26
Tobacco use 22.310 1.39-1.911 9 13
MASLD 25.2-29.812 3.6313 42.1a 16a
Type II Diabetes Mellitus 10.514 1.5315

Note: Denotes lack of sufficient data.

a

PAF for MASLD and type II diabetes mellitus merged based on overlapping data in metabolic factors data.

Abbreviations: MASLD, metabolic associated steatotic liver disease; PAF, population attributable fraction.

Burden of HCC

Liver cancer remains one of the three leading causes of cancer mortality worldwide and about 85% of liver cancer is HCC, thus the diagnosis and management is an important public health problem.16 Over the past 75 years, the incidence and mortality rates of HCC were greater in low-income and middle-income countries.17 The rates of HCC, however, rose rapidly in the late 20th and early 21st centuries in many formerly low-rate countries.18,19,20,21,22 In the United States, individuals with Hispanic ancestry account for the second-highest number of liver cancer deaths.23,24,25,26 In the most recent report on Cancer Health Disparities in the United States, the American Association for Cancer Research outlined that Hispanic persons have twice the rate of liver cancer mortality compared to other ethnic groups.27 While projections of future numbers of cases have wide confidence intervals, a recent study forecasts that by 2040 the number of liver cancer cases in the world will increase by nearly 55%, based on projections of growth and aging of populations.28 This increase might also be affected by the increasing numbers of MASLD.

The impact of HCC incidence by sex has been well documented.16 In many populations, there is a 2- to 5-fold greater prevalence of HCC in men.29 In contrast, across Central American countries where liver cancer rates are the highest in the Western Hemisphere, the incidence in men and women is almost equivalent.29,30 Since an increasing proportion of the US population is projected to have either immigrated from or be of, Central American ancestry, identifying their unique risk factors is crucial for early detection and prevention strategies, benefiting these high-risk communities and the US health care system.

This review examines the established and emerging environmental risk factors for HCC, with a focus on agents classified as group 1 human carcinogens by the International Agency for Research on Cancer (IARC). We define environmental exposures broadly to encompass all nongenetic factors that contribute to hepatocarcinogenesis. These include well-documented chemical carcinogens such as aflatoxins, alcohol, and tobacco constituents, as well as physical agents like particulate matter and air pollution. Additionally, we consider biological factors such as HBV, HCV, and metabolic conditions, including diabetes and MASLD, which, while not traditionally viewed as environmental, are significantly influenced by exogenous factors and lifestyle choices (Figure 1). We explore how established and emerging environmental factors interact with changing liver pathology, potentially increasing the overall risk of disease.

FIGURE 1.

FIGURE 1

Integrated model of major risk factors for liver cancer. Abbreviations: AFB1, aflatoxin; MASLD, metabolic dysfunction–associated steatotic liver disease; MASH, metabolic dysfunction–associated steatohepatitis; T2DM, type 2 diabetes mellitus.

AFLATOXIN

Discovery and relation to liver cancer

Aflatoxins, mycotoxins produced by fungi of the Aspergillus species, contaminate a variety of human foodstuffs, most notably, maize, ground nuts, and tree nuts.31 Although most extensively investigated in Africa and Asia, AFB1 contamination occurs in many locations around the world, especially in regions with warm, humid environments such as Central America.32 Animal studies first demonstrated AFB1’s hepatotoxicity and lethality at high doses. Human research lagged due to insufficient data on aflatoxin metabolism and susceptibility factors. The heterogeneous nature of food contamination made dietary assessments inadequate for establishing an AFB1-HCC link. However, the development of aflatoxin-specific biomarkers eventually enabled studies to provide convincing evidence of this association.32 Using AFB1 biomarkers, 2 cohort studies examined urinary levels of AFB1 and serum levels of HBV among a cohort of men in Shanghai, China. The study found that the risk of liver cancer was 3.4-fold higher among the men who tested positive for AFB1, 7-fold higher among the men who tested positive for HBV, and 59-fold higher among the men who tested positive for both AFB1 and HBV.33 Several years later, a subsequent cohort study from Taiwan confirmed the results.34 As was demonstrated by the studies in China and Taiwan, AFB1 is particularly carcinogenic when it co-occurs with chronic HBV infection, as the combination of the 2 factors has a synergistic effect on HCC risk.33,34 Based on the evidence presented by biomarker-based studies, an IARC expert working group concluded that there was sufficient evidence to classify aflatoxin as a group 1 human carcinogen.35 A 2012 meta-analysis estimated that AFB1 alone increased HCC risk by 6-fold, HBV alone by 11-fold, and the 2 factors together by 54-fold.36

Studies of HCC tumors identified a specific AFB1 exposure marker: a characteristic mutation in codon 249 of the TP53 tumor suppressor gene. High frequencies of the signature TP53 guanine to thymine transversion in codon 249 were observed in tumors from aflatoxin-exposed populations, but absent in HCCs from areas with little aflatoxin exposure.37 Notably, African immigrants to France show a decreasing likelihood of displaying the AFB1 signature in their tumors the longer they reside in France.38 Experimental models further confirm this link, demonstrating consistent mutational signature in liver tumors with aflatoxin exposure.39 In addition, whole genome sequencing of human HCC has identified a mutational signature of aflatoxin that correlates with the duration of time since AFB1 exposure.39

Beyond numerous studies linking AFB1 exposure to an increased risk of HCC, a natural experiment in the 1980s demonstrated that removing AFB1 from the environment reduces this risk. When the government facilitated a dietary switch from maize to rice in high-HCC regions, daily aflatoxin exposure decreased by nearly 4000% by 2012.40 This led to a 50% reduction in HCC mortality, occurring before HBV vaccination could influence rates. The dietary change accounted for an estimated 65% of the reduced liver cancer mortality, with 83% of this benefit conferred on HBV-infected individuals.41 Globally, the AFB1 population attributable fraction for HCC is estimated at 17%, ranging from 8% in HBV-absent populations to 21% in HBV-present populations.36

