Clin Mol Hepatol > Volume 28(4); 2022 > Article
Shin, Jun, and Yi: Impact of diabetes, obesity, and dyslipidemia on the risk of hepatocellular carcinoma in patients with chronic liver diseases

ABSTRACT

Despite the increasing prevalence of metabolic disorders, the potential effects of metabolic factors on hepatocellular carcinoma (HCC) development in individuals with chronic liver diseases (CLDs) are not well understood. For a metabolic factor to be identified as a risk factor for HCC in patients with CLDs, such as hepatitis B virus (HBV) and hepatitis C virus (HCV) infection, there should be a strong synergistic interaction between the carcinogenic mechanisms of the metabolic factor and the CLD itself. This review aims to comprehensively summarize the published data on the relationship between metabolic factors such as diabetes mellitus (DM), obesity, and blood lipids and the risk of HCC in patients with CLDs. DM consistently increases the risk of HCC in patients with CLD. When associated with DM, the risk of HCC seems to be highest in HCV and non-alcoholic fatty liver disease (NAFLD), followed by alcoholic liver disease (ALD) and HBV. Obesity may increase the risk of HCC. Among CLDs, the evidence is relatively consistent and clear for ALD, while clear evidence is limited in other CLDs including HBV, HCV, and NAFLD. Total cholesterol, potentially low-density lipoprotein cholesterol and triglyceride, seems to have strong inverse associations with HCC in individuals with CLDs. Despite evidence from observational studies, statins had no effect in preventing HCC in randomized controlled trials. Whether statins have a preventive effect against HCC is unclear. A better understanding and management of metabolic factors may be beneficial to reduce the risk of HCC in patients with CLDs.

INTRODUCTION

Liver cancer is the sixth most common cancer worldwide, and hepatocellular carcinoma (HCC) accounts for 75–85% of all primary liver cancers [1]. HCC is the third leading cause of cancer-related death among solid cancers, leading to approximately 1 million deaths yearly [2]. Chronic liver diseases (CLDs), such as hepatitis B virus (HBV) infection and liver cirrhosis (LC), are well known risk factors for the development of HCC.
Recently, increasing evidence has suggested that metabolic factors, including diabetes mellitus (DM), obesity, dyslipidemia, and metabolic syndromes, are risk factors for HCC [3-6]. In populations with a low prevalence of viral hepatitis, the overall impact of metabolic factors on HCC was suggested to be greater than that of viral hepatitis [7]. The effects of metabolic factors on HCC have not been well established in individuals with CLDs. Observational studies have suggested the potential beneficial effects of some pharmaceutical agents for metabolic disorders (e.g., statins) in preventing HCC.
The purpose of this review is to summarize the associations between metabolic factors such as DM, dyslipidemia, and obesity and the risk of HCC in patients with CLDs. In this review, we focused on HBV, hepatitis C virus (HCV) infection, alcoholic liver disease (ALD), non-alcoholic fatty liver disease (NAFLD), and LC among CLDs. The associations in the general population were briefly summarized for comparison. A clearer understanding of the role of metabolic factors in the development of HCC will help in informed decision-making and patient management aimed at reducing the risk of HCC in patients with CLDs and comorbid metabolic disorders.

ROLE OF METABOLIC FACTORS IN CLDS: A SYNERGISTIC INTERACTION

Patients with CLDs, such as HBV/HCV, have a several dozen-fold higher risk of HCC development compared with individuals without CLDs, while metabolic disorders, such as diabetes and obesity, are associated with a 2–3-fold higher risk of HCC in the general population. If patients with CLD and comorbid metabolic disorders have a higher risk of HCC, then carcinogenic mechanisms by CLDs and metabolic disorders should have a strong synergistic interaction. Let us assume that HBV patients without diabetes have a 50-fold higher risk for HCC development compared to the general population without diabetes and that diabetic patients have a 2-fold higher risk compared with general population without diabetes. If HBV and diabetes are independent risk factors without synergistic interaction, compared to individuals without HBV and diabetes, and HBV patients without diabetes, HBV patients with diabetes will have approximately 51-fold and 1.02-fold higher risk, respectively, for HCC development (likewise, if HBV patients with diabetes have a 2-fold higher risk compared to those without diabetes, then they have a 100-fold higher risk compared with individuals without diabetes and HBV) [8,9]. Additionally, the stronger the impact of a certain CLD (such as LC) on HCC development is, the stronger synergistic interaction should exist in individuals with CLD for a metabolic factor to be identified as a risk factor in individuals with CLD. It is common that the associations of a factor are weaker in individuals with diseases than in the general population. Potential mechanisms should be focused on the synergistic interactions of mechanisms by CLDs and metabolic factors (rather than on the general mechanism of a factor per se). However, few detailed mechanisms have been proposed, and further research would be needed to explain potential synergic interactions.

DM

General population

In general population studies, DM has been associated with an increased risk of HCC. A Korean study using the National Health Insurance Service-Health Screening Cohort (NHIS-HEALS) reported that DM was associated with a higher risk of HCC (hazard ratio [HR], 1.82) [10]. Two meta-analyses showed that DM was associated with a risk ratio (RR) of 2.01 and 2.31 for HCC development [11,12]. However, liver diseases themselves increase the risk of DM development [13-15]. Thus, at least some part of the association between DM and HCC in the general population may be due to reverse causation.

Patients with viral hepatitis

DM has been associated with an increased risk of HCC in patients with CLD. The magnitude of association between DM and HCC somewhat differed for each liver disease (Table 1) [16-30]. A recent Korean study on 214,167 male patients with HBV reported that the presence of DM was significantly associated with a higher risk of HCC (HR, 1.23) [16]. In a recent systematic review and meta-analysis of 36 studies, DM was associated with an HR of 1.36 for HCC development in patients with HBV [17]. A systematic review of seven studies in 10,700 patients with HCV reported that DM increased the risk of HCC by approximately 2-fold [19]. The HCC risk associated with DM appears to be stronger in HCV than in HBV [19,29]. However, in a US study in 52,671 HCV-LC patients (7,605 HCC cases), DM was not associated with the risk of HCC [20].

Patients with LC

In ALD-LC patients, DM was associated with approximately 1.5-fold increased risk of HCC [20,23]. A recent systematic review reported that DM increases 2.65-fold the risk of HCC incidence in NAFLD patients [30]. In NAFLD patients with/without LC, HRs associated with DM for HCC were 1.24 (407 HCC cases), 1.93 (608 HCC cases), 1.30 (291 HCC cases), and 2.77 (253 HCC cases) in studies with >200 HCC cases, while the corresponding HRs were 4.18 (30 HCC cases), 2.90 (28 HCC cases), 4.72 (41 HCC cases), and 3.21(16 HCC cases) in studies with <50 HCC cases [20,23-28]. The magnitude of association between DM and the risk of HCC in NAFLD patients may be comparable to that in HCV patients or in general populations.

Patients with ALD and NAFLD

The HCC risk associated with DM seemed to be highest in NAFLD, followed by ALD and HBV. However, HCV and NAFLD are well known to increase the DM incidence by approximately 2-fold [14,15]. Comorbid DM is considered a marker of severity in NAFLD [31,32]. Therefore, the strongest associations do not necessarily mean strongest causal effect of DM on HCC development in NAFLD. Overall, diabetes may have causal effects on HCC in patients with CLDs. However, part of the effects of DM may reflect the underlying severity of the liver disease.

Anti-diabetic drugs in CLDs

Only a few studies on the effects of anti-diabetic drugs on the HCC risk in patients with CLDs have been conducted, and the results have been inconsistent (Table 2). In HBV patient, some recent cohort studies reported that metformin increased the HCC risk [33,34]. Whereas, another cohort study reported that metformin was not associated with HCC [35]. While in studies on patients with NAFLD, metformin did not have a significant association with HCC risk [24,36]. In contrast, thiazolidinediones reduced the risk of HCC in patients with HBV (HR, 0.46) [35]. Kramer et al. [37] reported that insulin and sulfonylureas alone had no effect on HCC risk, but insulin in combination with oral medication (metformin, sulfonylurea) had a higher risk of HCC with NAFLD. Overall, there is a lack of evidence, and it is unclear whether the use of anti-diabetic medication itself affects HCC development or whether different types of diabetic medications differently affect the development of HCC in patients with CLD.

