Download PDF
Review  |  Open Access  |  22 Sep 2024

Chemoprevention of hepatocellular carcinoma associated with metabolic dysfunction-associated steatotic liver disease: an updated review

Views: 89 |  Downloads: 20 |  Cited:  0
Hepatoma Res 2024;10:37.
10.20517/2394-5079.2024.81 |  © The Author(s) 2024.
Author Information
Article Notes
Cite This Article

Abstract

With the predicted rise in metabolic dysfunction-associated steatotic liver disease (MASLD) prevalence over the next decade, strategies to prevent hepatocellular carcinoma (HCC), which is the third most common cause of cancer-related death, are paramount. In this narrative review, we present recent clinical and translational studies from 2020-2024, providing an updated overview of the literature on chemoprevention of HCC associated with MASLD. We specifically focus on statins, aspirin, metformin, and newer diabetes medications. These agents target specific steps in the development of HCC in MASLD, including steatosis resulting in oxidative stress, inflammation, and eventually fibrosis. All offer promising avenues for HCC chemoprevention, although statins have the strongest data at present. Further ongoing prospective studies are needed.

Keywords

MASLD, statins, aspirin, semaglutide, liver-related events

INTRODUCTION

Liver cancer is the third leading cause of cancer-related deaths globally, with hepatocellular carcinoma (HCC) representing 77% of all primary liver cancers in the United States (US)[1]. Unfortunately, the prognosis of HCC is extremely poor. According to the National Cancer Institute’s Surveillance, Epidemiology and End Results (SEER) database, HCC has a 5-year survival rate in the US of 19.6%, dropping as low as 2.5% in metastatic disease[2]. HCC arises in patients with chronic liver disease of multiple etiologies, including viral hepatitis, alcohol-associated liver diseases, genetic and autoimmune liver diseases. Exposure to environmental agents such as aflatoxins and tobacco contributes in some patients. This review focuses on metabolic dysfunction-associated steatotic liver disease (MASLD), formally known as non-alcoholic fatty liver disease (NAFLD). Our ultimate goal is to identify the most effective strategies for preventing MASLD-associated HCC. Consequently, articles exclusively examining alcohol-associated liver disease or viral hepatitis are not covered. Moving forward, we reference literature using the updated terminology MASLD instead of NAFLD and metabolic dysfunction-associated steatohepatitis (MASH) instead of non-alcoholic steatohepatitis (NASH).

MASLD affects approximately 30% of the global population and is now one of the leading causes of chronic liver disease worldwide[3]. The prevalence of MASLD is forecasted to increase by 21% in the US by 2030. This steep increase in cases parallels the rapid rise in the worldwide incidence of obesity and diabetes[4]. The global prevalence of MASLD has risen from 25% in 2006 to 38% in 2019, resulting in an increase of 50.4% by 2023[5]. Unsurprisingly, the incidence of MASLD-associated HCC is rising significantly. It is projected that MASLD-associated HCC will increase by 137% in the US between 2015 and 2030[6]. Given the striking increase in MASLD-associated HCC cases, it is imperative to focus on preventative measures to address this growing public health concern.

Mechanisms of MASLD-associated HCC

Understanding the cellular mechanisms of carcinogenesis in MASLD-associated HCC is crucial to isolate effective chemoprevention. MASLD is characterized by the abnormal accumulation of fat, primarily triglycerides, in hepatocytes. MASLD encompasses a spectrum of liver diseases that can ultimately progress to HCC.

The progression begins with simple hepatic steatosis, in which fat accumulates in the liver. If the fat results in oxidative stress, this can then advance to MASH, formerly known as NASH, which is characterized by inflammation, hepatocyte injury, and liver fibrosis[7]. Approximately 20% of patients diagnosed with simple steatosis progress to MASH, and of these, 2.6% may further progress to HCC[8]. Complicating the picture, MASLD is not a linear disease trajectory; the rates of progression vary significantly, and disease regression is possible under certain circumstances.

Recent studies have extensively investigated the pathogenesis and development of HCC. There is a proposed “two-hit hypothesis” for MASLD progression to MASH, then HCC. The first hit involves the sensitization of hepatocytes to inflammation and resistance to insulin, while the second hit involves increased inflammation resulting in fibrosis [Figure 1][9]. Oxidative stress in the liver may produce DNA damage and trigger a wound healing response, both of which contribute to hepatocarcinogenesis[10].

Chemoprevention of hepatocellular carcinoma associated with metabolic dysfunction-associated steatotic liver disease: an updated review

Figure 1. MASLD-associated HCC. Illustrating the pathogenesis of HCC, first with cardiometabolic risk factors contributing to the formation of steatosis. Then, chronic inflammation and fibrosis ultimately leading to HCC. Created with BioRender.com. TG: Triglycerides; HDL: high-density lipoprotein; BMI: body mass index; MASLD: metabolic dysfunction-associated steatotic liver disease; HCC: hepatocellular carcinoma.

Chronic liver disease prevention and cure

We briefly mention HCC chemoprevention specific to viral hepatitis, given the potential for this to dramatically influence the global burden of HCC. The incidence of viral hepatitis-associated HCC has significantly decreased due to the implementation of early screening, universal vaccination programs, and effective antiviral therapies. Since the recommendation for universal newborn vaccination for hepatitis B vaccine in 1991, there has been a marked decline in viral-related HCC, especially in the US[11]. Although there is no approved vaccine for hepatitis C virus (HCV), HCV-associated HCC has decreased with the advent of potent direct-acting antivirals (DAAs), though it has not completely been eliminated. Addressing the obesity and alcohol use disorder epidemics through public health interventions would be expected to dramatically reduce HCC incidence as well.

