Download PDF
Review  |  Open Access  |  22 Sep 2024

Impact of occult hepatitis B virus infection and high-fat diet on hepatocellular carcinoma development

Views: 78 |  Downloads: 22 |  Cited:  0
Hepatoma Res 2024;10:38.
10.20517/2394-5079.2024.85 |  © The Author(s) 2024.
Author Information
Article Notes
Cite This Article

Abstract

In the areas where viral infections are more common, the incidence of hepatocellular carcinoma (HCC) has declined. This is largely attributed to the availability of vaccines against hepatitis B virus (HBV), antiviral treatments, and a change in diet with healthier foods. However, in the Western world, the HCC incidence rate has risen steadily, probably due to an increased prevalence of metabolic dysfunction-associated fatty liver disease (MAFLD), a condition that has increasingly been considered an important risk factor for HCC. Despite this, in 2019, 41% of HCC cases were diagnosed globally and a majority of diagnosed cases from mainland China were related to HBV. Some HCC cases tested negative for hepatitis B surface antigen (HBsAg) but tested positive for HBV-DNA in blood. This is considered an occult HBV infection (OBI). OBI is related to various mutations in the viral genome, which modifies host immunity, subsequently leading to the long-term persistence of cccDNA in the nucleus of infected hepatocytes. Many OBIs are associated with HBV variants carrying mutations in preS/S genomic regions, which occur either spontaneously or as a result of antiviral treatments. OBIs are also frequently reported in HBV-vaccinated individuals. HBV variants post-HBV vaccination carry mutations in the preS area. This review discusses the relationship between preS variant-related OBIs and the development of HCC in the context of a high-fat diet, one of the preventable behavioral risk factors for MAFLD.

Keywords

Occult HBV infection, preS mutations, hepatocellular carcinoma, high-fat diet, hepatitis B vaccination, metabolic dysfunction associated fatty liver disease

INTRODUCTION

The age-standardized incidence rate of hepatocellular carcinoma (HCC) has decreased consistently in Oceania, sub-Saharan Africa, and Asia, such as in China[1]. This trend was greatly attributed to the strategies for reducing established HCC risk factors. It includes immunization against hepatitis B virus (HBV) using HepB vaccine[2-4], decreased exposure to carcinogen aflatoxins[5], treatment of patients with HBV using nucleos(t)ide analogs[6], and treatment of patients with hepatitis C virus (HCV) using direct-acting antiviral agents[7]. The vegetable-based dietary pattern was observed to reduce the risk of liver cancer, particularly in individuals with a history of chronic liver disease[8,9], and this pattern is considered increasingly more acceptable in China[10].

The HCC incidence rates have increased steadily in the Western world. It is likely associated with the increased prevalence of metabolic dysfunction-associated with fatty liver disease (MAFLD), formerly known as non-alcoholic fatty liver disease (NAFLD). MAFLD is an overarching term describing heterogenous liver diseases associated with known metabolic dysfunctions. It is associated with various factors such as age, gender, genetic predisposition, microbiota, hormonal status, and behavioral factors, including dietary patterns[11]. Epidemiologically, a major HCC etiology is a shift from viral to metabolic liver diseases[12-14]. The MAFLD prevalence has consistently increased in Asia, such as China, over the last two decades[15].

A study indicated that 41.0% of HCC cases diagnosed globally in 2019 were related to HBV[6]. In Mailand of China, 84.4% of diagnosed HCC cases during 2003-2020 were related to HBV. Some of these cases were serologically negative for HBV surface antigen (HBsAg) but positive for HBV-DNA[16]. A global systematic review and meta-analysis of 22 studies that included over 56,000 HCC individuals indicated that 49% of all HCC cases and 40% of HBV-related HCC cases have MAFLD, but only 12% of all HCC cases were associated with solitary MAFLD[14]. An 11-year follow-up study on the Chinese population with 13,032 company employees reported that the prevalence of NAFLD was 17.2% in 2006, which increased to 32.4% in 2016. However, the main clinical outcomes of NAFLD in this cohort population were diabetes, hypertension, and hyperuricemia. No patients with NAFLD progressed into HCC, except for one male patient who developed cirrhosis over the 11 years[17]. HepB vaccine is prophylactic and has no effect on established HBV infection. Antiviral therapies with nucleot(s)ide analogs inhibit viral DNA synthesis and delay the occurrence of HBV-related diseases including HCC but cannot cure HBV infections[18]. Global HBV prevalence in 2022 was estimated to be at 3.2%, corresponding to 257.5 million (216.6-316.4) HBsAg-positive individuals[19]. Primary prevention based on etiology offers the best cost-effective method for decreasing HCC incidence. Thus, it is important to understand the pathophysiology of HCC development in individuals with pre-existing HBV infection in the context of preventable behavioral risk factors that can lead to MAFLD, such as dietary patterns.