OBESITY

In this review, the international standard of body mass index (BMI) classification, namely underweight (BMI <18.5 kg/m2), normal weight (18.5–24.9 kg/m2), overweight (25–29.9 kg/m2), and obesity (≥30 kg/m2), was used, instead of the classification by the Korean Society for the Study of Obesity that defines obesity as BMI ≥25 kg/m2.

General population

Obesity is considered a risk factor for HCC, especially in populations of European origin [38,39]. Meta-analyses in the general, mostly Western, populations showed that obesity increased the risk of HCC (or primary liver cancer) by approximately two times [40,41]. In Asian populations, despite their slim body shape, obesity-related HCC risks were substantially lower than Western populations and the summed relative risk was approximately 1.5 in a meta-analysis [10,40-44].

Patients with viral hepatitis

Most studies on patients with HBV have been conducted in Asia, especially Taiwan (Table 3) [45-49]. The Taiwanese studies did not show consistent association between obesity and HCC risk [45-48]. A recent study on Korean adults with chronic HBV showed that obesity, compared to a BMI of 18.5–22.9, was associated with 22% (men: HR, 1.22; 95% confidence interval [CI], 1.09–1.36]) and 46% (women: HR, 1.46; 95% CI, 1.24–1.71) higher risk of HCC [49]. This large-scale study using five categories of BMI identified relatively weak associations between obesity and HCC. Previous studies may not have found these subtle results consistently, probably due to the small number of participants, the crude categories of obesity generally used (such as obesity vs. non-obesity), and the inclusion of only men in some cases [45-48]. Obesity may be a risk factor for HCC in patients with HBV; however, the effect size seemed to be weaker than that in the general population.
Most studies reported that BMI or obesity were not associated with HCC in patients with HCV infections [21,22,50-54]. Two studies, one Japanese and one Taiwanese, which are commonly cited concerning the higher HCC risk associated with obesity, have a major limitation [55,56]. In the Japanese study, relative risks associated with obesity were calculated by comparing with underweight, not with normal weight [55]. In the Taiwanese study, the higher HCC risk associated with obesity was based on a very small number of HCC cases (five cases of obesity) [56]. Overall, the association between obesity and HCC has been inconsistent in patients with HCV. Considering the similarity of the impact of HBV and HCV on HCC, obesity might impact HCC in individuals with HCV infections similarly to that in individuals with HBV infection; that is, obesity may modestly increase HCC development in individuals with HCV.

Patients with LC

In individuals with LC, the associations have been complex and inconsistent and have shown the potential to differ according to LC etiology, which includes ALD, NAFLD, HBV, and HCV [20,23,24,57-63]. In two studies on NAFLD-LC patients who received care in the US Veterans Affairs healthcare system from the same authors, one showed a potentially higher HCC risk associated with high BMI, whereas the other did not [20,23]. The associations appeared to be clearer and stronger in alcoholic LC than in viral LC [20,57]. These results suggest a strong synergistic interaction between ALD (or alcohol consumption) and obesity, which was also observed in general populations [47].

Patients with NAFLD

The studies conducted among NAFLD patients of Western populations, including those without cirrhosis/fibrosis, found that BMI or obesity was not statistically significantly associated with HCC [25,59,64]. It is surprising that the impact of obesity on HCC development has not been clearly shown in NAFLD patients. However, these NAFLD studies, including those conducted mostly on non-cirrhotic patients, mainly used crude categories of BMI (obesity vs. non-obesity) and possibly lacked statistical power to detect the true association [25,59,64]. Additionally, probably because cirrhosis is a main mechanism of NAFLD-related HCC, the association between obesity and HCC might not be apparent in NAFLD-LC patients [20,23,24,59].
In summary, obesity may increase the risk of HCC in patients with CLD, but probably with a weaker effect size than that in the general population [21,52,53]. Liver damage caused by obesity and CLD itself may synergistically interact together and further facilitate the progression and/or development of HCC in patients with CLD. However, it should be noted that there is limited evidence even in NAFLD cases.

Central obesity

Compared to the lowest category, the highest waist circumference (WC) category had a 1.59 times higher risk of HCC development in a recent systematic review, mostly in the general population [65]. A recent Chinese study on chronic HBV patients reported that central obesity defined by waist-to-height ratio, not by WC and waist-hip ratio, was associated with an increased risk of HCC (HR, 1.63; 95% CI, 1.11–2.38) compared to non-central obesity. However, the risk of HCC with changes in the waist-to-height ratio was associated with an increased risk of HCC both with a decrease and an increase in the ratio [66]. In a small Korean study among 102 chronic HBV patients (seven HCC cases), central obesity, defined by WC, waist-hip ratio, and visceral fat area, was not associated with HCC risk [67]. Despite the potential, it is difficult to conclude whether central obesity is a better predictor and risk factor than general obesity in individuals with CLDs, due to the very small number of studies.

DYSLIPIDEMIA

General population

As each type of dyslipidemia may have different associations with HCC, we focused on studies reporting specific lipid profiles, such as total cholesterol (TC), high-density lipoprotein cholesterol (HDL-C), low-density lipoprotein cholesterol (LDL-C), and triglyceride (TG). Most studies revealed that low TC levels were strongly associated with an increased risk of HCC development [6,68-74]. In a Korean study that used the NHIS-HEALS, a nearly 50% reduced risk of HCC was observed for every 39 mg/dL (1 mmol/L) increase in TC (HR, 0.54; 95% CI, 0.51–0.58) [71]. Only a few studies have examined the associations of other lipids with HCC. Low levels of LDL-C and TG were generally associated with a higher risk of HCC, while HDL-C showed unclear inverse associations [69,70,74]. Overall, TC and potentially LDL-C and TG, seemed to have clear inverse associations with HCC in the general population, whereas the dose-response association of HDL-C with HCC was unclear.

Patients with CLDs: TC

In patients with CLDs, like in the general population, TC levels were inversely associated with the risk of HCC. Higher TC levels were generally associated with a lower risk of HCC in individuals with viral hepatitis (including HBV and HCV), LC, and NAFLD (Table 4) [36,70,75-77]. Compared to the studies on TC, a relatively small number of studies have reported associations between other lipid profiles and HCC.
While low TC levels may be a useful marker for HCC development in patients with CLD, the potential causal effects of low TC levels are uncertain. The presence of CLD leads to hepatic damage, which, in turn, is related to changes in lipid metabolism. Serum cholesterol levels in patients with HBV, HCV, and HCC were reported to be lower than those in the normal control group [78-80]. The decrement in cholesterol levels positively correlated with the severity of liver disease [78,79,81-85]. Statin affected lower levels of cholesterol levels were not associated with higher risk of HCC [71]. Low TC levels seemed to be more a strong predictor, but less a risk factor for HCC. Future studies are needed to clarify the role of lipid profiles, such as whether they have independent effects on HCC development.

Patients with CLDs: other lipid profile

The association between LDL-cholesterol and HCC was similar to that between TC and HCC [70,77]. A recent population-based Korean study on HBV patients showed that HCC was negatively associated with increased TG levels [86]. In a large-scale Korean study, TG showed inverse associations and low HDL-cholesterol (L-curve) levels were associated with a higher risk of HCC in individuals with chronic viral hepatitis or LC, whereas a smaller study (89 HCC cases) reported that high TG levels, but not low HDL-cholesterol levels, were associated with a higher risk of HCC in patients with HBV [70,77]. Dyslipidemia, defined by high TG and/or low HDL-cholesterol levels, was not associated with a higher risk of HCC in American patients with NAFLD [25]. Overall, similar to the results in the general population, TC, potentially LDL-cholesterol and TG, seemed to have inverse associations with HCC in individuals with CLDs, whereas the association of low HDL-cholesterol with HCC was unclear.