Climate change is expected to expand AFB1’s impact. Warmer, more humid conditions favorable for AFB1 contamination are increasing globally, potentially raising its population attributable fraction. In previously low-risk regions like Central America, extreme weather events, increased rainfall, and higher temperatures are likely to promote mold growth and crop contamination.42 It is well documented that extreme weather events can lead to plant-related stress that increases the probability of mold growth on maize and peanuts. As mold spores are ubiquitous in soil, weather events that lead to a breakdown of the normal physical barriers in plants may increase aflatoxin contamination. A Texas study (2004–2014) showed significant increases in aflatoxin detection rates and median aflatoxin-albumin adduct biomarker levels over time. As maize cultivation expands into traditionally colder climates, aflatoxin contamination risks may spread geographically.43 Moreover, rising temperatures favor cyanobacterial blooms, potentially increasing exposure to liver-damaging cyanotoxins. Recent research confirms increased cyanotoxin prevalence due to climate-driven changes, aligning with studies linking environmental toxin exposures to higher liver disease risks. These findings underscore the need for integrated monitoring and response strategies to address these emerging health risks in Central America and beyond.

AIR POLLUTION

While air pollution is well-established as a risk factor for lung cancer, growing evidence suggests associations with other cancers, including liver cancer. The Effects of Low Level of Air Pollution: A Study in Europe collaboration, encompassing 6 European cohorts, has identified links between long-term exposure to air pollutants and increased liver cancer risk.44 Notably, this risk persists even at levels below current European Union air quality standards for nitrogen dioxide and fine particulate matter (PM2.5).44

The Effects of Low Level of Air Pollution: A Study in Europe study revealed statistically significant hazard ratios for various PM2.5 elemental components, with sulfur and vanadium showing the strongest correlations.44 In the United States, an ecological study demonstrated increased HCC incidence with rising ambient PM2.5 levels, though it lacked control for individual-level risk factors.45 A prospective cohort study in Taiwan further corroborated these findings, showing that higher residential ambient PM2.5 levels were associated with increased HCC risk (HR 1.22, 95% CI: 1.02–1.47).46

The mechanistic role of these pollutants appears to be mediated via inflammation and fibrosis.47 This inflammatory pathway may explain the carcinogenic potential of air pollutants in liver tissue. However, the complex nature of environmental exposures and potential confounding factors necessitate further well-designed prospective studies to definitively establish air pollutants as hepatocarcinogens.

Recent research has also begun to explore the potential synergistic effects of air pollution with other known liver cancer risk factors, such as HBV and MASLD.46,48,49 These interactions could amplify the carcinogenic impact of air pollutants on liver tissue. Additionally, emerging evidence suggests that certain populations may be more susceptible to the hepatotoxic effects of air pollution, including those with pre-existing liver conditions or genetic predispositions. With increasing urbanization and climate change impacting air quality, understanding the air pollution-liver cancer relationship is crucial. Future research must focus on specific pollutant mixtures and interventions for high-risk populations to address this emerging public health concern.

ALCOHOL CONSUMPTION

Excessive consumption of alcohol (ie, >45 or 50 g/d) increases the risk of liver cancer, with women showing a higher OR (3.89) than men (1.59).50 While women show higher individual risk, likely due to differences in alcohol metabolism, body composition, and hormonal factors; men face greater overall attributable risk due to their higher prevalence of alcohol consumption.51 A 2016 US study reported that 17% of HCCs among men were alcohol-associated, while only 6.2% of HCC among women were alcohol-associated.1 On the global scale, it has been estimated that 17% of liver cancer is related to alcohol, with a greater percentage among men (22.7%) than women (5.0%).51

The exact mechanism or mechanisms linking alcohol consumption to liver cancer are complex and multifaceted, although alcohol is metabolized to acetaldehyde, which is an established group 1 carcinogen that can directly damage DNA and proteins.52 In addition to the role that acetaldehyde may play, chronic alcohol consumption may (1) increase pro-oxidant states leading to oxidative stress and cellular damage, (2) alter the gut microbiome, resulting in deleterious consequences to the liver, (3) impair immune function, reducing the physiological response to detect and eliminate precancerous cells, and (4) induce chronic inflammation and fibrosis, leading to procarcinogenic environment in the liver.53,54,55,56,57

The impact of alcohol reduction or cessation on liver cancer risk remains understudied, with limited longitudinal data available.58 Current evidence suggests that alcohol abstinence may reduce HCC risk, particularly in patients with early-stage alcohol-associated liver disease. A retrospective study by Ganne-Carrié et al59 found that alcohol abstinence was associated with a decreased HCC incidence in patients with alcohol-associated cirrhosis (HR 0.58, 95% CI: 0.38–0.88). However, the risk reduction magnitude and time frame vary depending on factors such as the degree of pre-existing liver fibrosis, duration of heavy alcohol use, and potential genetic predispositions. For instance, Marot et al60 observed that the beneficial effects of abstinence on HCC risk were most pronounced in patients who had abstained for more than 1.5 years. Future prospective studies are needed to elucidate the molecular mechanisms underlying potential risk reduction, identify relevant biomarkers for monitoring, and establish evidence-based guidelines for alcohol cessation in HCC prevention strategies.

TOBACCO

Tobacco smoking is estimated to increase the risk of liver cancer with an estimated 70% higher risk among smokers compared to nonsmokes.61 Globally, 13% of liver cancers are related to smoking.2 Several mechanisms are hypothesized to explain the liver cancer-smoking association.62 The liver metabolizes a number of tobacco constituents, including 4-aminobiphenyl and polycyclic aromatic hydrocarbons, which are converted into reactive species. Other tobacco constituents, such as nitrosamines and benzene, undergo hepatic activation to procarcinogens by CYP2E1. In addition, tobacco exposure has been associated with immunosuppression and telomere shortening and can lead to the production of proinflammatory cytokines, such as IL-33, IL-1β, and TNFα. Mutational signature studies of liver cancer tumors have also noted the presence of signatures that could be related to tobacco smoking.38

The impact of smoking cessation on liver cancer risk has been investigated, with encouraging results. A US study reported an inverse association between the duration of smoking cessation and HCC risk. Individuals who ceased smoking for more than 30 years exhibited an HCC risk similar to that of never-smokers.63 This suggests a potential reversibility of smoking-induced liver cancer risk, though the time frame for risk reduction may be prolonged.