HBV LIFE CYCLE

HBV life cycle is described briefly here as it has been reviewed in detail elsewhere[20,21]. Infectious HBV viral particles that contain the genome of a 3.2 kb relaxed circular (rc) partially double-stranded DNA (rcDNA) attach first to heparan sulfate proteoglycans. Then, they enter hepatocytes via bile acid transporter sodium taurocholate co-transporting polypeptide (NTCP) through an interaction with the preS1 domain at the large envelop protein (L-HBsAg)[22]. Upon entry into the hepatocytes, the HBV viral particle is uncoated and nucleocapsids move to the hepatocyte nuclear pore complex, where rcDNA is released into the nucleus. By using host factors, rcDNA is converted into covalently closed circular DNA (cccDNA)[23,24], which serves as the template for transcription of viral pregenomic RNA (pgRNA) and viral mRNAs, including 3.5, 2.4, 2.1, and 0.7 kb HBV RNA. The 0.7 kb HBV RNA is transcribed from the X gene promoter and translated into a multifunctional HBx protein, which is known to activate viral and host gene transcriptions relating to hepatocarcinogenesis[20]. From 1,783 nt of the HBV genome, a 3.5 kb RNA is transcribed and translated to precore polypeptide which is trafficked into hepatocyte endoplasmic reticulum (ER) via a signal peptide in the N-terminal region. Within ER, the signal peptide and 34 amino acids (AA) of the C-terminal domain are removed, and hepatitis B e antigen (HBeAg) is matured to release from hepatocytes. From 1,818 nt of the HBV genome, a 3.5 kb pgRNA is transcribed and translated to HBV core protein (HBcAg) and viral polymerase (P), and encapsidated with P protein by the core protein dimers to form nucleocapsids particles. Reverse transcription is initiated within the particles and conducted by the P protein to generate rcDNA, or double-strand linear DNA (dslDNA) in some situations. The dslDNA is preferably integrated into the host genome. Some nucleocapsids are shuttled back to the nucleus to maintain a relatively stable pool of cccDNA and some are enveloped by L-HBsAg, which is translated from 2.4 kb HBV RNA. Meanwhile, middle (M-HBsAg) and small (S-HBsAg) envelop proteins are translated from 2.1 kb RNA. Synthesized L-, M-, and S-HBsAg are sorted into the ER for processing and then transferred to the Golgi apparatus. Through two cytoplasmic matrix domains, one located at the preS1-preS2 boundary and another at the C-terminal of S-HBsAg, the L-HBsAg contacts with nucleocapsids to pack tightly and drive the inward budding. The inward-budded nucleocapsids are processed by endosomal sorting complexes required for transport to catalyze the membrane fission and subsequently release outside the cells as virions[25,26]. The domains of L-HBsAg at preS1 and S are required for the generation of infectious viral particles (42 nm in diameter) and spherical particles (20 nm in diameter). The spherical particles contain no HBV genome, which mainly comprises S-HBsAg[20,26].

HCC RISK IN PATIENTS WITH CHRONIC HBV INFECTION COMBINED WITH METABOLIC DYSFUNCTION

MAFLD is a phenotype with complex and diverse causes[11]. Currently, HBV infection in combination with NAFLD leading to the progression of HCC remains unconfirmed. A previous review article summarized the interaction between HBV and NAFLD and the progression of the disease[27]. A study based on a healthy Korean adult cohort reported that HBsAg seropositivity was associated with a lower risk of developing NAFLD[28]. Most studies in the Chinese population also suggest that HBV infection reduces the NAFLD incidence[27]. In an HBV-immunocompetent mouse model with HBV genotype B infection, HBV replication was even attenuated in the context of high-fat diet-induced hepatic steatosis[29]. Many individuals with hepatic steatosis exhibit no liver inflammatory injury, and an individual with NAFLD can oscillate between steatosis and steatohepatitis even over a short timeframe[30].