Statin therapy in CLDs

In several observational studies, statin therapy has generally been associated with a reduced risk of HCC development in patients with CLDs (Table 5). Statin use was associated with a materially lower risk of HCC in patients with HBV, HCV, and NAFLD [17,33,36,75,76,87-94]. A recent meta-analysis of studies on CLD reported that statin use was associated with an HR of 0.57 for HCC development [94].
Caution should be exercised when interpreting the beneficial effects of statins on HCC based on evidence from observational studies. The higher the cholesterol level, the higher the possibility of statin use. The natural low cholesterol level, not the attained level after statin use, is an important predictor and a potential risk factor for HCC [71]. Therefore, when the association between statin use and HCC risk is examined in observational studies, only patients newly initiated on statin treatment should be enrolled as statin users and their natural cholesterol levels before statin use should be measured. The preventive effect of statins disappeared in such studies on the general population [71,95]. Importantly, in the ad hoc analysis of randomized controlled trials for cardiovascular disease prevention, statins had no effect in preventing HCC (HR, 1.06 for statin use/more use compared with non-use/less use) [96,97]. Overall, whether statins have a preventive effect against HCC remains unclear. In the meantime, for the prevention of cardiovascular diseases, there is no need to apply different criteria for starting statins in patients with CLD and the general population.

SYNERGISTIC EFFECTS OF METABOLIC FACTORS

Several studies have examined whether metabolic factors have a synergistic effect on the development of HCC in patients with CLDs. In a study on patients with cirrhosis, the HR for obesity alone was 2.1, and the HR for DM alone was 1.4. The coexistence of obesity and DM (compared to BMI <25 kg/m2 and no diabetes) increased the risk of HCC by >6 times [60]. However, this study only adjusted for age, sex, and a cirrhosis etiology. In a Taiwanese study on patients with HBV, a single metabolic factor did not increase the risk of HCC; however, the coexistence of three or more metabolic factors increased the risk of HCC [98]. In a retrospective study on 271,906 patients with NAFLD in the USA, obesity, hypertension, diabetes, and dyslipidemia had no clear synergistic effects on the composite occurrence of HCC and LC [25]. In a recent study on Korean patients with HBV, a higher number of metabolic syndrome components was negatively associated with the risk of liver cancer [86]. Overall, the evidence of a synergistic interaction among metabolic factors is insufficient. Further studies are needed to elucidate this issue.

LIMITATION

First, only a limited number of studies are available on the association between lipids other than TC and anti-diabetic agents and HCC development, although the number of studies conducted on patients with CLDs is increasing. Second, statistical models seemed to adjust for potential mediators of the effects of metabolic factors in most studies that included information on the associations between metabolic factors and HCC. For example, it is inadequate to adjust for diabetes, blood pressure, and cholesterol when examining the association between obesity and cardiovascular disease/mortality because these factors are considered as mediators of the effects of obesity [99]. Although this is understandable, since most of the studies were not specifically examining metabolic factors. Nonetheless, these studies may not accurately capture the specific associations of metabolic factors. Third, it is difficult to compare potential regional differences (such as between Western and Eastern populations) in this association. For example, most studies on HBV have been conducted in East Asia, while NAFLD and non-viral LC have been mainly studied in the Western population. Finally, most of the previous studies reviewed were retrospective studies, which are more prone to bias than prospective studies. More prospective, large-scale studies are needed to better capture these associations.

CONCLUSION

Comorbid DM increases the risk of HCC in patients with CLD. However, at least some of the effects of DM on HCC reflect the underlying severity of CLDs (reverse causality), especially for HCV and NAFLD. Obesity may increase the risk of HCC in patients with CLD, although the magnitude of the effect is weaker than that in the general population. Although obesity increases the risk of NAFLD, there is limited evidence on whether obesity increases the HCC risk in patients with NAFLD. Low lipid levels, especially TC, and perhaps LDL-C and TG, rather than dyslipidemia, are associated with an increased risk of HCC. TC levels may be used as a marker of liver disease severity associated with the risk of HCC. Whether statins or anti-diabetic agents have a preventive effect against HCC in patients with CLD remains unclear.

FOOTNOTES

Authors’ contributions
Study concept and design: Baek Gyu Jun and Sang-Wook Yi; Data analysis and interpretation: Baek Gyu Jun, Hwang Sik Shin, and Sang-Wook Yi; Wrote the paper: Baek Gyu Jun, Sang-Wook Yi, and Hwang Sik Shin; All authors have read and approved the final version of the manuscript.
Conflicts of Interest
The authors have no conflicts to disclose.

Table 1.
Association between diabetes mellitus and hepatocellular carcinoma
Study Region Etiology Study design Patients/HCC cases Result (95% CI) P-value
Kim et al. [16] (2018) Korea HBV Population-based cohort study 214,167/11,241 HR, 1.23 (1.15–1.34) <0.01
Campbell et al. [17] (2021) HBV Meta-analysis 40 studies (536,456) HR, 1.36 (1.20–1.32) <0.01
Hsu et al. [18] (2018) Taiwan HBV Population-based cohort study 23,851/596 HR, 1.3 (1.1–1.6)
Dyal et al. [19] (2016) HCV Meta-analysis 9 studies HR, 1.73 (1.30–2.30); RR, 3.52 (1.29–9.24)
Yang et al. [29] (2022) HBV Meta-analysis 11 studies RR, 1.37 (1.24–1.51); OR, 1.99 (0.73–5.48)
HCV 15 studies RR, 1.76 (1.42–2.17); OR, 1.77 (1.18–2.64)
Chen et al. [30] (2022) NAFLD-LC Meta-analysis 8 studies HR, 4.55 (2.34–8.87)
NAFLD HR, 2.65 (2.02–3.49)
Ioannou et al. [20] (2018) USA HCV-LC Retrospective cohort study 44,007/7,605 HR, 1.02 0.6
ALD-LC 29,326/1,388 HR, 1.54 <0.01
NAFLD-LC 13,456/608 HR, 1.93 <0.01
Huang et al. [21] (2015) Taiwan HCV Population-based cohort study 2,187/82 HR, 1.91 (1.10–3.30) 0.02
Arase et al. [22] (2013) Japan HCV Retrospective cohort study 4,302/393 HR, 1.73 (1.30–2.30) <0.01
Ioannou et al. [23] (2019) USA ALD-LC Retrospective cohort study 16,175/871 HR, 1.46 <0.01
NAFLD-LC 7,068/407 HR, 1.24 0.10
Yang et al. [24] (2020) USA NAFLD-LC Hospital-based cohort study 354/30 HR, 4.18 (1.23–14.2) 0.02
Retrospective cohort study 6,630/291 HR, 1.30 (1.02–1.66) 0.03
Kanwal et al. [25] (2020) USA NAFLD Retrospective cohort study 271,906/253 HR, 2.77 (2.03–3.77)
Bertot et al. [26] (2018) Australia NAFLD Hospital-based cohort study 284/28 HR, 2.9 (1.2–7.3) 0.02
Vilar-Gomez et al. [27] (2018) Spain, Australia, Hong Kong, and Cuba NAFLD Cohort study 456/41 HR, 4.72 (2.13–10.45) <0.01
Kawamura et al. [28] (2012) Japan NAFLD Retrospective cohort study 6,508/16 HR, 3.21 (1.09–9.50) 0.04

HCC, hepatocellular carcinoma; CI, confidence interval; HBV, hepatitis B virus; HR, hazard ratio; HCV, hepatitis C virus; RR, risk ratio; OR, odds ratio; NAFLD, non-alcoholic fatty liver disease; LC, liver cirrhosis; ALD, alcoholic liver disease.

Table 2.
Association between anti-diabetic drugs and hepatocellular carcinoma
Study Region Drug Etiology Study design Patients/HCC cases Result (95% CI) P-value
Chen et al. [33] (2015) Taiwan Metformin HBV Population-based cohort study 71,824/1,753 HR, 1.25 (1.06–1.47) <0.01
Hsu et al. [34] (2018) Taiwan Metformin HBV Population-based cohort study 27,820/802 HR, 2.20 (1.86–2.60) <0.01
Yip et al. [35] (2020) Hongkong Thiazolidinediones HBV Retrospective cohort study 28,999/2,307 HR, 0.46 (0.24–0.88) 0.19
Yang et al. [24] (2020) USA Metformin NAFLD Retrospective cohort study 354/30 HR, 1.93 (0.84–4.41) 0.12
Lee et al. [36] (2017) Taiwan Metformin NAFLD Population-based cohort study 18,080 HR, 1.29 (0.47–3.54) 0.62
Kramer et al. [37] (2022) USA Insulin NAFLD Retrospective cohort study 85,963/524 HR, 1.05 (0.88–1.27) 0.57
Sulfonylureas HR, 0.98 (0.84-.1.16) 0.84
Insulin-metformin HR, 1.53 (1.25-1.86) <0.01
Insulin-metformin-sulfonylureas HR, 1.71 (1.41-2.08) <0.01

HCC, hepatocellular carcinoma; CI, confidence interval; HBV, hepatitis B virus; HR, hazard ratio; NAFLD, non-alcoholic fatty liver disease.