Future research priorities include identifying the molecular mechanisms of tobacco-induced hepatocarcinogenesis, quantifying synergistic effects between smoking and viral hepatitis or alcohol consumption, and even assessing the hepatotoxic potential of e-cigarettes. Longitudinal studies are needed to determine the optimal duration of smoking cessation for significant HCC risk reduction. Additionally, investigating genetic susceptibility factors may help stratify smokers for targeted HCC prevention strategies.

OCCUPATIONAL CARCINOGENIC HAZARDS TO HUMANS

IARC has convened expert panels to evaluate carcinogenic risk to humans for more than 50 years, and many of these monographs summarize available epidemiologic data linking specific exposures to liver cancer (http://monographs.iarc.fr). While the preceding sections in this review summarize the dominant risk factors for human liver cancer, the following provides specific cases identified through epidemiologic and occupational studies. These include vinyl chloride as a risk factor for hepatic angiosarcoma, various pesticides, and a number of radioactive substances. Collectively, these represent at the present time a small fraction of the risk factors that contribute to liver cancer diagnosis. With the development of better biomarkers and higher throughput analytic methodologies, new causal associations will be characterized over the next decade.

Vinyl chloride Vinyl chloride exposure in occupational settings has been associated with angiosarcomas of the liver.64,65,66 Further, studies have reported a multiplicative interaction between vinyl chloride exposure in the workplace and alcohol consumption in the enhancement of HCC.67 Finally, a synergistic interaction between vinyl chloride workplace exposure and HBV status has been reported in a cohort in Taiwan.68

Arsenic: Chronic arsenic exposure, primarily through contaminated drinking water, has been established as a risk factor for HCC development through epidemiological studies in endemic regions. Studies from Taiwan and Bangladesh have demonstrated dose-dependent relationships between arsenic exposure and liver cancer mortality, with significantly increased risks observed at concentrations exceeding 50 μg/L in drinking water.69,70 Mechanistically, arsenic promotes hepatocarcinogenesis through oxidative stress, DNA damage, and epigenetic modifications, with enhanced risk observed in populations with concurrent HBV infection or heavy alcohol consumption. Recent prospective studies have further elucidated synergistic interactions between arsenic exposure and other environmental toxicants in HCC development.

Pesticide exposures: A number of epidemiologic studies, particularly among agricultural workers and other occupational settings, have reported associations between liver cancer and DDT and it’s most persistent metabolite, dichlorodiphenyldichloroethylene. All 3 studies found significant associations with either DDT or dichlorodiphenyldichloroethylene, suggesting that other organochlorine pesticides might have similar effects.71

EXPOSURES TO RADIOACTIVE SUBSTANCES: PLUTONIUM, THORIUM-232, AND THOROTRAST

There have been a number of reports of both occupational and iatrogenic exposures to specific forms of radiation-causing liver cancers. This has been observed among workers exposed to alpha emitters such as plutonium.72 In these circumstances, the ongoing exposure assessments required for these workers helped to define the dose of radiation, and ongoing health assessments facilitated disease diagnoses. Similar to this exposure and dose situation, the alpha-emitter Thorium-232 that historically was incorporated into an imaging product for the liver was subsequently shown to induce HCC in experimental models and in patients.73,74 These studies illustrate that high-quality exposure and health assessment can identify these etiologies, and in turn, this accelerates preventive interventions.

HEPATITIS B VIRUS

Chronic infection with HBV is associated with more than half (56%) of the HCCs in the world, making it the dominant risk factor.75 The lifetime risk of developing HCC among persons with chronic HBV infections varies between 10% and 25%,21 depending on characteristics of the virus, other concomitant viral infections, demographic characteristics, and the presence of other HCC risk factors, such as aflatoxin B1.76

Approximately 300 million individuals are chronically infected with HBV, with the highest infection rates (>8%) in sub-Saharan Africa and the Western Pacific. Early-life transmission is a key factor in developing chronic infections, particularly in high-prevalence regions. HBV vaccination, available since 1981, offers primary prevention. Taiwan’s pioneering neonatal HBV vaccination program, implemented in 1984, demonstrated a decline in HCC incidence in vaccinated cohorts after 2 decades. As of 2022, 190 WHO member states have introduced neonatal HBV vaccination, although complete coverage remains a challenge in some countries. For individuals with established chronic HBV infections, antiviral treatment reduces risk of HCC, although long-term therapy must be maintained.77,78,79

HEPATITIS C VIRUS

Chronic infection with HCV is associated with 20% of HCCs in the world.75 In the majority (90%) of cases, HCV-associated HCC is preceded by cirrhosis, with the annual incidence of HCC ranging from 0.5% to 10%.19 As cirrhosis is almost invariably present in HCV-associated HCC, hepatocarcinogenesis is likely the result of repetitive damage, which results in regeneration, fibrosis, and cirrhosis.80,81 There is no HCV vaccine at the current time, but curative therapy exists. Direct-acting antiviral therapy, first introduced in 2014, effectively eradicates the virus. HCC risk is reduced by 50-80% among persons who achieve a sustained virologic response.82 However, it’s important to note that HCC risk persists even after viral clearance, particularly in patients with advanced fibrosis or cirrhosis. In the majority (90%) of cases, HCV-associated HCC is preceded by cirrhosis, with the annual incidence of HCC ranging from 1% to 7%, emphasizing the need for continued surveillance in high-risk individuals even after successful HCV treatment.19

TYPE 2 DIABETES

Approximately 7% of liver cancers in the world have been linked to diabetes, which is associated with a 2 to 3-fold increased risk of HCC.2 The risk is significantly greater among men than women and among persons with long-standing and poorly controlled diseases.83 The association between type 2 diabetes and HCC may be due to various mechanisms, many of which are related to insulin resistance and activation of the insulin receptor and IGF 1 pathways.2 Chronic hyperinsulinemia in type 2 diabetes promotes hepatic lipogenesis and inflammation, creating a procarcinogenic environment. Additionally, diabetes-associated oxidative stress and advanced glycation end products contribute to DNA damage and genomic instability in hepatocytes. Recent studies suggest that metformin, a common diabetes medication, may have a protective effect against HCC development, potentially through its anti-inflammatory and antiproliferative properties.84,85