Most HCCs develop with a background of chronic liver injury, hepatic inflammation, and liver fibrosis. Clinical evidence and a few animal models suggested that hepatic steatosis in patients with chronic hepatitis B (CHB) might reduce HCC risks. Virus per se is the most important factor in the development of HBV-related HCC. Among the HBsAg-positive population, the HBeAg positivity significantly increases HCC risk[31]. However, among CHB patients with steatosis, the portions of serum HBeAg positivity and HBV viremia reduced, and the positive staining of intrahepatic HBsAg and hepatitis B core antigen (HBcAg) decreased in liver tissue[32]. A meta-analysis in CHB patients found that hepatic steatosis was not significantly associated with liver fibrosis[33]. CHB patients with MAFLD increased the overall risk of liver fibrosis, although atherosclerosis was protected[34]. A prospective study conducted on CHB patients from Hong Kong reported that reduced hepatic steatosis significantly increased the HCC risk[35]. Analyzing the database of the Korean National Health Insurance Service, which included 1,504,880 adults with HBV infection, it was reported that metabolic syndromes increased the risk for most malignancies but were negatively associated with HCC[36]. However, the 5-year follow-up results from 10,546 treatment-naive CHB patients reported that hepatic steatosis and metabolic dysfunction displayed distinct effects on HCC development[37]. A study based on 5,373 male Taiwanese civil servants indicated that the cumulative incidence of HCC and liver-related deaths are significantly different in HBV carriers with different metabolic risk factors. Metabolic risk factors and insulin resistance presented the largest effect on HCC risk in CHB patients. More importantly, the association between HCC and lower levels of serum HBV-DNA (less than 10,000 copies/mL) was stronger compared to that in patients with higher levels of HBV-DNA[38]. HBV retention within hepatocytes might play an important role in promoting HCC development [Figure 1]. Indeed, reduced HBV secretion from hepatocytes promotes HCC development, such as the infection of preS/S variants that lead to an imbalance in HBV replication and secretion from hepatocytes[39-41].

Impact of occult hepatitis B virus infection and high-fat diet on hepatocellular carcinoma development

Figure 1. Schematic representation of the impact of HBV infection on HCC development. (1) Hepatic steatosis with high-fat diet can reduce HBV replication, increase HBsAg secretion, and inhibit liver inflammation, and probably reduce the risk of HCC; (2) Some types of HBV preS/S variants can induce an imbalanced HBV production and secretion, leading to ER stress-dependent and/or independent signals to promote HCC development (Reviewed by Pollicino et al.[40]); (3) PreS mutations related to OBI post HepB vaccination occur. The OBI-related variants display the alteration of amino acid properties in large-S protein and result in HBsAg retention in hepatocyte ER. PreS variant-infected hepatocytes elevate the generation of abnormal ceramides after overload of free fatty acids. The abnormal ceramides, together with HBV proteins, co-activate NLRP3 inflammasome to increase the production of inflammatory cytokines from liver macrophages, increasing the risk of HCC. HBV: Hepatitis B virus; HCC: hepatocellular carcinoma; HBsAg: hepatitis B surface antigen; ER: endoplasmic reticulum; OBI: occult hepatitis B infection; NLRP3: NOD-like receptor protein 3.