Table 3.
Association between obesity and hepatocellular carcinoma
Study Region Etiology Study design Patients/HCC cases Result (95% CI) P-value
Yu et al. [45] (2008) Taiwan HBV Cohort study 2,903/134 HR, 1.48 (1.04–2.12); BMI 25–30
HR, 1.96 (0.72–5.38); BMI >30
Fu et al. [46] (2015) Taiwan HBV Cohort study 4,179/111 HR, 0.31 (0.04–2.24); obesity* 0.25
Loomba et al. [47] (2010) Taiwan HBV Cohort study 2,260/135 HR, 1.00 (0.93–1.06)|| 0.54
Chao et al. [48] (2011) Taiwan HBV Cohort study 1,142/124 HR, 1.58 (1.16–2.17); BMI ≥25* <0.01
Kim et al. [49] (2018) Korea HBV Population-based cohort study 370,322/14,609 HR, 1.04 (0.99–1.09); men; BMI 25.0–29.9
HR, 1.22 (1.09–1.36); men; BMI ≥30 <0.01
HR, 1.25 (1.15–1.36); women; BMI 25.0–29.9 <0.01
HR, 1.46 (1.24–1.71); women; BMI ≥30 <0.01
Huang et al. [21] (2015) Taiwan HCV Population-based cohort study 2,187/82 HR, 1.79 (0.43–7.44); obesity* 0.42
Arase et al. [22] (2013) Japan HCV Retrospective cohort study 4,302/393 uHR, 1.37 (1.12–1.66); BMI ≥22* <0.01
Veldt et al. [50] (2008) Europe, Canada HCV Prospective 541 HR, 0.94 (0.84–1.05); BMI|| 0.29
Arano et al. [51] (2011) Japan HCV Retrospective cohort study 325/122 HR, 1.01 (0.93–1.09); men|| 0.76
HR, 1.09 (0.99–1.19); women|| 0.06
Yang et al. [52] (2016) USA HCV Hospital-based cohort study 739/69 uHR, 1.0 (1.0–1.0); BMI|| 0.74
Lok et al. [53] (2009) USA HCV Cohort study 1,005/48 Lower BMI was associated with HCC (unadjusted) 0.04
McMahon et al. [54] (2017) USA HCV Population-based cohort study 1,080/40 uHR, 1.06 (0.5–2.1); BMI ≥30* 0.87
Ohki et al. [55] (2008) Japan HCV Retrospective cohort study 1,431/122 HR, 1.86 (1.09–3.16); BMI 25–30§ 0.02
HR, 3.10 (1.41–6.81); BMI >30§ 0.01
Hung et al. [56] (2011) Taiwan HCV Cohort study 1,470/87 HR, 1.31 (0.83–2.05); BMI ≥25* 0.24
Ioannou et al. [20] (2018) USA LC Retrospective study 116,404/10,042 HR, 1.0; BMI 24.5–28; HCV–LC†† 0.96
HR, 0.93; BMI 28–32; HCV–LC†† 0.04
HR, 0.84; BMI >32; HCV–LC <0.01
HR, 1.2; BMI 24.5–28; NAFLD–LC 0.48
HR, 1.52; BMI 28–32; NAFLD–LC 0.09
HR, 1.45; BMI >32; NAFLD–LC 0.10
HR, 1.59; BMI 24.5–28; ALD–LC†† <0.01
HR, 1.74; BMI 28–32; ALD–LC†† <0.01
HR, 1.88; BMI >32; ALD–LC†† <0.01
Ioannou et al. [23] (2019) USA LC Retrospective cohort study 23,243/1,278 HR, 1.32; BMI 25.2–29.3; ALD-LC‡‡ <0.01
HR, 1.39; BMI 29.3–33.8; ALD-LC ‡‡ <0.01
HR, 1.49; BMI 33.8–38.5; ALD-LC‡‡ <0.01
HR, 1.31; BMI >38.5; ALD-LC ‡‡ 0.06
HR, 0.91; BMI 25.2–29.3; NAFLD-LC ‡‡ 0.64
HR, 0.9; BMI 29.3–33.8; NAFLD-LC ‡‡ 0.60
HR, 0.8; BMI 33.8–38.5; NAFLD-LC‡‡ 0.28
HR, 0.77; BMI >38.5; NAFLD-LC ‡‡ 0.22
Yang et al. [24] (2020) USA LC Hospital-based cohort study 354/30 uHR, 0.99 (0.95–1.03); Mayo cohort; BMI|| 0.59
Cohort study 6,630/291 uHR, 1.00 (0.98–1.02); UNOS cohort; BMI|| 0.83
Nair et al. [57] (2002) USA LC Large-population database study 19,271/659 HR, 1.65 (1.22–2.22); BMI ≥30* <0.01
Pais et al. [58] (2015) France LC Retrospective study 110/29 OR, 8.24 (2.04–33.32); BMI >25* <0.01
Grimaudo et al. [59] (2020) Italy LC Prospective study 162/13 HR, 0.86 (0.21–3.51); BMI ≥30* 0.83
N’Kontchou et al. [60] (2006) France LC Retrospective cohort study 771/220 HR, 2.0 (1.4–2.7); BMI 25.0–29.9** <0.01
HR, 2.8 (2.0–4.0); BMI ≥30** <0.01
Archambeaud et al. [61] (2015) France LC Case-control study 905/282 OR, 1.56 (1.02–2.37); past obesity* 0.03
uOR, 1.04 (0.72–1.48); current obesity* 0.84
Brichler et al. [62] (2019) France LC Prospective cohort study 317/27 HR, 2.67 (1.04–6.84); BMI ≥30* 0.04
Ioannou et al. [63] (2007) USA LC Cohort study 2,126/173 HR, 2.8 (1.4–5.4); BMI 25.0–29.9**
HR, 2.5 (1.3–4.9); BMI ≥30
Kanwal et al. [25] (2020) USA NAFLD Retrospective cohort study 271,906/253 HR, 1.31 (0.98–1.74); BMI >30*
Ito et al. [64] (2020) Japan NAFLD Retrospective study 179/7 uHR, 1.07 (0.21–5.51); BMI ≥25* 0.94
Fan et al. [66] (2021) China HBV Cohort study 5,754/142 HR, 1.63 (1.11–2.38); waist-to-height ratio >0.50* 0.01
Lee et al. [67] (2016) Korea HBV Cohort study 102/7 uHR, 0.88 (0.17–4.53); WC ≥90 (men) or ≥80 (women)* 0.88
uHR, 1.94 (0.23–15.87); waist-to-hip ratio ≥0.9* 0.39
uHR, 1.69 (0.43–8.71), visceral fat area ≥100 cm2 * 0.50

HCC, hepatocellular carcinoma; CI, confidence interval; HBV, hepatitis B virus; HR, hazard ratio; BMI, body mass index (kg/m2); HCV, hepatitis C virus; uHR, unadjusted HR; LC, liver cirrhosis; HCV, hepatitis C virus; NAFLD, non-alcoholic fatty liver disease; ALD, alcoholic liver disease; UNOS, United Network for Organ Sharing; OR, odds ratio; uOR, unadjusted OR; WC, waist circumference (cm).

* Two group analysis, for example obesity vs. no obesity, BMI ≥25 vs. BMI <25 kg/m2.

Reference: BMI 18.5–24.9 kg/m2.

Reference: BMI 18.5–22.9 kg/m2.

§ Reference: BMI ≤18.5 kg/m2.

|| HR per 1 unit increase in BMI.

** Reference: BMI <25 kg/m2.

†† Reference: BMI 18.0–24.5 kg/m2.