The importance of fibrosis

Despite the reduction in viral hepatitis cases, the overall incidence of HCC continues to rise, attributable to the increase in MASLD-associated HCC. Fibrosis is a critical factor in liver cancer development, as shown by recent data findings that 80%-90% of HCCs are preceded by cirrhosis[12]. In exploring chemoprevention strategies, a key focus is on agents that actively reduce hepatic fibrosis or promote fibrosis regression. Therefore, as expected, most drugs discussed in this article help reduce liver fibrosis and, in turn, have a potential preventative effect against HCC.

ASPIRIN

Mechanisms

Non-steroidal anti-inflammatory drugs (NSAIDS) such as Aspirin have been extensively studied for their chemoprotective effects against various malignancies, including but not limited to colorectal, prostate, and ovarian cancer[13,14]. Chronic inflammation acts as a catalyst for the development of fibrosis, which is the primary driver of HCC. Moreover, data in animal models suggest that platelets may facilitate the accumulation of T lymphocytes in the liver in acute viral hepatitis, thereby contributing to inflammation and potentially fibrosis in the liver[15]. Consequently, both aspirin’s anti-inflammatory and anti-platelet properties may contribute to its ability to protect against the development of HCC. Specifically, in patients with MASLD, there is enhanced intrahepatic prostaglandin synthase-2 (COX-2) and prostaglandin E2. This promotes lipid droplet formation and further activates hepatic stellate cells, leading to fibrosis[16]. One proposed mechanism by which aspirin prevents fibrosis is through its antagonism and irreversible inhibition of proinflammatory COX-2 isozymes[16]. Studies have shown that COX-2 overexpression is present in many patients with HCC, suggesting its causative role in hepatocarcinogenesis[17]. One study demonstrated that enhancing COX-2 expression in mice was sufficient to induce HCC[18]. Conversely, COX-2 expression is not detected in normal liver tissue without chronic inflammatory diseases[19]. Furthermore, COX-2 single nucleotide polymorphisms (SNPs) have been associated with the presence of HCC in humans[20].

Literature review of aspirin chemoprevention

In alignment with previous literature, recent studies continue to affirm that aspirin is an effective chemoprevention for HCC. A 2023 meta-analysis involving over 2.2 million patients demonstrated that aspirin use was associated with a 30% reduction in the risk of developing HCC[21]. This protective effect was observed with aspirin use in both patients with cirrhosis (HR 0.78; 95%CI: 0.69-0.88) and those without cirrhosis (HR 0.86; 95%CI: 0.78-0.94)[21]. Yan et al. reported a reduced risk of HCC with daily aspirin use in their meta-analysis (HR 0.64; 95%CI: 0.56-0.75), with the most significant benefit observed in patients with cirrhosis (HR 0.60; 95%CI: 0.45-0.81)[22]. Additionally, a 2021 meta-analysis by Tan et al. found that aspirin use not only reduced the incidence of HCC (HR 0.51; 95%CI: 0.36-0.72), but also improved liver-related mortality (OR 0.32; 95%CI: 0.15-0.70)[23]. Another proposed mechanism by which aspirin may act against hepatocarcinogenesis is through the reduction and prevention of hepatic fat accumulation. In a 2024 randomized control trial, Simon et al. demonstrated that six months of daily aspirin use led to a significant reduction in hepatic fat quantity in MASLD patients. The study reported a mean absolute change in hepatic fat content of -6.6% in the aspirin group compared to 3.6% in the placebo group, resulting in a mean difference of -10.2% (95%CI: -27.7% to -2.6%)[24]. Overall, these findings underscore the potential and promising use of aspirin as a valuable agent in the chemoprevention of HCC.

Dose- and time-dependent aspirin use

A dose- and time-dependent risk reduction in HCC has been observed, with one study showing the greatest benefits seen in high-dose aspirin (100 mg/day) used for more than three years[22]. Similarly, Ma et al. found a higher dose response with the risk of HCC decreasing by 10% for each 50 mg/day increment in aspirin use and reduction by 6% with each additional year of aspirin exposure[25]. These findings are consistent with our current understanding that while low-dose aspirin (75-100 mg) is sufficient to irreversibly inhibit COX-1, higher doses (> 100 mg) are required to completely inhibit COX-2, thereby exerting optimal anti-inflammatory effects[26-28]. Although low-dose aspirin does not act through the conventional COX pathway, it exhibits anti-inflammatory properties by reducing the accumulation of polymorphonuclear leukocytes and macrophages[29] which are implicated in the development of steatohepatitis.

Conversely, Abdelmalak et al. found that aspirin reduced the risk of HCC by approximately 30% (HR 0.70, 95%CI: 0.60-0.81). However, only low-dose aspirin (< 163 mg/day) led to significant HCC risk reduction (HR 0.39; 95%CI: 0.17-0.91), as opposed to high-dose (HR 0.67; 95%CI: 0.42-1.08)[30]. Additionally, a 2022 meta-analysis by Wang et al. reported a significant inverse association between aspirin dose and liver cancer risk, with effective doses hitting a limit up to ~100 mg/day; doses higher than this threshold did not show a significant impact on reducing HCC incidence[31]. These findings highlight the complexity behind using aspirin for HCC chemoprevention and the mixed results of prior studies. There is also a reasonable concern regarding the risk of gastrointestinal bleeding associated with the use of higher doses of aspirin. Overall, while both high- and low-dose aspirin may offer protective benefits, confirming this effect with prospective studies and determining the optimal dosage for maximizing efficacy while minimizing risks require further investigation [Table 1][21-23,25,27,30-38].