OCCULT HBV INFECTION POST HepB VACCINATION AT INFANCY

The definition of occult HBV infection (OBI) was updated in October 2018. It is now defined as the presence of replication-competent HBV (the episomal cccDNA) in liver and/or HBV-DNA in blood of individuals who are seronegative for HBsAg with currently available assays. The pathogenesis of OBI is related to the long-term persistence of cccDNA in the nucleus of infected hepatocytes[42]. The infection with HBV S variants carrying mutations in the S gene generates modified HBsAg, which cannot be recognized by some of the commercially available HBsAg assays. The infection with HBV variants carrying mutations in the promoter area of S-gene, including preS1 and/or preS2 area, and the HBV splice variants can reduce HBsAg production/secretion that falls below the detection threshold of currently available assays[42-46]. In addition to viral factors, host immunity also plays an important role in controlling virus replication in the “occult” status[42,47]. The gold standard OBI diagnostic method is the detection of HBV-DNA in liver. However, commonly available methods use blood to detect HBV-DNA. Detection of anti-HBc in blood is often used as a surrogate technique[42]. Integrated HBV in the genome of hepatocytes, which is not replication-competent, might be detected in the blood when cell death occurs after spontaneous or treatment-induced HBsAg clearance. The true OBI should contain the replication-competent HBV-DNA[42]. An assay of over-gap PCR amplification can identify the presence of replication-competent HBV-DNA in blood (rcDNA) and liver tissues (cccDNA), distinguishing the replication-competent HBV-DNA from integrated HBV-DNA[48].

HepB vaccine contains only the small S-HBsAg and does not include the preS1 domain that presents in the infectious virion[49]. Since the implementation of HepB vaccination to Chinese infants in 1992, the HBsAg-positive rate has dropped from 10% to 0.32%, 0.94%, and 4.38% for age groups of 1-4 years, 5-14 years, and 15-29 years, respectively[50]. After 10-15 years of vaccination, the vaccination conferred anti-HBs levels waned or reached undetectable levels in a vast majority of individuals[4,51,52]. A study involving 215,627 Chinese children who received HepB vaccination reported that the HBsAg-positive rate was 0.55% (ranging from 0.30%-0.64%) among those aged 1-9 years, and it increased to 1.40% (ranging from 1.00%-2.04%) in age groups older than 9 years. The positive rate of anti-HBc stayed relatively stable (5.69%) between 1-10 years but increased to 7.80% for age groups of 11-16 years[53]. Among individuals immunized with the HepB vaccine, HBV-DNA was frequently detected in blood samples with serological markers indicating HBsAg-negative but anti-HBc-positive status, suggesting the presence of OBI[43,54-57]. HBV rcDNA in blood is in the form of replication-component HBV[20]. To confirm the presence of true OBI, the method of over-gap PCR amplification was used[48]. It reiterated the presence of HBV rcDNA in the blood of few HepB-vaccinated young adults who tested negative for HBsAg but positive for anti-HBc[48,57]. The serum level of rcDNA among the HepB-vaccinated young adults with OBI was between 100 to 1,000 IU/mL[43,57].

HepB vaccination significantly reduced the risk of HCC by lowering the seroprevalence of HBsAg[3,4,58]. However, the increasing amount of evidence reveals that the individuals who completed the HepB vaccination series during infancy have an increased risk of HBV infection as they age and the infection can be chronic[43,52,53,59]. A meta-analysis has reported that OBI is significantly associated with an increased risk of HCC[42,60]. Wong reported that 24 of 33 cryptogenic HCC patients (73%) were related to OBI. Among them, HBV was detected more in the non-tumorous than the tumorous tissues. When compared with the HCC patients with overt chronic hepatitis B, intrahepatic pgRNA levels were detected at lower levels. The investigators hypothesized that HBV integration was the likely cause of HCC in OBI patients[61]. In cases of OBI, HBV maintains its pro-oncogenic properties[39]. However, the hepatocarcinogenesis remains to be elucidated.