‡‡ Reference: BMI ≤25.2 kg/m2.

Table 4.
Association between dyslipidemia and hepatocellular carcinoma
Study Region Etiology Study design Patients/HCC cases Result (95% CI) P-value
Lee et al. [36] (2017) Taiwan NAFLD Population-based cohort study 18,080/41 HR, 0.41 (0.15–1.11) 0.08
Cho et al. [70] (2021) Korea LC Large-population database study 6,399/561 HR, 0.78 (0.64–0.96); TC Q2*
HR, 0.65 (0.51–0.83); TC Q3
HR, 0.30 (0.21–0.44); TC Q4
HR, 0.78 (0.58–1.05); TG Q2
HR, 0.69 (0.44–1.07); TG Q3
HR, 0.47 (0.23–0.94); TG Q4
HR, 0.77 (0.63–0.94); LDL Q2
HR, 0.64 (0.51–0.81); LDL Q3
HR, 0.29 (0.21–0.40); LDL Q4
HR, 0.95 (0.75–1.20); HDL Q2
HR, 1.01 (0.80–1.28); HDL Q3
HR, 1.18 (0.94–1.49); HDL Q4
Viral hepatitis 86,368/806 HR, 0.69 (0.58–0.82); TC Q2*
HR, 0.50 (0.41–0.61); TC Q3
HR, 0.31 (0.24–0.39); TC Q4
HR, 0.78 (0.58–1.05); TG Q2
HR, 0.69 (0.44–1.07); TG Q3
HR, 0.47 (0.23–0.94); TG Q4
HR, 0.77 (0.63–0.94); LDL Q2
HR, 0.64 (0.51–0.81); LDL Q3
HR, 0.29 (0.21–0.40); LDL Q4
HR, 0.95 (0.75–1.20); HDL Q2
HR, 1.01 (0.80–1.28); HDL Q3
HR, 1.18 (0.94–1.49); HDL Q4
Goh et al. [75] (2020) Korea HBV Retrospective cohort study 7,713/702 HR, 0.77 (0.63–0.95); high TC 0.01
Wu et al. [76] (2014) Taiwan HBV Population-based cohort study 43,190/5,446 HR, 0.87 (0.61–1.23) 0.43
Tan et al. [77] (2019) Taiwan HBV Population-based cohort study 6,564/89 HR, 0.67 (0.39–1.16); high TG 0.15
HR, 0.99 (0.54–1.83); low HDL 0.98
Choe et al. [86] (2021) Korea HBV Large-population database study 1,504,880/29,412 HR, 0.46 (0.45-0.47); high TG
HR, 0.68 (0.66-0.70); low HDL
Kanwal et al. [25] (2020) USA NAFLD Retrospective cohort study 271,906/253 HR, 1.31 (0.84–2.04); high TG and/or low HDL-cholesterol

HCC, hepatocellular carcinoma; CI, confidence interval; NAFLD, non-alcoholic fatty liver disease; HR, hazard ratio; LC, liver cirrhosis; TC, total cholesterol; Q, quartile; TG, triglyceride; LDL, low-density lipoprotein; HDL, high-density lipoprotein; HBV, hepatitis B virus.

* Quartile 1 (Q1, lowest), Q2, Q3, and Q4 (highest). The reference was Q1.

Table 5.
Association between statin use and hepatocellular carcinoma
Study Region Etiology Study design Patients/HCC cases Result (95% CI) P-value
Campbell et al. [17] (2020) HBV Meta-analysis 40 studies (536,456) HR, 0.36 (0.19–0.68) to HR, 0.81 (0.73–0.90)
Chen et al. [33] (2015) Taiwan HBV Population-based cohort study 71,824/1,735 HR, 0.34 (0.27–0.42) <0.001
Goh et al. [75] (2020) Korea HBV Retrospective cohort study 7,713/702 HR, 0.36 (0.19–0.68) 0.002
Wu et al. [76] (2014) Taiwan HBV Population-based cohort study 43,190/5,446 HR, 0.55 (0.47–0.63) <0.001
Tsan et al. [87] (2012) Taiwan HBV Population-based cohort study 33,413/1,021 HR, 0.47 (0.36–0.61) <0.001
Hsiang et al. [88] (2015) China HBV Cohort study 73,499/6,883 HR, 0.68 (0.48–0.97) 0.033
Tsan et al. [89] (2013) Taiwan HCV Population-based cohort study 260,864/27,883 HR, 0.53 (0.49–0.58) <0.001
Butt et al. [90] (2015) USA HCV Cohort study 7,248 HR, 0.52 (0.35–0.78) <0.001
Mohanty et al. [91] (2016) USA HCV Retrospective cohort study 2,747/173 HR, 0.42 (0.27–0.64) <0.001
Simon et al. [92] (2016) USA HCV Cohort study 9,135/239 HR, 0.85 (0.47–1.53) 0.580
Li et al. [93] (2020) HBV, HCV Meta-analysis 13 studies (519,707) RR, 0.54 (0.44–0.66) <0.001
Wong et al. [94] (2021) HBV, HCV, LC Meta-analysis 13 studies (1,742,260) HR, 0.57 (0.52–0.62) 0.060
Lee et al. [36] (2017) Taiwan NAFLD Population-based cohort study 18,080/41 HR, 0.29 (0.12–0.68) 0.005

HCC, hepatocellular carcinoma; CI, confidence interval; HBV, hepatitis B virus; HR, hazard ratio; HCV, hepatitis C virus; LC, liver cirrhosis; RR, risk ratio; NAFLD, non-alcoholic fatty liver disease.

Abbreviations

ALD
alcoholic liver disease
BMI
body mass index
CI
confidence interval
CLD
chronic liver disease
DM
diabetes mellitus
HBV
hepatitis B virus
HCC
hepatocellular carcinoma
HCV
hepatitis C virus
HDL
high-density lipoprotein
HR
hazard ratio
LC
liver cirrhosis
LDL
low-density lipoprotein
NAFLD
non-alcoholic fatty liver disease
NHIS-HEALS
National Health Insurance Service-Health Screening Cohort
RR
risk ratio
TC
total cholesterol
TG
triglyceride
WC
waist circumference