METABOLIC DYSFUNCTION–ASSOCIATED STEATOTIC LIVER DISEASE

MASLD, thought to be the hepatic manifestation of metabolic syndrome, is the most rapidly rising risk factor for HCC in the world. MASLD is defined as hepatic steatosis in the presence of other cardiometabolic risk factors or without discernible secondary causes.86,87 The global prevalence of MASLD is approximately 30%, with the highest prevalence in the world estimated to be in Latin America.12,88,89 HCC risk is significantly higher in persons with MASLD who also have diabetes, obesity, dyslipidemia, and hypertension.90 Diabetes doubles the risk of HCC, and when associated with MASLD, it increases HCC risk nearly fivefold.91 The prevalence of MASLD in persons with type 2 diabetes mellitus is estimated to be 55%–68%, with 37% having metabolic dysfunction–associated steatohepatitis, with 17% demonstrating fibrosis.92

Obesity also significantly impacts HCC risk in patients with MASLD. Body mass index >30 tracks with a 2-fold increase in HCC risk, and body mass index >35 is correlated with a fourfold increase in HCC risk.12,91 In the United States, MASLD-related HCC accounts for 19.2%–35.6% of HCC cases in Medicare patients.91,93 Importantly, MASLD-related HCC was associated with the lowest rates of surveillance when compared to other major etiologies of HCC.93

While MASLD and obesity are strongly associated, “lean” MASLD also occurs in 7%–12% of normal-weight individuals in the United States.94,95 In a recent study by Gobali et al,95 lean MASLD was associated with higher rates of diabetes and insulin resistance, hypertension, and cardiovascular events when compared to lean individuals without MASLD. Interestingly, patients with cirrhosis with lean MASLD and obesity-related MASLD have similar HCC rates, approximately 20%.94,95 The annual incidence of HCC among persons with metabolic dysfunction–associated steatohepatitis cirrhosis ranges from 0.5% to 2.6%.12 The rising prevalence of MASLD, particularly in urban environments, underscores the urgent need for effective prevention, screening, and treatment strategies, especially for high-risk groups such as individuals with diabetes.

CONCLUSION AND PERSPECTIVES FOR FUTURE

The landscape of HCC risk factors is shifting globally. While viral hepatitis prevalence is decreasing, MASLD is emerging as a dominant driver of HCC. Traditional factors like alcohol and aflatoxins remain significant, while air pollution and climate change-induced mycotoxin spread present new challenges. These evolving risk factors intersect with broader societal issues, particularly socioeconomic disparities, creating a complex web of HCC determinants. This disparity underscores the need for targeted interventions in vulnerable populations, as addressing socioeconomic factors is crucial for comprehensive HCC prevention.

Key priorities for prevention should include beyond expanding HBV vaccination and HCV treatment access to implementing effective MASLD screening programs and addressing lifestyle and metabolic factors. For air pollution, prevention strategies should focus on policy-level interventions, such as enforcing stricter emissions standards, promoting clean energy technologies, and improving urban planning to reduce exposure. Individual-level measures like using air purifiers and limiting outdoor activities during high pollution days can also help mitigate risk. Aflatoxin prevention requires a multifaceted approach. Implementing better agricultural practices, such as proper crop rotation and storage techniques, can reduce contamination. Biocontrol methods, like introducing nontoxigenic fungal strains, have shown promise in reducing aflatoxin levels. Additionally, improving food safety regulations and increasing public awareness about proper food storage and handling can further reduce exposure. Table 2 summarizes various categories to effectively mitigate the risk factors that may guide the next steps (Table 2).

TABLE 2.

Comprehensive strategies for reducing HCC environmental and lifestyle risk factors

Risk factor Solution Details
Aflatoxin Agriculture Control crop drying/storage, use biocontrol agents
Diet Promote dietary diversity, educate about moldy food risks
Regulation Enforce aflatoxin limits, adopt aflatoxin-resistant crops
Air pollution Emissions Enforce stricter industrial and vehicle emission controls, promote clean energy technologist including electric vehicles (EVs)
Urban planning Increase green spaces, enforce zoning laws
Health Monitor air quality, educate on exposure reduction
Alcohol Policy Implement minimum unit pricing, restrict availability and advertising
Health care Screen for alcohol use disorders, provide interventions and referrals
Education Raise awareness about safe drinking limits and risks
Tobacco Cessation Offer counseling, nicotine replacement therapy (NRT), pharmacotherapy
Policy Increase taxes, enact smoke-free laws, restrict advertising
Occupational exposure to carcinogenic Enhanced protective equipment Use advanced personal protective equipment (PPE), such as respirators and full-body suits, minimizes exposure to radioactive substances and hazardous chemicals
Engineering controls Implement changes to the workplace, such as installing adequate ventilation systems, containment structures, and shielding barriers, effectively reduces airborne exposure and contamination risks
Type 2 diabetes Prevention Promote healthy diet and physical activity, workplace wellness programs
Screening Conduct regular screenings in high-risk populations for early intervention
Management Improve access to care, encourage medication adherence and lifestyle changes
MASLD Diet Promote Mediterranean diet, reduce processed foods and sugary drinks
Exercise Promote physical activity, improve access to fitness and safe outdoor spaces
Screening Develop improved screening protocols specific to MASLD and obese patients
Treatment Improve access to weight loss interventions, including behavioral therapy and bariatric surgery
Viral hepatitis HBV Expand HBV vaccination, screen and treat chronic infections
HCV Implement widespread HCV screening, improve access to direct-acting antiviral treatments, provide clean needle access
Education Raise awareness about transmission and prevention

Abbreviations: EVs, electric vehicles; MASLD, metabolic dysfunction–associated steatotic liver disease; NRT, nicotine replacement therapy; PPE, personal protective equipment.