HepB-VACCINATED INDIVIDUALS WITH OBI WERE FREQUENTLY ASSOCIATED WITH VARIANTS CARRYING MUTATIONS IN THE HBV preS REGION

Some “vaccine-selected escape mutants”, in which one amino acid is replaced by another within the “a” determinant, including G145R, have been identified in children receiving the HepB vaccine and they generated anti-HBs[62,63]. In 2009, Mu reported that 5/46 (10.9%) of children were serologically HBsAg-negative but HBV-DNA positive as tested using nested PCR in at least two regions. A low titer of HBV-DNA was detected in five children. Analysis of the S-gene sequencing found no G145R, but C139S vaccine escape mutant was detected. Variations and deletions were more common in the preS region of these OBI isolates[55]. In 2010, Xu reported that 124 out of 2,919 (4.2%) young adults receiving the HepB vaccine at infancy were HBsAg-negative but anti-HBs and anti-HBc-positive. Among these 124 individuals, 37 (29.8%) had HBV-DNA at the levels of 500-10,000 copies/mL and 73 (58.9%) had it below 500 copies/mL. After analysis of 41 HBV isolates, the mutations of “a” epitope in S-HBsAg were found to be few but more frequent in the preS region. All the sequences were recorded in GenBank (from HM191578 to HM191618). Alignment analysis showed that most isolates carried mutations that altered the properties of encoded AA[43,52,57,64]. Most of those were seropositive for anti-HBs and carried the mutations at the preS region, particularly in the preS1 area[57].

HEPATOCYTE INFECTION OF preS VARIANTS CAUSES HBsAg RETENTION WITHIN ER, GENERATING ABNORMAL CERAMIDES

In L-HBsAg, the domains located at the preS1 and S region are required for the generation of infectious viral particles and for HBsAg secretion[26]. Pollicino reported that the ratio of HBsAg to HBV-DNA concentrations was significantly lower in patients with preS/S gene variants, which included a 183-nucleotide deletion within the preS1 region, the deletion of preS2 start codon, and an anormal stop signal within the S gene[41]. Infection of HepG2 cells with these three specified types of specified preS-S gene variants resulted in the reduction of HBsAg secretion, retention of envelope proteins in the ER, and decreased efficiency in virion secretion, alongside the significantly increased accumulation of cccDNA in the nucleus. Both experimental data and human studies show that these specified mutations in the preS/S gene cause HBsAg retention within the hepatocyte ER, resulting in ER stress[41]. Infection of preS/S variants has been reported to be associated with fulminant hepatitis, fibrosing cholestatic hepatitis, cirrhosis, and HCC development via various mechanisms related to ER stress[40].

PreS-S of HBV from 35 HCC patients related to OBI were sequenced and recognized to carry extensive mutations in the large S area. Most variants carried mutations with altered AA properties of L-HBsAg, leading to the conversion of hydrophobic properties to hydrophilic ones, and vice versa, along with charge modifications[65]. The mutations of HBV isolated from OBI-related HCC were similar to those detected in people who received HepB vaccination during infancy[43,57]. The preS1 domain of L-HBsAg can be either projected toward cytoplasm or oriented toward ER lumen[20,26]. To address the effects of preS mutations, which changed AA properties of L-HBsAg on infected hepatocytes, the mtPreS1 or mtPreS2 plasmids were generated, which contained 1.3 × HBV genome. As a control, the plasmid of Ref-HBV was generated based on an HBV that was isolated from an HCC patient with overt HBsAg-positive HBV infection. When the hepatocytes were infected with the two types of preS variants, HBsAg secretion from hepatocytes reduced compared to the cells infected with Ref-HBV. HBsAg retention was mainly detected in ER compartment.

Hepatocyte ER is intricately linked to a wide spectrum of cellular functions. It is a critical component in maintaining and restoring metabolic health. Therefore, unsolved ER stress can lead to aberrant metabolism, organelle dysfunction, insulin resistance, and inflammation[66]. Three ER transmembrane proteins have been described as ER stress sensors: (1) activating transcription factor 6 (ATF6), (2) inositol-requiring enzyme 1α (IRE1α), and (3) PRKR-like ER kinase (PERK). Upon activation, multiple mechanisms are involved for reinstating the ER homeostasis, such as unfolded protein response (UPR), ER-associated protein degradation (ERAD), or autophagy[66]. In responding to HBsAg retention, the IRE1α-XBP1 pathway, which is key to lipid metabolic homeostasis[66], was found to be significantly activated in hepatocytes[65]. Abnormal accumulation of ceramides is a hallmark of metabolism-related disorders, and the ceramides exert markedly different effects depending on amide-linked fatty acid chain lengths[67]. Using HPLC-MS/MS, we determined the amounts and species of ceramides associated with various lengths of fatty acyl chains. HBsAg retention within ER, caused by preS variations due to AA property alterations in HBV envelop proteins, disturbed hepatocyte lipid metabolism homeostasis. This led to the generation of abnormal amounts and species of ceramides, mainly the C16:0 ceramide[65]. Modern diets are rich in unhealthy fat and ingestion of excessive fat is one of the major risk factors for MAFLD development. In a cell culture system, we observed that loading free fatty acids into hepatocytes infected with hepatitis B virus (HBV), particularly those with preS variants, significantly increased the production of abnormal ceramides[65].