REFERENCES

1. Singal AG, Lampertico P, Nahon P. Epidemiology and surveillance for hepatocellular carcinoma: new trends. J Hepatol 2020;72:250-261.
crossref pmid pmc
2. Lombardi A, Grimaldi A, Zappavigna S, Misso G, Caraglia M. Hepatocarcinoma: genetic and epigenetic features. Minerva Gastroenterol Dietol 2018;64:14-27.
crossref pmid
3. 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.
pmid pmc
4. Simon TG, King LY, Chong DQ, Nguyen LH, Ma Y, VoPham T, et al. Diabetes, metabolic comorbidities, and risk of hepatocellular carcinoma: results from two prospective cohort studies. Hepatology 2018;67:1797-1806.
crossref pmid pmc pdf
5. Jinjuvadia R, Patel S, Liangpunsakul S. The association between metabolic syndrome and hepatocellular carcinoma: systemic review and meta-analysis. J Clin Gastroenterol 2014;48:172-177.
pmid
6. Inoue M, Noda M, Kurahashi N, Iwasaki M, Sasazuki S, Iso H, et al. Impact of metabolic factors on subsequent cancer risk: results from a large-scale population-based cohort study in Japan. Eur J Cancer Prev 2009;18:240-247.
crossref pmid
7. 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.
crossref pmid pmc
8. Ahlbom A, Alfredsson L. Interaction: a word with two meanings creates confusion. Eur J Epidemiol 2005;20:563-564.
crossref pmid pdf
9. Andersson T, Alfredsson L, Källberg H, Zdravkovic S, Ahlbom A. Calculating measures of biological interaction. Eur J Epidemiol 2005;20:575-579.
crossref pmid pdf
10. Yi SW, Choi JS, Yi JJ, Lee YH, Han KJ. Risk factors for hepatocellular carcinoma by age, sex, and liver disorder status: a prospective cohort study in Korea. Cancer 2018;124:2748-2757.
crossref pmid pdf
11. Wang P, Kang D, Cao W, Wang Y, Liu Z. Diabetes mellitus and risk of hepatocellular carcinoma: a systematic review and meta-analysis. Diabetes Metab Res Rev 2012;28:109-122.
crossref pmid
12. Wang C, Wang X, Gong G, Ben Q, Qiu W, Chen Y, et al. Increased risk of hepatocellular carcinoma in patients with diabetes mellitus: a systematic review and meta-analysis of cohort studies. Int J Cancer 2012;130:1639-1648.
crossref pmid
13. Cai C, Zeng J, Wu H, Shi R, Wei M, Gao Y, et al. Association between hepatitis B virus infection and diabetes mellitus: a meta-analysis. Exp Ther Med 2015;10:693-698.
crossref pmid pmc
14. Mantovani A, Petracca G, Beatrice G, Tilg H, Byrne CD, Targher G. Non-alcoholic fatty liver disease and risk of incident diabetes mellitus: an updated meta-analysis of 501 022 adult individuals. Gut 2021;70:962-969.
crossref pmid
15. Fabiani S, Fallahi P, Ferrari SM, Miccoli M, Antonelli A. Hepatitis C virus infection and development of type 2 diabetes mellitus: systematic review and meta-analysis of the literature. Rev Endocr Metab Disord 2018;19:405-420.
crossref pmid pdf
16. Kim K, Choi S, Park SM. Association of fasting serum glucose level and type 2 diabetes with hepatocellular carcinoma in men with chronic hepatitis B infection: a large cohort study. Eur J Cancer 2018;102:103-113.
crossref pmid
17. Campbell C, Wang T, McNaughton AL, Barnes E, Matthews PC. Risk factors for the development of hepatocellular carcinoma (HCC) in chronic hepatitis B virus (HBV) infection: a systematic review and meta-analysis. J Viral Hepat 2021;28:493-507.
crossref pmid pmc pdf
18. Hsu YC, Yip TC, Ho HJ, Wong VW, Huang YT, El-Serag HB, et al. Development of a scoring system to predict hepatocellular carcinoma in Asians on antivirals for chronic hepatitis B. J Hepatol 2018;69:278-285.
crossref pmid
19. Dyal HK, Aguilar M, Bartos G, Holt EW, Bhuket T, Liu B, et al. Diabetes mellitus increases risk of hepatocellular carcinoma in chronic hepatitis C virus patients: a systematic review. Dig Dis Sci 2016;61:636-645.
crossref pmid pdf
20. Ioannou GN, Green P, Lowy E, Mun EJ, Berry K. Differences in hepatocellular carcinoma risk, predictors and trends over time according to etiology of cirrhosis. PLoS One 2018;13:e0204412.
crossref pmid pmc
21. Huang YW, Wang TC, Yang SS, Lin SY, Fu SC, Hu JT, et al. Increased risk of hepatocellular carcinoma in chronic hepatitis C patients with new onset diabetes: a nation-wide cohort study. Aliment Pharmacol Ther 2015;42:902-911.
crossref pmid pdf
22. Arase Y, Kobayashi M, Suzuki F, Suzuki Y, Kawamura Y, Akuta N, et al. Effect of type 2 diabetes on risk for malignancies includes hepatocellular carcinoma in chronic hepatitis C. Hepatology 2013;57:964-973.
crossref pmid pdf
23. Ioannou GN, Green P, Kerr KF, Berry K. Models estimating risk of hepatocellular carcinoma in patients with alcohol or NAFLD-related cirrhosis for risk stratification. J Hepatol 2019;71:523-533.
crossref pmid pmc
24. Yang JD, Ahmed F, Mara KC, Addissie BD, Allen AM, Gores GJ, et al. Diabetes is associated with increased risk of hepatocellular carcinoma in patients with cirrhosis from nonalcoholic fatty liver disease. Hepatology 2020;71:907-916.
crossref pmid pmc pdf
25. Kanwal F, Kramer JR, Li L, Dai J, Natarajan Y, Yu X, et al. Effect of metabolic traits on the risk of cirrhosis and hepatocellular cancer in nonalcoholic fatty liver disease. Hepatology 2020;71:808-819.
crossref pmid pdf
26. Bertot LC, Jeffrey GP, de Boer B, MacQuillan G, Garas G, Chin J, et al. Diabetes impacts prediction of cirrhosis and prognosis by non-invasive fibrosis models in non-alcoholic fatty liver disease. Liver Int 2018;38:1793-1802.
crossref pmid pdf
27. Vilar-Gomez E, Calzadilla-Bertot L, Wai-Sun Wong V, Castellanos M, Aller-de la Fuente R, Metwally M, et al. Fibrosis severity as a determinant of cause-specific mortality in patients with advanced nonalcoholic fatty liver disease: a multi-national cohort study. Gastroenterology 2018;155:443-457.e17.
crossref pmid
28. Kawamura Y, Arase Y, Ikeda K, Seko Y, Imai N, Hosaka T, et al. Large-scale long-term follow-up study of Japanese patients with non-alcoholic fatty liver disease for the onset of hepatocellular carcinoma. Am J Gastroenterol 2012;107:253-261.
crossref pmid pdf
29. Yang C, Wan M, Lu Y, Yang X, Yang L, Wang S, et al. Associations between diabetes mellitus and the risk of hepatocellular carcinoma in Asian individuals with hepatitis B and C infection: systematic review and a meta-analysis of cohort studies. Eur J Cancer Prev 2022;31:107-116.
crossref pmid
30. Chen J, Song S, Li X, Bian D, Wu X. Association of metabolic traits with occurrence of nonalcoholic fatty liver disease-related hepatocellular carcinoma: a systematic review and meta-analysis of longitudinal cohort studies. Saudi J Gastroenterol 2022;28:92-100.
crossref pmid pmc
31. Hum J, Jou JH. The link between hepatitis C virus and diabetes mellitus: improvement in insulin resistance after eradication of hepatitis C virus. Clin Liver Dis (Hoboken) 2018;11:73-76.
crossref pmid pmc pdf
32. Targher G, Corey KE, Byrne CD, Roden M. The complex link between NAFLD and type 2 diabetes mellitus - mechanisms and treatments. Nat Rev Gastroenterol Hepatol 2021;18:599-612.
crossref pmid pdf
33. Chen CI, Kuan CF, Fang YA, Liu SH, Liu JC, Wu LL, et al. Cancer risk in HBV patients with statin and metformin use: a population-based cohort study. Medicine (Baltimore) 2015;94:e462.
pmid pmc
34. Hsu YC, Ho HJ, Lee TY, Huang YT, Wu MS, Lin JT, et al. Temporal trend and risk determinants of hepatocellular carcinoma in chronic hepatitis B patients on entecavir or tenofovir. J Viral Hepat 2018;25:543-551.
crossref pmid pdf