Table 1

Summary of articles on aspirin chemoprevention of HCC

StudyYearDesignSample size, nResults (HR/OR/RR, 95%CI)
Wang et al.[21]2023Meta-analysis/systemic review2,217,7120.70 (0.63-0.76)
Yan et al.[22]2022Meta-analysis2,531,7420.64 (0.56-0.75)
Tan et al.[23]2021Meta-analysis/systemic review147,2830.51 (0.36-0.72)
Ma et al.[25]2022Meta-analysis/systemic review3,273,5240.75 (0.71-0.80)
Zhou et al.[27]2022Meta-analysis/systemic review2,781,1000.56 (0.46-0.69)
Abdelmalak et al.[30]2023Meta-analysis/systemic review2,404,8760.70 (0.60-0.81)
Wang et al.[31]2020Systematic review2,604,3190.59 (0.47-0.75)
Lee et al.[32]2023Case control35,8980.57 (0.37-0.87)
Liu et al.[33]2022Meta-analysis/systemic review2,659,6290.53 (0.43-0.65)
Memel et al.[34]2021Meta-analysis/systemic review2,389,0190.61 (0.51-0.73)
Singh et al.[35]2022Retrospective5210.35 (0.12-0.98)
Tan et al.[36]2023Meta-analysis/systemic review71,2110.46 (0.31-0.67)
Wang et al.[37]2021Meta-analysis3,000,000+0.54 (0.44-0.66)
Zeng et al.[38]2022Meta-analysis2,190,2850.48 (0.27-0.87)

STATINS

Statin medications are well-known for their cholesterol-lowering effects; however, they also possess antifibrotic and anti-inflammatory properties that make them valuable as chemopreventive agents against HCC. Statins reduce the expression of transforming growth factor-beta, a potent cytokine that promotes the activation of hepatic stellate cells[39]. The anti-inflammatory effects of statins are evident through their reduction of macrophage activity, cytokine production, and inflammatory markers such as soluble CD40 ligand[40,41]. Additionally, statins activate peroxisome proliferator-activated receptors (PPAR-α, PPAR-γ), which leads to a reduction in inflammation and fibrosis[42]. Given that inflammation precedes fibrosis and hepatocarcinogenesis, inhibiting inflammation is a crucial step in preventing the development of HCC.

Literature review of statin chemoprevention

In a recent meta-analysis with over 1.7 million patients, Zeng et al. found that the use of statins was associated with a significantly reduced overall risk of HCC compared to non-users (HR 0.52; 95%CI: 0.37-0.72). This study also revealed a reduced risk of HCC in MASLD patients using statins (HR 0.68; 95%CI: 0.59-0.77)[38]. Additionally, among 272,431 adults diagnosed with MASLD, Zou et al. found that statin users exhibited a 53% diminished risk of HCC development in contrast to non-users (HR 0.47; 95%CI: 0.36-0.60)[43]. Similar protective effects of statins in preventing HCC were observed in another study focusing on patients with chronic liver disease (OR 0.52; 95%CI: 0.40-0.68)[44]. Sung et al. further highlighted that the overall HCC incidence was 2.8 times higher in the non-statin cohort compared to the statin cohort[45]. Although the above meta-analyses have found a chemopreventive effect of statins, it should be noted that not every individual study found such effects. In fact, multiple large studies of individual patient data from randomized controlled trials (e.g., Matsushita et al. 2010, Emberson et al. 2012) have not found such effects[46,47]. Therefore, ongoing investigation with prospective studies, as discussed below, will be important to confirm these findings.

Hydrophilic vs. lipophilic statin use

Hydrophilic and lipophilic statins appear to have different effects on HCC risk. According to Zeng et al., only lipophilic statins exhibited a preventive effect against HCC (HR 0.46; 95%CI: 0.37-0.57), whereas hydrophilic statins did not (HR 0.48; 95%CI: 0.18-1.27)[38]. Similarly, Wang et al. reported that lipophilic statins could prevent HCC (OR 0.51; 95%CI: 0.40-0.68), while hydrophilic statins could not (OR 0.77; 95%CI: 0.58-1.02)[44]. Facciorusso et al. found that all lipophilic statins tested (atorvastatin, lovastatin, simvastatin) were associated with reduced HCC incidence (HR 0.49; 95%CI: 0.39-0.62), with atorvastatin showing the greatest magnitude of protection (HR 0.43; 95%CI: 0.28-0.65)[48]. When looking exclusively at MASH patients, one retrospective study showed that statin use was linked to a lower risk of developing HCC (HR 0.40; 95%CI: 0.24-0.67)[49]. Specifically, patients using lipophilic statins had a significant HCC risk reduction (adjusted HR 0.31; 95%CI: 0.17-0.56), in contrast to those who used hydrophilic statins (aHR 0.85; 95%CI: 0.42-1.72)[49].

The higher efficacy of lipophilic statins in reducing HCC risk is likely related to the difference in the ease with which they can enter cells. Lipophilic stains enter cells through passive diffusion (because of their ability to cross cell membranes) and are thus widely distributed within tissues, including hepatocytes. In contrast, hydrophilic statins require a specific carrier-mediated mechanism in order to enter the hepatocytes[28]. Lipophilic statins are thought to have more pleiotropic and off-target effects as well. A recent study on MASLD patients found a reduced risk of HCC development with both the use of lipophilic statins (HR 0.49; 0.37-0.65) and hydrophilic statins (HR 0.40; 0.21-0.76)[43].

Future directions

Statins have other beneficial effects, including a reduction in portal hypertension and liver-related events. Multiple prospective studies of statins in patients with cirrhosis are ongoing. The RESCU Trial in the Liver Cirrhosis Network (NCT05832229) will evaluate rosuvastatin 40 mg daily in patients with compensated cirrhosis. The SACRED Trial (NCT03654053) will evaluate simvastatin in patients with cirrhosis followed in the Veterans Affairs Health Care System. The STAT NASH Trial (NCT04679376) is evaluating statins in the treatment of MASH specifically. Hopefully, these will provide valuable insights into the safety, and optimal types and dosing of statins. The challenge in performing clinical trials for chemoprevention is low event rates and the need for long-term follow-up. One way to address this is to use biomarkers as surrogate endpoints. PLS-NAFLD, a blood-based signature developed to predict HCC in patients with MASLD and validated in multiple independent cohorts, is modified by statins and may serve as a surrogate endpoint [Table 2][37,38,43-45,48-59].