INFECTION OF HBV preS VARIANTS WITH HIGH-FAT DIET SYNERGISTICALLY PROMOTED AUTOCHTHONOUS HCC RELATING TO LIVER INFLAMMATION MEDIATED BY CERAMIDE AND HBV ANTIGENS IN MURINE MODEL

Individuals infected with HBV variants mutated in the preS-S genomic region are frequent. These variants might result from Hep vaccination or antiviral treatments, or also may occur spontaneously. As discussed above, some HBV variants carry mutations that modify AA properties of L-HBsAg, leading to HBsAg retention in hepatic ER. The impacts of these types of variant infections on HCC development and their underlying mechanisms were examined in male C57BL/6J mice. The mice that were infected with the same amount of replicating plasmids consisting of 1.3 × HBV genome of mtPreS1, or mtPreS2, or Ref-HBV were followed up by giving normal chow (NC) or high-fat (HF) diet for five weeks. A higher ratio of liver weight to body weight was observed in HBV-infected mice, particularly among those infected with the variants at PreS1 or PreS2 regions. Notably, an elevated ratio of liver weight to body weight was also detected in NC-diet mice after infection of preS variants. Intrinsic genetic lesions are crucial for tumor initiation. Mice were then administered carcinogen diethylnitrosamine (DEN) to induce liver cancer[68]. By 14 weeks post DEN injection, liver cancer did not develop in mice that received empty-vector even if they were fed HF diet. After HBV-plasmid injection, liver cancer developed by 14 weeks in all mice that were given the HF diet but not the NC diet. Compared to mice infected with Ref-HBV isolated from an HCC patient with overt HBV infection, those infected with preS variants derived from OBI patients exhibited a higher liver tumor burden. In a subset of HF-diet mice that received DEN and mtPreS1 variants, no liver cancer was detected following myriocin administration for inhibition of ceramide de novo synthesis. Notably, mice that did not receive DEN injections did not develop liver cancer by 14 weeks, even when fed the HF diet.

In cancer development, chronic inflammation has been suggested as a factor in different settings[69-71]. As an endogenous “danger signal” to trigger inflammation, ceramide can trigger activation of NOD-like receptor protein 3 (NLRP3), promoting the development of metabolism-related disorders and liver disease progression[67,72]. Owing to their location at the interface in the tissue, liver macrophages constantly receive signals from hepatocytes[73]. The IL-1β and IL-18 production mediated by NLRP3 activation was enhanced in macrophages after stimulation with HBV proteins in the presence of ceramides. HBV proteins and ceramides displayed synergistic effects on the activation of macrophage NLRP3 and inflammatory cytokine production depending on ceramide concentration. In the livers of HF-diet mice, inflammatory macrophages increased threefold compared with liver cells in NC diet-fed mice. Remarkably, NLRP3 activation after HF diet was significantly activated in liver macrophages of HBV-infected mice, especially those with preS variant infection, generating larger amounts of IL-1β and IL-18[65]. These results from murine model suggest that hepatocyte infection with preS variants altered amino acid properties of L-HBsAg and generated abnormal ceramides. In the presence of HBV proteins, ceramides co-activated NLRP3 inflammasome in liver macrophages to promote autochthonous HCC development [Figure 1]. HBV infection, particularly with variants leading to HBV retention, might synergistically promote HCC development in the context of HF diet.

EFFECTS OF OCCULT HEPATITIS B VIRUS INFECTION AND METABOLIC DISORDERS ON HCC DEVELOPMENT: IS THERE A SYNERGISTIC EFFECT?