35. Yip TC, Wong VW, Chan HL, Tse YK, Hui VW, Liang LY, et al. Thiazolidinediones reduce the risk of hepatocellular carcinoma and hepatic events in diabetic patients with chronic hepatitis B. J Viral Hepat 2020;27:904-914.
crossref pmid pdf
36. Lee TY, Wu JC, Yu SH, Lin JT, Wu MS, Wu CY. The occurrence of hepatocellular carcinoma in different risk stratifications of clinically noncirrhotic nonalcoholic fatty liver disease. Int J Cancer 2017;141:1307-1314.
crossref pmid pdf
37. Kramer JR, Natarajan Y, Dai J, Yu X, Li L, El-Serag HB, et al. Effect of diabetes medications and glycemic control on risk of hepatocellular cancer in patients with nonalcoholic fatty liver disease. Hepatology 2022;75:1420-1428.
crossref pmid pmc pdf
38. Borena W, Strohmaier S, Lukanova A, Bjørge T, Lindkvist B, Hallmans G, et al. Metabolic risk factors and primary liver cancer in a prospective study of 578,700 adults. Int J Cancer 2012;131:193-200.
crossref pmid
39. Samanic C, Chow WH, Gridley G, Jarvholm B, Fraumeni JF Jr. Relation of body mass index to cancer risk in 362,552 Swedish men. Cancer Causes Control 2006;17:901-909.
crossref pmid pdf
40. Chen Y, Wang X, Wang J, Yan Z, Luo J. Excess body weight and the risk of primary liver cancer: an updated meta-analysis of prospective studies. Eur J Cancer 2012;48:2137-2145.
crossref pmid
41. Gupta A, Das A, Majumder K, Arora N, Mayo HG, Singh PP, et al. Obesity is independently associated with increased risk of hepatocellular cancer-related mortality: a systematic review and meta-analysis. Am J Clin Oncol 2018;41:874-881.
pmid pmc
42. Parr CL, Batty GD, Lam TH, Barzi F, Fang X, Ho SC, et al. Bodymass index and cancer mortality in the Asia-Pacific cohort studies collaboration: pooled analyses of 424,519 participants. Lancet Oncol 2010;11:741-752.
crossref pmid pmc
43. Oh SW, Yoon YS, Shin SA. Effects of excess weight on cancer incidences depending on cancer sites and histologic findings among men: Korea national health insurance corporation study. J Clin Oncol 2005;23:4742-4754.
crossref pmid
44. Batty GD, Barzi F, Huxley R, Chang CY, Jee SH, Jamrozik K, et al. Obesity and liver cancer mortality in Asia: the Asia Pacific cohort studies collaboration. Cancer Epidemiol 2009;33:469-472.
crossref pmid pmc
45. Yu MW, Shih WL, Lin CL, Liu CJ, Jian JW, Tsai KS, et al. Body-mass index and progression of hepatitis B: a population-based cohort study in men. J Clin Oncol 2008;26:5576-5582.
crossref pmid
46. Fu SC, Huang YW, Wang TC, Hu JT, Chen DS, Yang SS. Increased risk of hepatocellular carcinoma in chronic hepatitis B patients with new onset diabetes: a nationwide cohort study. Aliment Pharmacol Ther 2015;41:1200-1209.
crossref pmid
47. Loomba R, Yang HI, Su J, Brenner D, Iloeje U, Chen CJ. Obesity and alcohol synergize to increase the risk of incident hepatocellular carcinoma in men. Clin Gastroenterol Hepatol 2010;8:891-898.e8982.
crossref pmid
48. Chao LT, Wu CF, Sung FY, Lin CL, Liu CJ, Huang CJ, et al. Insulin, glucose and hepatocellular carcinoma risk in male hepatitis B carriers: results from 17-year follow-up of a population-based cohort. Carcinogenesis 2011;32:876-881.
crossref pmid pmc
49. Kim K, Choi S, Park SM. Association of high body mass index and hepatocellular carcinoma in patients with chronic hepatitis B virus infection: a Korean population-based cohort study. JAMA Oncol 2018;4:737-739.
crossref pmid pmc
50. Veldt BJ, Chen W, Heathcote EJ, Wedemeyer H, Reichen J, Hofmann WP, et al. Increased risk of hepatocellular carcinoma among patients with hepatitis C cirrhosis and diabetes mellitus. Hepatology 2008;47:1856-1862.
crossref pmid
51. Arano T, Nakagawa H, Tateishi R, Ikeda H, Uchino K, Enooku K, et al. Serum level of adiponectin and the risk of liver cancer development in chronic hepatitis C patients. Int J Cancer 2011;129:2226-2235.
crossref pmid
52. Yang JD, Mohamed HA, Cvinar JL, Gores GJ, Roberts LR, Kim WR. Diabetes mellitus heightens the risk of hepatocellular carcinoma except in patients with hepatitis C cirrhosis. Am J Gastroenterol 2016;111:1573-1580.
crossref pmid pmc pdf
53. Lok AS, Seeff LB, Morgan TR, di Bisceglie AM, Sterling RK, Curto TM, et al. Incidence of hepatocellular carcinoma and associated risk factors in hepatitis C-related advanced liver disease. Gastroenterology 2009;136:138-148.
crossref pmid pmc
54. McMahon BJ, Bruden D, Townshend-Bulson L, Simons B, Spradling P, Livingston S, et al. Infection with hepatitis C virus genotype 3 is an independent risk factor for end-stage liver disease, hepatocellular carcinoma, and liver-related death. Clin Gastroenterol Hepatol 2017;15:431-437.e2.
crossref pmid pmc
55. Ohki T, Tateishi R, Sato T, Masuzaki R, Imamura J, Goto T, et al. Obesity is an independent risk factor for hepatocellular carcinoma development in chronic hepatitis C patients. Clin Gastroenterol Hepatol 2008;6:459-464.
crossref pmid
56. Hung CH, Lee CM, Wang JH, Hu TH, Chen CH, Lin CY, et al. Impact of diabetes mellitus on incidence of hepatocellular carcinoma in chronic hepatitis C patients treated with interferon-based antiviral therapy. Int J Cancer 2011;128:2344-2352.
crossref pmid pdf
57. Nair S, Mason A, Eason J, Loss G, Perrillo RP. Is obesity an independent risk factor for hepatocellular carcinoma in cirrhosis? Hepatology 2002;36:150-155.
crossref pmid pdf
58. Pais R, Lebray P, Rousseau G, Charlotte F, Esselma G, Savier E, et al. Nonalcoholic fatty liver disease increases the risk of hepatocellular carcinoma in patients with alcohol-associated cirrhosis awaiting liver transplants. Clin Gastroenterol Hepatol 2015;13:992-999.e2.
crossref pmid
59. Grimaudo S, Pipitone RM, Pennisi G, Celsa C, Cammà C, Di Marco V, et al. Association between PNPLA3 rs738409 C>G variant and liver-related outcomes in patients with nonalcoholic fatty liver disease. Clin Gastroenterol Hepatol 2020;18:935-944.e3.
crossref pmid
60. N’Kontchou G, Paries J, Htar MT, Ganne-Carrie N, Costentin L, Grando-Lemaire V, et al. Risk factors for hepatocellular carcinoma in patients with alcoholic or viral C cirrhosis. Clin Gastroenterol Hepatol 2006;4:1062-1068.
crossref pmid
61. Archambeaud I, Auble H, Nahon P, Planche L, Fallot G, Faroux R, et al. Risk factors for hepatocellular carcinoma in Caucasian patients with non-viral cirrhosis: the importance of prior obesity. Liver Int 2015;35:1872-1876.
crossref pmid pdf
62. Brichler S, Nahon P, Zoulim F, Layese R, Bourcier V, Audureau E, et al. Non-virological factors are drivers of hepatocellular carcinoma in virosuppressed hepatitis B cirrhosis: results of ANRS CO12 CirVir cohort. J Viral Hepat 2019;26:384-396.
crossref pmid pdf
63. Ioannou GN, Splan MF, Weiss NS, McDonald GB, Beretta L, Lee SP. Incidence and predictors of hepatocellular carcinoma in patients with cirrhosis. Clin Gastroenterol Hepatol 2007;5:938-945 945.e1-e4.
crossref pmid
64. Ito T, Ishigami M, Ishizu Y, Kuzuya T, Honda T, Ishikawa T, et al. Serum nutritional markers as prognostic factors for hepatic and extrahepatic carcinogenesis in Japanese patients with nonalcoholic fatty liver disease. Nutr Cancer 2020;72:884-891.
crossref pmid
65. Rahmani J, Kord Varkaneh H, Kontogiannis V, Ryan PM, Bawadi H, Fatahi S, et al. Waist circumference and risk of liver cancer: a systematic review and meta-analysis of over 2 million cohort study participants. Liver Cancer 2020;9:6-14.
crossref pmid pmc pdf
66. Fan R, Niu J, Ma H, Xie Q, Cheng J, Rao H, et al. Association of central obesity with hepatocellular carcinoma in patients with chronic hepatitis B receiving antiviral therapy. Aliment Pharmacol Ther 2021;54:329-338.