Table 2

Summary of articles on statin chemoprevention of HCC

StudyYearDesignSample size, nResults (HR/OR/RR, 95%CI)
Wang et al.[37]2021Meta-analysis1,772,4630.57 (0.49-0.65)
Zeng et al.[38]2022Meta-analysis1,774,4760.52 (0.37-0.72)
Zou et al.[43]2023Retrospective study272,4310.47 (0.36-0.60)
Wang et al.[44]2022Meta-analysis4,963,5180.58 (0.51-0.67)
Sung et al.[45]2022Cohort study1,545,6710.36 (0.35-0.38)
Facciorusso et al.[48]2020Meta-analysis1,925,9640.73 (0.69-0.76)
Pinyopornpanish et al.[49]2021Retrospective study1,0720.40 (0.24-0.67)
Azit et al.[51]2021Matched case-control study4240.37 (0.21-0.65)
Chang et al.[52]2020Systemic review/meta-analysis1,611,5960.54 (0.42-0.66)
German et al.[53]2020Retrospective case-control study1030.20 (0.07-0.60)
Hashemi Rafsanjani et al.[54]2024Systemic review/meta-analysis5,732,9480.56 (0.50-0.63)
Islam et al.[55]2020Meta-analysis59,0730.54 (0.47-0.61)
Khazaaleh et al.[56]2022Systemic review/meta-analysis2,668,4970.57 (0.49-0.67)
Vell et al.[57]2023Cohort study1,785,4910.58 (0.35-0.96)
Wong et al.[58]2021Systemic review/meta-analysis1,742,2600.57 (0.52-0.62)
Zhang et al.[59]2023Systemic review/meta-analysis684,3630.59 (0.39-0.89)

METFORMIN

Type 2 diabetes mellitus (T2DM) is a leading predisposing factor for HCC, with studies indicating that it doubles the risk of developing HCC[60]. This risk then increases tenfold in patients with both cirrhosis and T2DM[61]. The exact mechanism by which T2DM leads to HCC is not fully understood, but it is believed to involve the excessive production of reactive oxygen species, which activate nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) and hepatic stellate cells, thereby promoting hepatocarcinogenesis [Figure 2][62]. A recent study by Ribback et al. showed that ballooned hepatocytes, a distinct diagnostic feature of MASLD, were present in 43.8% of cirrhotic livers[63]. Ballooned hepatocyte formation is attributed to dysregulation of the glycogen metabolism, resulting in highly pronounced glycogenosis, which is often associated with the development of steatosis. Therefore, ballooned hepatocytes may link insulin resistance to MASLD. Additionally, insulin resistance in T2DM leads to elevation in levels of insulin growth factor-1, which stimulates hepatocyte proliferation and also inhibits cellular apoptosis, contributing to fibrosis[62]. This then raises the question of whether antidiabetic drugs such as metformin can help reduce HCC risk.

Chemoprevention of hepatocellular carcinoma associated with metabolic dysfunction-associated steatotic liver disease: an updated review

Figure 2. HCC chemoprevention drug mechanisms. Created with BioRender.com. HCC: Hepatocellular carcinoma.

Metformin is thought to exert anti-cancer effects on the liver by suppressing the mTOR pathway through the activation of AMPK, thereby affecting protein synthesis and growth[64]. It also decreases hepatic triglyceride accumulation, which is especially helpful in MAFLD patients as this assists in the reduction of obesity-induced inflammation[64]. In a mouse model study, metformin intervention inhibited hepatic stellate cell activation, reduced liver fibrosis, decreased lipid accumulation in hepatocytes, and halted the progression to cirrhosis and HCC development[65].

Literature review of metformin chemoprevention

Compared to statin and aspirin, there is less literature on metformin’s effects on HCC prevention. Further, the available literature is mixed with regard to the presence and degree of chemoprevention seen with metformin. In a retrospective study involving 1,061 cirrhotic patients with T2DM, the five-year incidence of HCC was lower in the metformin exposure group, compared to the non-exposure group (HR 0.42; 95%CI: 0.33-0.54) [Table 3][62]. Another study by Li et al. reported a decreased risk of HCC in T2DM patients with daily metformin use (OR/RR 0.59; 95%CI: 0.51-0.68), along with an overall reduction in HCC-related mortality (HR 0.74; 95%CI: 0.66-0.83)[64]. In a large cohort of patients with T2DM and NAFLD, metformin use was associated with a 20% reduction in HCC risk (HR 0.80; 95%CI: 0.93-0.98) compared to insulin use alone which did not have any effect on HCC risk (HR 1.02; 95%CI: 0.85-1.22)[66].

Table 3

Summary of articles on metformin chemoprevention of HCC

StudyYearDesignSample size, nResults
Huynh et al.[68]2023Retrospective study36,626No protective effect with metformin monotherapy
Metformin + SGLT2i: HR 0.43; 95%CI: 0.21-0.88
Kramer et al.[66]2021Retrospective cohort study85,962HR 0.80; 95%CI: 0.93-0.98
Li et al.[64]2022Meta-analysis1,452,265OR/RR 0.59; 95%CI: 0.51-0.68
Tangjarusritaratorn
et al.[62]
2021Retrospective study719HR 0.48; 95%CI: 0.36-0.61
Vilar-Gomez et al.[67]2021Cohort study299aHR 0.78; 95%CI: 0.69-0.96
Zeng et al.[38]2022Meta-analysis125,458No protective effect. HR 0.57; 95%CI: 0.31-1.06

The difference between HCC risk reduction with metformin compared to insulin raises the question of whether improved glycemic control, independent of the specific drug, is what is truly protective against HCC. Indeed, good glycemic control (A1c < 7% for > 80% of the time) was associated with a 32% lower risk of HCC (HR 0.69; 95%CI: 0.60-0.77)[66]. Among T2DM patients with MASH cirrhosis, metformin use was associated with significant HCC risk reduction (aHR 0.78; 95%CI: 0.69-0.96)[67]. Another study of 3,358 patients with T2DM and MASH cirrhosis found that dual therapy with metformin and a sodium-glucose cotransport 2 inhibitor (SGLT2i) significantly reduced HCC risk (HR 0.43; 95%CI: 0.21-0.88), whereas metformin monotherapy did not[68]. While the potential of metformin as a chemopreventive agent against HCC is encouraging, the limited number of studies and variability in their results both indicate a critical need for further research to fully establish its efficacy.