Implementing these comprehensive prevention programs presents significant challenges, particularly in resource-limited countries where the burden of HCC is projected to increase. Reducing the global impact of HCC necessitates a coordinated international effort, with high-income countries collaborating closely with lower-income nations to address both environmental risk factors and socioeconomic disparities. By focusing on these diverse risk factors through targeted, evidence-based interventions, the global health community can strive to substantially reduce HCC incidence in the coming decades. While the task ics formidable, the potential to save lives and reduce the societal burden of HCC makes these efforts imperative.

Footnotes

Abbreviations: AFB1, aflatoxin B1; IARC, International Agency for Research on Cancer; MASLD, metabolic dysfunction–associated steatotic liver disease; PM2.5, particulate matter.

Contributor Information

Divya Rayapati, Email: drayapa1@jhmi.edu.

Katherine A. McGlynn, Email: mcglynnk@mail.nih.gov.

John D. Groopman, Email: jgroopm1@jhu.edu.

Amy K. Kim, Email: akim97@jhmi.edu.

CONFLICTS OF INTEREST

The authors have no conflicts to report.

REFERENCES

  • 1.Makarova-Rusher OV, Altekruse SF, McNeel TS, Ulahannan S, Duffy AG, Graubard BI, et al. Population attributable fractions of risk factors for hepatocellular carcinoma in the United States. Cancer. 2016;122:1757–1765. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Baecker A, Liu X, La Vecchia C, Zhang ZF. Worldwide incidence of hepatocellular carcinoma cases attributable to major risk factors. Eur J Cancer Prev. 2018;27:205–212. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Hepatitis. World Health Organization . Accessed June 1, 2024. https://www.who.int/data/gho/data/themes/chronic-viral-hepatitis
  • 4.El-Serag HB. Epidemiology of viral hepatitis and hepatocellular carcinoma. Gastroenterology. 2012;142:1264–1273.e1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Oyebamiji YO, Ajijolakewu KA, Adebayo IA. Worldwide Prevalence of Aflatoxins in Food and Feed. In: Hakeem KR, Oliveira CAF, Ismail A, eds. Aflatoxins in Food: A Recent Perspective. Springer International Publishing; 2021. 41-65. [Google Scholar]
  • 6.Chu YJ, Yang HI, Wu HC, Lee MH, Liu J, Wang LY, et al. Aflatoxin B(1) exposure increases the risk of hepatocellular carcinoma associated with hepatitis C virus infection or alcohol consumption. Eur J Cancer. 2018;94:37–46. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Air Pollution Data Portal . World Health Organization. https://www.who.int/data/gho/data/themes/air-pollution
  • 8.Bryazka D, Reitsma MB, Griswold MG, Abate KH, Abbafati C, Abbasi-Kangevari M, et al. Population-level risks of alcohol consumption by amount, geography, age, sex, and year: A systematic analysis for the Global Burden of Disease Study 2020. Lancet. 2022;400:185–235. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Matsushita H, Takaki A. Alcohol and hepatocellular carcinoma. BMJ Open Gastroenterol. 2019;6:e000260. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Tobacco. World Health Organization . https://www.who.int/health-topics/tobacco
  • 11.Yoo JJ, Park MY, Cho EJ, Yu SJ, Kim SG, Kim YJ, et al. Smoking increases the risk of hepatocellular carcinoma and cardiovascular disease in patients with metabolic-associated fatty liver disease. J Clin Med. 2023;12:3336. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Riazi K, Azhari H, Charette JH, Underwood FE, King JA, Afshar EE, et al. The prevalence and incidence of NAFLD worldwide: A systematic review and meta-analysis. Lancet Gastroenterol Hepatol. 2022;7:851–861. [DOI] [PubMed] [Google Scholar]
  • 13.Xie X, Zheng M, Guo W, Zhou Y, Xiang Z, Li Y, et al. Correlation analysis of metabolic characteristics and the risk of metabolic-associated fatty liver disease - related hepatocellular carcinoma. Sci Rep. 2022;12:13969. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Facts & Figures. International Diabetes Federation. https://idf.org/about-diabetes/diabetes-facts-figures/
  • 15.Doycheva I, Zhang T, Amjad W, Thuluvath PJ. Diabetes and hepatocellular carcinoma: Incidence trends and impact of liver disease etiology. J Clin Exp Hepatol. 2020;10:296–303. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A, et al. Global Cancer Statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2021;71:209–249. [DOI] [PubMed] [Google Scholar]
  • 17.Wild CP, Miller JD, Groopman JD. Mycotoxin Control in Low- and Middle-Income Countries. 2015. (IARC Working Group Reports; 9) . http://www.iarc.fr/en/publications/pdfs-online/wrk/wrk9/index.php [PubMed]
  • 18.Ferlay J, Soerjomataram I, Dikshit R, Eser S, Mathers C, Rebelo M, et al. Cancer incidence and mortality worldwide: Sources, methods and major patterns in GLOBOCAN 2012. Int J Cancer. 2015;136:E359–E386. [DOI] [PubMed] [Google Scholar]
  • 19.El-Serag HB, Kanwal F. Epidemiology of hepatocellular carcinoma in the United States: Where are we? Where do we go? Hepatology. 2014;60:1767–1775. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Petrick JL, Kelly SP, Altekruse SF, McGlynn KA, Rosenberg PS. Future of hepatocellular carcinoma incidence in the United States Forecast through 2030. J Clin Oncol. 2016;34:1787–1794. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.McGlynn KA, Petrick JL, London WT. Global epidemiology of hepatocellular carcinoma: An emphasis on demographic and regional variability. Clin Liver Dis. 2015;19:223–238. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Groopman JD, Smith JW, Rivera-Andrade A, Alvarez CS, Kroker-Lobos MF, Egner PA, et al. Aflatoxin and the etiology of liver cancer and Its Implications for Guatemala. World Mycotoxin J. 2021;14:305–317. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Pinheiro PS, Callahan KE, Kobetz EN. Disaggregated hispanic groups and cancer: Importance, methodology, and current knowledge. In: Ramirez AG, Trapido EJ, eds. Advancing the Science of Cancer in Latinos. Springer Publishing; 2020:17–34. [PubMed] [Google Scholar]