crossref pmid pdf
67. Lee J, Yoo SH, Sohn W, Kim HW, Choi YS, Won JH, et al. Obesity and hepatocellular carcinoma in patients receiving entecavir for chronic hepatitis B. Clin Mol Hepatol 2016;22:339-349.
crossref pmid pmc pdf
68. Osaki Y, Taniguchi S, Tahara A, Okamoto M, Kishimoto T. Metabolic syndrome and incidence of liver and breast cancers in Japan. Cancer Epidemiol 2012;36:141-147.
crossref pmid
69. Häggström C, Jonsson H, Bjørge T, Nagel G, Manjer J, Ulmer H, et al. Linear age-course effects on the associations between body mass index, triglycerides, and female breast and male liver cancer risk: an internal replication study of 800,000 individuals. Int J Cancer 2020;146:58-67.
crossref pmid pdf
70. Cho Y, Cho EJ, Yoo JJ, Chang Y, Chung GE, Jeong SM, et al. Association between lipid profiles and the incidence of hepatocellular carcinoma: a nationwide population-based study. Cancers (Basel) 2021;13:1599.
crossref pmid pmc
71. Yi SW, Kim SH, Han KJ, Yi JJ, Ohrr H. Higher cholesterol levels, not statin use, are associated with a lower risk of hepatocellular carcinoma. Br J Cancer 2020;122:630-633.
crossref pmid pmc pdf
72. Sinn DH, Kang D, Cho SJ, Paik SW, Guallar E, Cho J, et al. Risk of hepatocellular carcinoma in individuals without traditional risk factors: development and validation of a novel risk score. Int J Epidemiol 2020;49:1562-1571.
crossref pmid pdf
73. Nderitu P, Bosco C, Garmo H, Holmberg L, Malmström H, Hammar N, et al. The association between individual metabolic syndrome components, primary liver cancer and cirrhosis: a study in the Swedish AMORIS cohort. Int J Cancer 2017;141:1148-1160.
crossref pmid pdf
74. Chiang CH, Lee LT, Hung SH, Lin WY, Hung HF, Yang WS, et al. Opposite association between diabetes, dyslipidemia, and hepatocellular carcinoma mortality in the middle-aged and elderly. Hepatology 2014;59:2207-2215.
crossref pmid
75. Goh MJ, Sinn DH, Kim S, Woo SY, Cho H, Kang W, et al. Statin use and the risk of hepatocellular carcinoma in patients with chronic hepatitis B. Hepatology 2020;71:2023-2032.
crossref pmid pdf
76. Wu CY, Lin JT, Ho HJ, Su CW, Lee TY, Wang SY, et al. Association of nucleos(t)ide analogue therapy with reduced risk of hepatocellular carcinoma in patients with chronic hepatitis B: a nationwide cohort study. Gastroenterology 2014;147:143-151.e5.
crossref pmid
77. Tan Y, Zhang X, Zhang W, Tang L, Yang H, Yan K, et al. The influence of metabolic syndrome on the risk of hepatocellular carcinoma in patients with chronic hepatitis B infection in mainland China. Cancer Epidemiol Biomarkers Prev 2019;28:2038-2046.
crossref pmid pdf
78. Arain SQ, Talpur FN, Channa NA, Ali MS, Afridi HI. Serum lipid profile as a marker of liver impairment in hepatitis B cirrhosis patients. Lipids Health Dis 2017;16:51.
crossref pmid pmc pdf
79. Vere CC, Streba CT, Streba L, Rogoveanu I. Lipid serum profile in patients with viral liver cirrhosis. Med Princ Pract 2012;21:566-568.
crossref pmid pdf
80. Jiang J, Nilsson-Ehle P, Xu N. Influence of liver cancer on lipid and lipoprotein metabolism. Lipids Health Dis 2006;5:4.
crossref pmid pmc pdf
81. Chrostek L, Supronowicz L, Panasiuk A, Cylwik B, Gruszewska E, Flisiak R. The effect of the severity of liver cirrhosis on the level of lipids and lipoproteins. Clin Exp Med 2014;14:417-421.
crossref pmid pmc pdf
82. Janičko M, Veselíny E, Leško D, Jarčuška P. Serum cholesterol is a significant and independent mortality predictor in liver cirrhosis patients. Ann Hepatol 2013;12:581-587.
pmid
83. Ghadir MR, Riahin AA, Havaspour A, Nooranipour M, Habibinejad AA. The relationship between lipid profile and severity of liver damage in cirrhotic patients. Hepat Mon 2010;10:285-288.
pmid pmc
84. Bassani L, Fernandes SA, Raimundo FV, Harter DL, Gonzalez MC, Marroni CA. Lipid profile of cirrhotic patients and its association with prognostic scores: a cross-sectional study. Arq Gastroenterol 2015;52:210-215.
crossref pmid
85. Tauseef A, Zafar M, Rashid B, Thirumalareddy J, Chalfant V, Farooque U, et al. Correlation of fasting lipid profile in patients with chronic liver disease: a descriptive cross-sectional study in Tertiary Care Hospital. Cureus 2020;12:e11019.
crossref pmid pmc
86. Choe JW, Hyun JJ, Kim B, Han KD. Influence of metabolic syndrome on cancer risk in HBV carriers: a nationwide population based study using the national health insurance service database. J Clin Med 2021;10:2401.
crossref pmid pmc
87. Tsan YT, Lee CH, Wang JD, Chen PC. Statins and the risk of hepatocellular carcinoma in patients with hepatitis B virus infection. J Clin Oncol 2012;30:623-630.
crossref pmid
88. Hsiang JC, Wong GL, Tse YK, Wong VW, Yip TC, Chan HL. Statin and the risk of hepatocellular carcinoma and death in a hospital-based hepatitis B-infected population: a propensity score landmark analysis. J Hepatol 2015;63:1190-1197.
crossref pmid
89. Tsan YT, Lee CH, Ho WC, Lin MH, Wang JD, Chen PC. Statins and the risk of hepatocellular carcinoma in patients with hepatitis C virus infection. J Clin Oncol 2013;31:1514-1521.
crossref pmid
90. Butt AA, Yan P, Bonilla H, Abou-Samra AB, Shaikh OS, Simon TG, et al. Effect of addition of statins to antiviral therapy in hepatitis C virus-infected persons: results from ERCHIVES. Hepatology 2015;62:365-374.
crossref pmid
91. Mohanty A, Tate JP, Garcia-Tsao G. Statins are associated with a decreased risk of decompensation and death in veterans with hepatitis C-related compensated cirrhosis. Gastroenterology 2016;150:430-440.e1.
crossref pmid pmc
92. Simon TG, Bonilla H, Yan P, Chung RT, Butt AA. Atorvastatin and fluvastatin are associated with dose-dependent reductions in cirrhosis and hepatocellular carcinoma, among patients with hepatitis C virus: results from ERCHIVES. Hepatology 2016;64:47-57.
crossref pmid pmc
93. Li X, Sheng L, Liu L, Hu Y, Chen Y, Lou L. Statin and the risk of hepatocellular carcinoma in patients with hepatitis B virus or hepatitis C virus infection: a meta-analysis. BMC Gastroenterol 2020;20:98.
crossref pmid pmc pdf
94. 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;55:615-623.
pmid
95. Friedman GD, Achacoso N, Fireman B, Habel LA. Statins and reduced risk of liver cancer: evidence for confounding. J Natl Cancer Inst 2016;108:djw109.
pmid pmc
96. Jeong GH, Lee KH, Kim JY, Eisenhut M, Kronbichler A, van der Vliet HJ, et al. Effect of statin on cancer incidence: an umbrella systematic review and meta-analysis. J Clin Med 2019;8:819.
crossref pmid pmc
97. Cholesterol Treatment Trialists’ (CTT) Collaboration. 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.
crossref pmid pmc
98. Yu MW, Lin CL, Liu CJ, Yang SH, Tseng YL, Wu CF. Influence of metabolic risk factors on risk of hepatocellular carcinoma and liver-related death in men with chronic hepatitis B: a large cohort study. Gastroenterology 2017;153:1006-1017.e5.
crossref pmid
99. Yi SW, Ohrr H, Shin SA, Yi JJ. Sex-age-specific association of body mass index with all-cause mortality among 12.8 million Korean adults: a prospective cohort study. Int J Epidemiol 2015;44:1696-1705.
crossref pmid pmc

Editorial Office
The Korean Association for the Study of the Liver
Room A1210, 53 Mapo-daero(MapoTrapalace, Dowha-dong), Mapo-gu, Seoul, 04158, Korea
TEL: +82-2-703-0051   FAX: +82-2-703-0071    E-mail: kasl@kams.or.kr
Copyright © The Korean Association for the Study of the Liver.         
COUNTER
TODAY : 2967
TOTAL : 1738975
Close layer