NEWER DIABETES MEDICATIONS

While available data point to the beneficial effects of multiple diabetes medications on MASLD histology and progression, it is unclear whether these have chemopreventive effects on HCC beyond their influence on the natural history of MASLD. Glucagon-like peptide-1 receptor agonists (GLP-1RAs), which increase insulin secretion and slow gastric emptying, have gotten attention in the field of MASLD due to the substantial weight loss they produce, and associated beneficial effects on liver histology. GLP-1RAs including semaglutide, liraglutide, and dulaglutide may be associated with reduced risk of HCC as well. A recent study from 2024 of 1,890,020 patients with T2DM found that GLP-1RAs were associated with a lower risk of HCC compared to insulin (HR 0.20; 95%CI: 0.14-0.31) and sulfonylureas (HR 0.39; 95%CI: 0.21-0.69). There was no statistically significant difference between GLP-1RA and metformin[69]. Another very recent retrospective cohort study from 2024 involving MASLD cirrhosis patients with type 2 diabetes found that GLP-1RAs were associated with a lower risk of liver-related events including decompensation (HR 0.74; 95%CI: 0.61-0.88) and HCC (HR 0.37; 95%CI: 0.20-0.63)[70].

SGLT2is are another newer class of diabetes medications that result in increased glucose excretion in the kidneys. In both human and animal studies, SGLT2is have been associated with reductions in steatosis and fibrosis[71]. Two large claims database studies from Korea, both published in 2024, found associations between SGLT2i use and HCC incidence or survival. In one study, SGLT2i use in patients with T2DM without chronic liver disease (N ~ 4 million) was associated with a lower incidence of HCC compared to the use of dipeptidyl peptidase-4 inhibitors[72]. The other study, which included patients with T2DM and MASLD (N = 201,542), found that among 4,936 patients with both MASLD and chronic viral hepatitis, SGLT2i use was associated with a lower risk of HCC even after matching for propensity scores (adjusted HR 2.32; 95%CI: 1.06-5.06)[73]. Although the authors found that the risk of HCC was also lower among patients with T2DM and MASLD only who took SGLT2i, the association was not statistically significant after matching for propensity scores (HR 0.88; 95%CI: 0.62-1.25).

Studies of the association between GLP-1RA and SGLT2i and HCC are generally limited by a lack of data on the magnitude of associated changes in weight and diabetes control. These agents require further study, including in prospective cohorts with systematic measurement of improvement in metabolic parameters. Overall, whether observed effects are products of improved glycemic control is not known.

CONCLUSION

The cascade of steatosis to oxidative stress to inflammation to fibrosis is key in the development of HCC in patients with MASLD. Aspirin, statins, and metformin all address key parts of this pathway, offering promising avenues for HCC chemoprevention.

Aspirin’s anti-inflammatory and anti-platelet properties help prevent HCC; however, optimal dosing and long-term bleeding risks remain unclear. Statins have demonstrated efficacy in reducing fibrosis and hepatocarcinogenesis, yet further research is needed to determine the most effective dosage and types of statins. Overall, statins have the strongest evidence of the agents we discuss, and momentum is gathering for the use of statins to prevent liver-related complications in patients with MASLD and cirrhosis. Metformin’s role in mitigating insulin resistance and reducing hepatic triglycerides build-up is reassuring, though the current body of research is limited, warranting additional studies. Several key questions remain unanswered: how specifically is each agent mediating reduction in HCC risk? Is combination therapy beneficial? Is all glycemic control or lipid lowering the same, or are the pleiotropic properties of these agents unique?

Future research should focus on both existing and emerging therapies, including newer antidiabetic medications, immunotherapies, various selective COX inhibitors, and antifibrotic therapies. In addition, studies specifically targeting MASLD patients are essential. With numerous clinical trials and studies currently underway, significant advancements in HCC chemoprevention are anticipated. In the next 5-10 years, especially with the projected rise in MASLD prevalence, we expect to have more definitive guidance on effective treatments.

DECLARATIONS

Authors’ contributions

Manuscript drafting: Dickinson A

Manuscript review and critical revision: Dinani A

Conceptualization, supervision, manuscript review and critical revision: Wegermann K

Availability of data and materials

Not applicable.

Financial support and sponsorship

None.

Conflicts of interest

All authors declared that there are no conflicts of interest.

Ethical approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Copyright

© The Author(s) 2024.

REFERENCES

1. Rumgay H, Ferlay J, de Martel C, et al. Global, regional and national burden of primary liver cancer by subtype. Eur J Cancer 2022;161:108-18.

2. Chidambaranathan-Reghupaty S, Fisher PB, Sarkar D. Hepatocellular carcinoma (HCC): epidemiology, etiology and molecular classification. Adv Cancer Res 2021;149:1-61.

3. Miao L, Targher G, Byrne CD, Cao YY, Zheng MH. Current status and future trends of the global burden of MASLD. Trends Endocrinol Metab 2024;35:697-707.

4. Younossi ZM. Non-alcoholic fatty liver disease - a global public health perspective. J Hepatol 2019;70:531-44.

5. 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;77:1335-47.

6. Estes C, Razavi H, Loomba R, Younossi Z, Sanyal AJ. Modeling the epidemic of nonalcoholic fatty liver disease demonstrates an exponential increase in burden of disease. Hepatology 2018;67:123-33.

7. Bessone F, Razori MV, Roma MG. Molecular pathways of nonalcoholic fatty liver disease development and progression. Cell Mol Life Sci 2019;76:99-128.