  • 24.Zamora SM, Pinheiro PS, Gomez SL, Hastings KG, Palaniappan LP, Hu J, et al. Disaggregating Hispanic American Cancer Mortality Burden by Detailed Ethnicity. Cancer Epidemiol Biomarkers Prev. 2019;28:1353–1363. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Pinheiro PS, Callahan KE, Jones PD, Morris C, Ransdell JM, Kwon D, et al. Liver cancer: A leading cause of cancer death in the United States and the role of the 1945-1965 birth cohort by ethnicity. JHEP Rep. 2019;1:162–169. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Callahan KE, Pinheiro PS. Hepatocellular carcinoma among Hispanics: An understudied public health priority. Clin Gastroenterol Hepatol. 2019;17:1649–1650. [DOI] [PubMed] [Google Scholar]
  • 27. Cancer Disparities Progress Report. 2022:200. Accessed June 1, 2024. http://www.CancerDisparitiesProgressReport.org/ [DOI] [PubMed]
  • 28.Rumgay H, Arnold M, Ferlay J, Lesi O, Cabasag CJ, Vignat J, et al. Global burden of primary liver cancer in 2020 and predictions to 2040. J Hepatol. 2022;77:1598–1606. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Bray F, Laversanne M, Sung H, Ferlay J, Siegel RL, Soerjomataram I, et al. Global cancer statistics 2022: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2024;74:229–263. [DOI] [PubMed] [Google Scholar]
  • 30.Piñeros M, Frech S, Frazier L, Laversanne M, Barnoya J, Garrido C, et al. Advancing reliable data for cancer control in the Central America four region. J Glob Oncol. 2018;4:1–11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Eaton DL, Groopman JD. The Toxicology of Aflatoxins: Human Health, Veterinary, and Agricultural Significance. Academic Press, Inc; 1994. [Google Scholar]
  • 32.Kensler TW, Roebuck BD, Wogan GN, Groopman JD. Aflatoxin: A 50-year odyssey of mechanistic and translational toxicology. Toxicol Sci. 2011;120(Suppl 1):S28–S48. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Ross RK, Yu MC, Henderson BE, Yuan JM, Qian GS, Tu JT, et al. Urinary aflatoxin biomarkers and Risk of hepatocellular carcinoma. Lancet. 1992;339:943–946. [DOI] [PubMed] [Google Scholar]
  • 34.Wang LY, Hatch M, Chen CJ, Levin B, You SL, Lu SN, et al. Aflatoxin exposure and risk of hepatocellular carcinoma in Taiwan. Research Support, Non-U.S. Gov't Research Support, U.S. Gov't, P.H.S. Int Cancer. 1996;67:620–625. [DOI] [PubMed] [Google Scholar]
  • 35.IARC monographs . Some naturally occurring substances: Food items and constitutents, heterocyclic aromatic amines and mycotoxins. IARC Monogr Eval Carcinog Risk Hum. 1993;56. [Google Scholar]
  • 36.Liu Y, Chang CC, Marsh GM, Wu F. Population attributable risk of aflatoxin-related liver cancer: Systematic review and meta-analysis. Meta-Analysis Research Support, N.I.H., Extramural Review. Eur J Cancer. 2012;48:2125–2136. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Gouas D, Shi H, Hainaut P. The aflatoxin-induced TP53 mutation at codon 249 (R249S): Biomarker of exposure, early detection and target for therapy. Research Support, Non-U.S. Gov't Review. Cancer Lett. 2009;286:29–37. [DOI] [PubMed] [Google Scholar]
  • 38.Letouzé E, Shinde J, Renault V, Couchy G, Blanc JF, Tubacher E, et al. Mutational signatures reveal the dynamic interplay of risk factors and cellular processes during liver tumorigenesis. Nat Commun. 2017;8:1315. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Fedeles BI, Chawanthayatham S, Croy RG, Wogan GN, Essigmann JM. Early detection of the aflatoxin B1 mutational fingerprint: A diagnostic tool for liver cancer. Mol Cell Oncol. 2017;4:e1329693. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Chen JG, Egner PA, Ng D, Jacobson LP, Muñoz A, Zhu YR, et al. Reduced aflatoxin exposure presages decline in liver cancer mortality in an endemic region of China. Cancer Prev Res (Phila). 2013;6:1038–1045. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Chen J, Zhu J, Wang G, Groopman JD, Kensler TW. Qidong: A crucible for studies on liver cancer etiology and prevention. Cancer Biol Med. 2019;16:24–37. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Romanello M, McGushin A, Di Napoli C, Drummond P, Hughes N, Jamart L, et al. The 2021 report of the Lancet Countdown on health and climate change: Code red for a healthy future. Lancet. 2021;398:1619–1662. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Xue KS, Tang L, Shen CL, Pollock BH, Guerra F, Phillips TD, et al. Increase in aflatoxin exposure in two populations residing in East and West Texas, United States. Int J Hyg Environ Health. 2021;231:113662. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.So R, Chen J, Mehta AJ, Liu S, Strak M, Wolf K, et al. Long-term exposure to air pollution and liver cancer incidence in six European cohorts. Int J Cancer. 2021;149:1887–1897. [DOI] [PubMed] [Google Scholar]
  • 45.VoPham T, Bertrand KA, Tamimi RM, Laden F, Hart JE. Ambient PM(2.5) air pollution exposure and hepatocellular carcinoma incidence in the United States. Cancer Causes Control. 2018;29:563–572. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Pan WC, Wu CD, Chen MJ, Huang YT, Chen CJ, Su HJ, et al. Fine particle pollution, alanine transaminase, and liver cancer: A Taiwanese Prospective Cohort Study (REVEAL-HBV). J Natl Cancer Inst. 2016;108. doi: 10.1093/jnci/djv341 [DOI] [PubMed] [Google Scholar]