8. Ascha MS, Hanouneh IA, Lopez R, Tamimi TA, Feldstein AF, Zein NN. The incidence and risk factors of hepatocellular carcinoma in patients with nonalcoholic steatohepatitis. Hepatology 2010;51:1972-8.

9. Fang YL, Chen H, Wang CL, Liang L. Pathogenesis of non-alcoholic fatty liver disease in children and adolescence: from “two hit theory” to “multiple hit model”. World J Gastroenterol 2018;24:2974-83.

10. Peiseler M, Schwabe R, Hampe J, Kubes P, Heikenwälder M, Tacke F. Immune mechanisms linking metabolic injury to inflammation and fibrosis in fatty liver disease - novel insights into cellular communication circuits. J Hepatol 2022;77:1136-60.

11. Nelson NP, Easterbrook PJ, McMahon BJ. Epidemiology of hepatitis B virus infection and impact of vaccination on disease. Clin Liver Dis 2016;20:607-28.

12. Leong TY, Leong AS. Epidemiology and carcinogenesis of hepatocellular carcinoma. HPB 2005;7:5-15.

13. Hurwitz LM, Townsend MK, Jordan SJ, et al. Modification of the association between frequent aspirin use and ovarian cancer risk: a meta-analysis using individual-level data from two ovarian cancer consortia. J Clin Oncol 2022;40:4207-17.

14. Shah D, Di Re A, Toh JWT. Aspirin chemoprevention in colorectal cancer: network meta-analysis of low, moderate, and high doses. Br J Surg 2023;110:1691-702.

15. Iannacone M, Sitia G, Isogawa M, et al. Platelets mediate cytotoxic T lymphocyte-induced liver damage. Nat Med 2005;11:1167-9.

16. Simon TG, Henson J, Osganian S, et al. Daily aspirin use associated with reduced risk for fibrosis progression in patients with nonalcoholic fatty liver disease. Clin Gastroenterol Hepatol 2019;17:2776-84.e4.

17. Koga H, Sakisaka S, Ohishi M, et al. Expression of cyclooxygenase-2 in human hepatocellular carcinoma: relevance to tumor dedifferentiation. Hepatology 1999;29:688-96.

18. Chen H, Cai W, Chu ESH, et al. Hepatic cyclooxygenase-2 overexpression induced spontaneous hepatocellular carcinoma formation in mice. Oncogene 2017;36:4415-26.

19. Tang TC, Poon RT, Lau CP, Xie D, Fan ST. Tumor cyclooxygenase-2 levels correlate with tumor invasiveness in human hepatocellular carcinoma. World J Gastroenterol 2005;11:1896-902.

20. Lu SC, Zhong JH, Tan JT, et al. Association between COX-2 gene polymorphisms and risk of hepatocellular carcinoma development: a meta-analysis. BMJ Open 2015;5:e008263.

21. Wang S, Zuo L, Lin Z, Yang Z, Chen R, Xu Y. The relationship between aspirin consumption and hepatocellular carcinoma: a systematic review and meta-analysis. Eur J Med Res 2023;28:226.

22. Yan LJ, Yao SY, Li HC, et al. Efficacy and safety of aspirin for prevention of hepatocellular carcinoma: an updated meta-analysis. J Clin Transl Hepatol 2022;10:835-46.

23. Tan RZH, Lockart I, Abdel Shaheed C, Danta M. Systematic review with meta-analysis: the effects of non-steroidal anti-inflammatory drugs and anti-platelet therapy on the incidence and recurrence of hepatocellular carcinoma. Aliment Pharmacol Ther 2021;54:356-67.

24. Simon TG, Wilechansky RM, Stoyanova S, et al. Aspirin for metabolic dysfunction-associated steatotic liver disease without cirrhosis: a randomized clinical trial. JAMA 2024;331:920-9.

25. Ma S, Qu G, Sun C, et al. Does aspirin reduce the incidence, recurrence, and mortality of hepatocellular carcinoma? A GRADE-assessed systematic review and dose-response meta-analysis. Eur J Clin Pharmacol 2023;79:39-61.

26. Chan AT. Aspirin, non-steroidal anti-inflammatory drugs and colorectal neoplasia: future challenges in chemoprevention. Cancer Causes Control 2003;14:413-8.

27. Zhou X, Zhang T, Sun Y, et al. Systematic review and meta-analysis: association of aspirin with incidence of hepatocellular carcinoma. Front Pharmacol 2022;13:764854.

28. Goh MJ, Sinn DH. Statin and aspirin for chemoprevention of hepatocellular carcinoma: time to use or wait further? Clin Mol Hepatol 2022;28:380-95.

29. Morris T, Stables M, Hobbs A, et al. Effects of low-dose aspirin on acute inflammatory responses in humans. J Immunol 2009;183:2089-96.

30. Abdelmalak J, Tan N, Con D, et al. The effect of aspirin use on incident hepatocellular carcinoma - an updated systematic review and meta-analysis. Cancers 2023;15:3518.

31. Wang S, Yu Y, Ryan PM, et al. Association of aspirin therapy with risk of hepatocellular carcinoma: a systematic review and dose-response analysis of cohort studies with 2.5 million participants. Pharmacol Res 2020;151:104585.

32. Lee CH, Hsu CY, Yen TH, Wu TH, Yu MC, Hsieh SY. Daily aspirin reduced the incidence of hepatocellular carcinoma and overall mortality in patients with cirrhosis. Cancers 2023;15:2946.

33. Liu Y, Ren T, Xu X, Jin J. Association of aspirin and nonaspirin NSAIDs therapy with the incidence risk of hepatocellular carcinoma: a systematic review and meta-analysis on cohort studies. Eur J Cancer Prev 2022;31:35-43.

34. Memel ZN, Arvind A, Moninuola O, et al. Aspirin use is associated with a reduced incidence of hepatocellular carcinoma: a systematic review and meta-analysis. Hepatol Commun 2021;5:133-43.