  • 47.VoPham T, Jones RR. State of the science on outdoor air pollution exposure and liver cancer risk. Environ Adv. 2023;11. doi: 10.1016/j.envadv.2023.100354 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Jang TY, Ho CC, Wu CD, Dai CY, Chen PC. Air pollution as a potential risk factor for hepatocellular carcinoma in Taiwanese patients after adjusting for chronic viral hepatitis. J Chin Med Assoc. 2024;87:287–291. [DOI] [PubMed] [Google Scholar]
  • 49.Barouki R, Samson M, Blanc EB, Colombo M, Zucman-Rossi J, Lazaridis KN, et al. The exposome and liver disease—How environmental factors affect liver health. J Hepatol. 2023;79:492–505. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Bagnardi V, Rota M, Botteri E, Tramacere I, Islami F, Fedirko V, et al. Alcohol consumption and site-specific cancer risk: A comprehensive dose-response meta-analysis. Br J Cancer. 2015;112:580–593. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Rumgay H, Shield K, Charvat H, Ferrari P, Sornpaisarn B, Obot I, et al. Global burden of cancer in 2020 attributable to alcohol consumption: A population-based study. Lancet Oncol. 2021;22:1071–1080. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.IARC monographs. A revew of human carcinogens: Personal habits and indoor combusion. IARC Monogr Eval Carcinog Risks Hum. 2012;100E:1–538. [PMC free article] [PubMed] [Google Scholar]
  • 53.Sureshchandra S, Raus A, Jankeel A, Ligh BJK, Walter NAR, Newman N, et al. Dose-dependent effects of chronic alcohol drinking on peripheral immune responses. Sci Rep. 2019;9:7847. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Yang XH, Yamagiwa S, Ichida T, Matsuda Y, Sugahara S, Watanabe H, et al. Increase of CD4+ CD25+ regulatory T-cells in the liver of patients with hepatocellular carcinoma. J Hepatol. 2006;45:254–262. [DOI] [PubMed] [Google Scholar]
  • 55.Åberg F, Helenius-Hietala J, Puukka P, Färkkilä M, Jula A. Interaction between alcohol consumption and metabolic syndrome in predicting severe liver disease in the general population. Hepatology. 2018;67:2141–2149. [DOI] [PubMed] [Google Scholar]
  • 56.Rumgay H, Murphy N, Ferrari P, Soerjomataram I. Alcohol and cancer: Epidemiology and biological mechanisms. Nutrients. 2021;13:3173. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Yu LX, Schwabe RF. The gut microbiome and liver cancer: Mechanisms and clinical translation. Nat Rev Gastroenterol Hepatol. 2017;14:527–539. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Gapstur SM, Bouvard V, Nethan ST, Freudenheim JL, Abnet CC, English DR, et al. The IARC perspective on alcohol reduction or cessation and cancer risk. N Engl J Med. 2023;389:2486–2494. [DOI] [PubMed] [Google Scholar]
  • 59.Ganne-Carrié N, Chaffaut C, Bourcier V, Archambeaud I, Perarnau JM, Oberti F, et al. Estimate of hepatocellular carcinoma incidence in patients with alcoholic cirrhosis. J Hepatol. 2018;69:1274–1283. [DOI] [PubMed] [Google Scholar]
  • 60.Marot A, Henrion J, Knebel JF, Moreno C, Deltenre P. Alcoholic liver disease confers a worse prognosis than HCV infection and non-alcoholic fatty liver disease among patients with cirrhosis: An observational study. PLoS One. 2017;12:e0186715. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.The health consequences of smoking - 50 years of progress: A report of the Surgeon General. 2014.
  • 62.Marti-Aguado D, Clemente-Sanchez A, Bataller R. Cigarette smoking and liver diseases. J Hepatol. 2022;77:191–205. [DOI] [PubMed] [Google Scholar]
  • 63.Petrick JL, Campbell PT, Koshiol J, Thistle JE, Andreotti G, Beane-Freeman LE, et al. Tobacco, alcohol use and risk of hepatocellular carcinoma and intrahepatic cholangiocarcinoma: The Liver Cancer Pooling Project. Br J Cancer. 2018;118:1005–1012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Falk H, Creech JL, Jr, Heath CW, Jr, Johnson MN, Key MM. Hepatic disease among workers at a vinyl chloride polymerization plant. JAMA. 1974;230:59–63. [PubMed] [Google Scholar]
  • 65.Dragani TA, Zocchetti C. Occupational exposure to vinyl chloride and risk of hepatocellular carcinoma. Cancer Causes Control. 2008;19:1193–1200. [DOI] [PubMed] [Google Scholar]
  • 66.Forman D, Bennett B, Stafford J, Doll R. Exposure to vinyl chloride and angiosarcoma of the liver: A report of the register of cases. Br J Ind Med. 1985;42:750–753. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Mastrangelo G, Fedeli U, Fadda E, Valentini F, Agnesi R, Magarotto G, et al. Increased risk of hepatocellular carcinoma and liver cirrhosis in vinyl chloride workers: Synergistic effect of occupational exposure with alcohol intake. Environ Health Perspect. 2004;112:1188–1192. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Wong RH, Chen PC, Wang JD, Du CL, Cheng TJ. Interaction of vinyl chloride monomer exposure and hepatitis B viral infection on liver cancer. JOccMed. 2003;45:379–383. [DOI] [PubMed] [Google Scholar]
  • 69.Lin HJ, Sung TI, Chen CY, Guo HR. Arsenic levels in drinking water and mortality of liver cancer in Taiwan. J Hazard Mater. 2013;262:1132–1138. [DOI] [PubMed] [Google Scholar]
  • 70.Chen Y, Ahsan H. Cancer burden from arsenic in drinking water in Bangladesh. Am J Public Health. 2004;94:741–744. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.VoPham T, Bertrand KA, Hart JE, Laden F, Brooks MM, Yuan JM, et al. Pesticide exposure and liver cancer: A review. Cancer Causes Control. 2017;28:177–190. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Goerlitz DS, Blancato J, Ramesh A, Islam M, Graham GT, Revina V, et al. Somatic mutation signatures in primary liver tumors of workers exposed to ionizing radiation. Sci Rep. 2019;9:18199. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Ito Y, Kojiro M, Nakashima T, Mori T. Pathomorphologic characteristics of 102 cases of thorotrast-related hepatocellular carcinoma, cholangiocarcinoma, and hepatic angiosarcoma. Cancer. 1988;62:1153–1162. [DOI] [PubMed] [Google Scholar]