35. Singh J, Wozniak A, Cotler SJ, et al. Combined use of aspirin and statin is associated with a decreased incidence of hepatocellular carcinoma. J Clin Gastroenterol 2022;56:369-73.

36. Tan JL, Sidhu-Brar S, Woodman R, Chinnaratha MA. Regular aspirin use is associated with a reduced risk of hepatocellular carcinoma (HCC) in chronic liver disease: a systematic review and meta-analysis. J Gastrointest Cancer 2023;54:325-31.

37. Wang J, Li X. Impact of statin use on the risk and prognosis of hepatocellular carcinoma: a meta-analysis. Eur J Gastroenterol Hepatol 2021;33:1603-9.

38. Zeng RW, Yong JN, Tan DJH, et al. Meta-analysis: chemoprevention of hepatocellular carcinoma with statins, aspirin and metformin. Aliment Pharmacol Ther 2023;57:600-9.

39. Zhang S, Ren X, Zhang B, Lan T, Liu B. A systematic review of statins for the treatment of nonalcoholic steatohepatitis: safety, efficacy, and mechanism of action. Molecules 2024;29:1859.

40. Sharpton SR, Loomba R. Emerging role of statin therapy in the prevention and management of cirrhosis, portal hypertension, and HCC. Hepatology 2023;78:1896-906.

41. Cipollone F, Mezzetti A, Porreca E, et al. Association between enhanced soluble CD40L and prothrombotic state in hypercholesterolemia: effects of statin therapy. Circulation 2002;106:399-402.

42. Yano M, Matsumura T, Senokuchi T, et al. Statins activate peroxisome proliferator-activated receptor gamma through extracellular signal-regulated kinase 1/2 and p38 mitogen-activated protein kinase-dependent cyclooxygenase-2 expression in macrophages. Circ Res 2007;100:1442-51.

43. Zou B, Odden MC, Nguyen MH. Statin use and reduced hepatocellular carcinoma risk in patients with nonalcoholic fatty liver disease. Clin Gastroenterol Hepatol 2023;21:435-44.e6.

44. Wang Y, Wang W, Wang M, Shi J, Jia X, Dang S. A meta-analysis of statin use and risk of hepatocellular carcinoma. Can J Gastroenterol Hepatol 2022;2022:5389044.

45. Sung FC, Yeh YT, Muo CH, Hsu CC, Tsai WC, Hsu YH. Statins reduce hepatocellular carcinoma risk in patients with chronic kidney disease and end-stage renal disease: a 17-year longitudinal study. Cancers 2022;14:825.

46. Matsushita Y, Sugihara M, Kaburagi J, et al. Pravastatin use and cancer risk: a meta-analysis of individual patient data from long-term prospective controlled trials in Japan. Pharmacoepidemiol Drug Saf 2010;19:196-202.

47. Cholesterol Treatment Trialists’ (CTT) Collaboration; Emberson JR, Kearney PM, Blackwell L, et al. Lack of effect of lowering LDL cholesterol on cancer: meta-analysis of individual data from 175,000 people in 27 randomised trials of statin therapy. PLoS One 2012;7:e29849.

48. Facciorusso A, Abd El Aziz MA, Singh S, et al. Statin use decreases the incidence of hepatocellular carcinoma: an updated meta-analysis. Cancers 2020;12:874.

49. Pinyopornpanish K, Al-Yaman W, Butler RS, Carey W, McCullough A, Romero-Marrero C. Chemopreventive effect of statin on hepatocellular carcinoma in patients with nonalcoholic steatohepatitis cirrhosis. Am J Gastroenterol 2021;116:2258-69.

50. Fujiwara N, Kubota N, Crouchet E, et al. Molecular signatures of long-term hepatocellular carcinoma risk in nonalcoholic fatty liver disease. Sci Transl Med 2022;14:eabo4474.

51. Azit NA, Sahran S, Voon Meng L, Subramaniam M, Mokhtar S, Mohammed Nawi A. Risk factors of hepatocellular carcinoma in type 2 diabetes patients: a two-centre study in a developing country. PLoS One 2021;16:e0260675.

52. Chang Y, Liu Q, Zhou Z, et al. Can statin treatment reduce the risk of hepatocellular carcinoma? A systematic review and meta-analysis. Technol Cancer Res Treat 2020;19:1533033820934881.

53. German MN, Lutz MK, Pickhardt PJ, Bruce RJ, Said A. Statin use is protective against hepatocellular carcinoma in patients with nonalcoholic fatty liver disease: a case-control study. J Clin Gastroenterol 2020;54:733-40.

54. Hashemi Rafsanjani MR, Rahimi R, Heidari-Soureshjani S, Darvishi M, Adeli OA, Abbaszadeh S. Statin use and hepatocellular carcinoma risk: a comprehensive meta-analysis and systematic review. Recent Pat Anticancer Drug Discov 2024.

55. Islam MM, Poly TN, Walther BA, Yang HC, Jack Li YC. Statin use and the risk of hepatocellular carcinoma: a meta-analysis of observational studies. Cancers 2020;12:671.

56. Khazaaleh S, Sarmini MT, Alomari M, et al. Statin use reduces the risk of hepatocellular carcinoma: an updated meta-analysis and systematic review. Cureus 2022;14:e27032.

57. Vell MS, Loomba R, Krishnan A, et al. Association of statin use with risk of liver disease, hepatocellular carcinoma, and liver-related mortality. JAMA Netw Open 2023;6:e2320222.

58. Wong YJ, Qiu TY, Ng GK, Zheng Q, Teo EK. Efficacy and safety of statin for hepatocellular carcinoma prevention among chronic liver disease patients: a systematic review and meta-analysis. J Clin Gastroenterol 2021;55:615-23.

59. Zhang J, Fu S, Liu D, Wang Y, Tan Y. Statin can reduce the risk of hepatocellular carcinoma among patients with nonalcoholic fatty liver disease: a systematic review and meta-analysis. Eur J Gastroenterol Hepatol 2023;35:353-8.

60. Rasha F, Paul S, Simon TG, Hoshida Y. Hepatocellular carcinoma chemoprevention with generic agents. Semin Liver Dis 2022;42:501-13.

61. Garcia-Compean D, Jaquez-Quintana JO, Gonzalez-Gonzalez JA, Maldonado-Garza H. Liver cirrhosis and diabetes: risk factors, pathophysiology, clinical implications and management. World J Gastroenterol 2009;15:280-8.

62. Tangjarusritaratorn T, Tangjittipokin W, Kunavisarut T. Incidence and survival of hepatocellular carcinoma in type 2 diabetes patients with cirrhosis who were treated with and without metformin. Diabetes Metab Syndr Obes 2021;14:1563-74.

63. Ribback S, Peters K, Yasser M, et al. Hepatocellular ballooning is due to highly pronounced glycogenosis potentially associated with steatosis and metabolic reprogramming. J Clin Transl Hepatol 2024;12:52-61.

64. Li Q, Xu H, Sui C, Zhang H. Impact of metformin use on risk and mortality of hepatocellular carcinoma in diabetes mellitus. Clin Res Hepatol Gastroenterol 2022;46:101781.

65. Shankaraiah RC, Callegari E, Guerriero P, et al. Metformin prevents liver tumourigenesis by attenuating fibrosis in a transgenic mouse model of hepatocellular carcinoma. Oncogene 2019;38:7035-45.

66. Kramer JR, Natarajan Y, Dai J, et al. Effect of diabetes medications and glycemic control on risk of hepatocellular cancer in patients with nonalcoholic fatty liver disease. Hepatology 2022;75:1420-8.

67. Vilar-Gomez E, Calzadilla-Bertot L, Wong VW, et al. Type 2 diabetes and metformin use associate with outcomes of patients with nonalcoholic steatohepatitis-related, child-pugh a cirrhosis. Clin Gastroenterol Hepatol 2021;19:136-45.e6.

68. Huynh DJ, Renelus BD, Jamorabo DS. Reduced mortality and morbidity associated with metformin and SGLT2 inhibitor therapy in patients with type 2 diabetes mellitus and cirrhosis. BMC Gastroenterol 2023;23:450.

69. Wang L, Berger NA, Kaelber DC, Xu R. Association of GLP-1 Receptor agonists and hepatocellular carcinoma incidence and hepatic decompensation in patients with type 2 diabetes. Gastroenterology 2024;167:689-703.

70. Elsaid MI, Li N, Firkins SA, et al. Impacts of glucagon-like peptide-1 receptor agonists on the risk of adverse liver outcomes in patients with metabolic dysfunction-associated steatotic liver disease cirrhosis and type 2 diabetes. Aliment Pharmacol Ther 2024;59:1096-110.

71. Dwinata M, Putera DD, Hasan I, Raharjo M. SGLT2 inhibitors for improving hepatic fibrosis and steatosis in non-alcoholic fatty liver disease complicated with type 2 diabetes mellitus: a systematic review. Clin Exp Hepatol 2020;6:339-46.

72. Bea S, Jeong HE, Kim JH, Yu OH, Azoulay L, Shin JY. Sodium-glucose cotransporter-2 inhibitors and risk of hepatocellular carcinoma among patients with type 2 diabetes. Clin Gastroenterol Hepatol 2023;21:3451-4.e4.

73. Cho HJ, Lee E, Kim SS, Cheong JY. SGLT2i impact on HCC incidence in patients with fatty liver disease and diabetes: a nation-wide cohort study in South Korea. Sci Rep 2024;14:9761.

Cite This Article

Review
Open Access
Chemoprevention of hepatocellular carcinoma associated with metabolic dysfunction-associated steatotic liver disease: an updated review
Averie Dickinson, ... Kara WegermannKara Wegermann

How to Cite

Dickinson, A.; Dinani A.; Wegermann K. Chemoprevention of hepatocellular carcinoma associated with metabolic dysfunction-associated steatotic liver disease: an updated review. Hepatoma. Res. 2024, 10, 37. http://dx.doi.org/10.20517/2394-5079.2024.81

Download Citation

If you have the appropriate software installed, you can download article citation data to the citation manager of your choice. Simply select your manager software from the list below and click on download.

Export Citation File:

Type of Import

Tips on Downloading Citation

This feature enables you to download the bibliographic information (also called citation data, header data, or metadata) for the articles on our site.

Citation Manager File Format

Use the radio buttons to choose how to format the bibliographic data you're harvesting. Several citation manager formats are available, including EndNote and BibTex.

Type of Import

If you have citation management software installed on your computer your Web browser should be able to import metadata directly into your reference database.

Direct Import: When the Direct Import option is selected (the default state), a dialogue box will give you the option to Save or Open the downloaded citation data. Choosing Open will either launch your citation manager or give you a choice of applications with which to use the metadata. The Save option saves the file locally for later use.

Indirect Import: When the Indirect Import option is selected, the metadata is displayed and may be copied and pasted as needed.

About This Article

© The Author(s) 2024. Open Access This article is licensed under a Creative Commons Attribution 4.0 International License (https://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, sharing, adaptation, distribution and reproduction in any medium or format, for any purpose, even commercially, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made.

Data & Comments

Data

Views
89
Downloads
20
Citations
0
Comments
0
0

Comments

Comments must be written in English. Spam, offensive content, impersonation, and private information will not be permitted. If any comment is reported and identified as inappropriate content by OAE staff, the comment will be removed without notice. If you have any queries or need any help, please contact us at support@oaepublish.com.

0
Download PDF
Share This Article
Scan the QR code for reading!
See Updates
Contents
Figures
Related
Hepatoma Research
ISSN 2454-2520 (Online) 2394-5079 (Print)

Portico

All published articles are preserved here permanently:

https://www.portico.org/publishers/oae/

Portico

All published articles are preserved here permanently:

https://www.portico.org/publishers/oae/