  • 74.H.IARC Working Group on the Evaluation of Carcinogenic Risks to . Radiation. IARC Monogr Eval Carcinog Risks Hum. 2012;100(Pt D):7–303. [PMC free article] [PubMed] [Google Scholar]
  • 75.Maucort-Boulch D, de Martel C, Franceschi S, Plummer M. Fraction and incidence of liver cancer attributable to hepatitis B and C viruses worldwide. Int J Cancer. 2018;142:2471–2477. [DOI] [PubMed] [Google Scholar]
  • 76.El-Serag HB. Epidemiology of viral hepatitis and hepatocellular carcinoma. Research Support, N.I.H., Extramural Research Support, Non-U.S. Gov't Review. Gastroenterology. 2012;142:1264–1273. e1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Singal AG, El-Serag HB. Hepatocellular carcinoma from epidemiology to prevention: Translating knowledge into practice. Clin Gastroenterol Hepatol. 2015;13:2140–2151. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Hsu YC, Wu CY, Lane HY, Chang CY, Tai CM, Tseng CH, et al. Determinants of hepatocellular carcinoma in cirrhotic patients treated with nucleos(t)ide analogues for chronic hepatitis B. J Antimicrob Chemother. 2014;69:1920–1927. [DOI] [PubMed] [Google Scholar]
  • 79.Jeng WJ, Lok ASF. What will it take to cure hepatitis B? Hepatol Commun. 2023;7:e0084. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.De Mitri MS, Pisi E, Poussin K, Paterlini P, Bréchot C, Baccarini P, et al. HCV-associated liver cancer without cirrhosis. Lancet. 1995;345:413–415. [DOI] [PubMed] [Google Scholar]
  • 81.Global change in hepatitis C virus prevalence and cascade of care between 2015 and 2020: a modelling study . Polaris Observatory HCV Collaborators. Lancet Gastroenterol Hepatol. 2022;7:396–415. [DOI] [PubMed] [Google Scholar]
  • 82.Waziry R, Hajarizadeh B, Grebely J, Amin J, Law M, Danta M, et al. Hepatocellular carcinoma risk following direct-acting antiviral HCV therapy: A systematic review, meta-analyses, and meta-regression. J Hepatol. 2017;67:1204–1212. [DOI] [PubMed] [Google Scholar]
  • 83.Ohkuma T, Peters SAE, Woodward M. Sex differences in the association between diabetes and cancer: A systematic review and meta-analysis of 121 cohorts including 20 million individuals and one million events. Diabetologia. 2018;61:2140–2154. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Lai SW, Chen PC, Liao KF, Muo CH, Lin CC, Sung FC. Risk of hepatocellular carcinoma in diabetic patients and risk reduction associated with anti-diabetic therapy: A population-based cohort study. Am J Gastroenterol. 2012;107:46–52. [DOI] [PubMed] [Google Scholar]
  • 85.Ruiter R, Visser LE, van Herk-Sukel MPP, Coebergh JWW, Haak HR, Geelhoed-Duijvestijn PH, et al. Lower risk of cancer in patients on metformin in comparison with those on sulfonylurea derivatives: Results from a large population-based follow-up study. Diabetes Care. 2012;35:119–124. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.New MASLD Nomenclature. https://www.aasld.org/new-masld-nomenclature
  • 87.Chan WK, Chuah KH, Rajaram RB, Lim LL, Ratnasingam J, Vethakkan SR. Metabolic dysfunction-associated steatotic liver disease (MASLD): A state-of-the-art review. J Obes Metab Syndr. 2023;32:197–213. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Alexander M, Loomis AK, Fairburn-Beech J, van der Lei J, Duarte-Salles T, Prieto-Alhambra D, et al. Real-world data reveal a diagnostic gap in non-alcoholic fatty liver disease. BMC Med. 2018;16:130. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Younossi ZM, Golabi P, Paik JM, Henry A, Van Dongen C, Henry L. The global epidemiology of nonalcoholic fatty liver disease (NAFLD) and nonalcoholic steatohepatitis (NASH): A systematic review. Hepatology. 2023;77:1335–1347. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Huang DQ, El-Serag HB, Loomba R. Global epidemiology of NAFLD-related HCC: trends, predictions, risk factors and prevention. Nat Rev Gastroenterol Hepatol. 2021;18:223–238. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Younossi ZM, Henry L. Epidemiology of non-alcoholic fatty liver disease and hepatocellular carcinoma. JHEP Rep. 2021;3:100305. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Younossi ZM. Non-alcoholic fatty liver disease—A global public health perspective. J Hepatol. 2019;70:531–544. [DOI] [PubMed] [Google Scholar]
  • 93.Karim MA, Singal AG, Kum HC, Lee YT, Park S, Rich NE, et al. Clinical Characteristics and outcomes of nonalcoholic fatty liver disease-associated hepatocellular carcinoma in the United States. Clin Gastroenterol Hepatol. 2023;21:670–680.e18. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Almomani A, Kumar P, Onwuzo S, Boustany A, Krishtopaytis E, Hitawala A, et al. Epidemiology and prevalence of lean nonalcoholic fatty liver disease and associated cirrhosis, hepatocellular carcinoma, and cardiovascular outcomes in the United States: A population-based study and review of literature. J Gastroenterol Hepatol. 2023;38:269–273. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Golabi P, Paik J, Fukui N, Locklear CT, de Avilla L, Younossi ZM. Patients With Lean Nonalcoholic Fatty Liver Disease Are Metabolically Abnormal and Have a Higher Risk for Mortality. Clin Diabetes. 2019;37:65–72. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Hepatology Communications are provided here courtesy of Wolters Kluwer Health

RESOURCES