Over the past two decades, NAFLD/MAFLD has been extensively described in populations from different areas. Experts in this area have proposed the term MAFLD and described the disease as an overarching one. They appreciated that MAFLD is a phenotype with complex and different causes and its pathogenesis is heterogeneous[11]. Due to the high prevalence of HBV infection, many studies in the Asian population also address disease progression per se to liver and other body organs/systems in individuals with chronic HBV infection, even among those considered “healthy”. Patients with MAFLD have a higher incidence of HCC than those without the disease, and HCC can develop in cases of metabolic syndromes with no liver cirrhosis[74-78]. MAFLD, therefore, has received significant attention and is considered an important risk factor for HCC[11]. Concurrent fatty liver is common in HBV-infected patients and it is an independent risk factor potentiating HBV-associated HCC development[79]. Obesity, one of the components of MAFLD, was reported to be much stronger in individuals with concomitant HCV infection than in individuals with HBV infection[80]. Since few cases were also positive for HCV[60], a few reviewers questioned the OBI as an independent risk factor for HCC but considered the OBI as a cofactor with HCV infection that promotes HCC development[81]. Nevertheless, clinical and laboratory experimental studies with solitary HBV infection confirm the importance of OBI in HCC development[39,42,61,65].

Due to the crucial role of HBx in HCC development, some laboratory experimental studies and animal models suggested that chronic HBV infection can promote hepatic steatosis, which is mainly dependent on HBx-induced expression of liver fatty acid binding protein[82]. However, clinical data showed that hepatic steatosis and metabolic dysfunction have distinct effects on HCC risk[32,37]. Hepatic steatosis was not associated with liver fibrosis[33], a proven HCC risk factor[1]. In cases of hepatic steatosis induced by high-fat diet, HBV replication was even attenuated, accompanied by reduced serum HBeAg positivity, HBV viremia, and positive staining of intrahepatic HBsAg and HBcAg[29,32,37]. Reduced hepatic steatosis even significantly increased the HCC risk[35]. Although HBV infection in combination with NAFLD for the progression of HCC remains debatable, increasing clinical evidence suggests that HBV retention within hepatocytes might play an important role in promoting HCC development [Figure 1]. Our experimental data indicated that preS variant-related OBI increases the risk of HCC in the context of a high-fat diet and these two factors act synergistically[65]. Molecular mechanisms relating to HBV retention within hepatocytes remain to be clarified. Population-based longitudinal studies are required to clarify the effects of OBI in the context of a high-fat diet, one of the preventable behavioral risk factors leading to MAFLD, such as in OBI individuals after HepB vaccination.

SUMMARY

Antiviral therapies with nucleot(s)ide analogs delay the occurrence of HBV-related diseases including HCC, but cannot cure HBV infections due to the persistence of cccDNA in hepatocytes[18]. As an emerging HCC risk factor, MAFLD is influenced by heterogeneous and multiple factors, including dietary patterns[11,83]. Hepatic ER action is significantly challenged by excess lipid exposure derived from diet[66]. Although the evidence of OBI as an independent risk factor for HCC has been debated[81], experimental data strongly suggest that preS variant-related OBI synergistically promotes HCC development in the context of a high-fat diet[65]. Modern diets are rich in unhealthy fat. To reduce HCC risk, individuals infected with PreS variant-related OBI should pay more attention to preventing metabolic disorders by, at least, improving their diet pattern and receiving proper intervention when metabolic disorders appear. People who tested positive for HBsAg have typically received more focused attention for HCC monitoring to help reduce HCC mortality[84,85]. Going forward, individuals with MAFLD who also test positive for anti-HBc should also be closely monitored for HCC screening.

DECLARATIONS

Authors’ contributions

Conceptualized the article, conducted the literature review, and prepared the final draft of the manuscript: Qu C

Performed the literature review and contributed to the writing and editing of the manuscript: Qu C, Chen K

Availability of data and materials

Not applicable.

Financial support and sponsorship

This work is supported by the National Natural Science Foundation of China (81972628), and the Innovation Fund for Medical Sciences of Chinese Academy of Medical Sciences (CIFMS, 2021-I2M-1-066 and 2023-I2M-2-004).

Conflicts of interest

Qu C is an Editorial Board member of Hepatoma Research. The other author 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. Singal AG, Kanwal F, Llovet JM. Global trends in hepatocellular carcinoma epidemiology: implications for screening, prevention and therapy. Nat Rev Clin Oncol 2023;20:864-84.

2. Cao M, Fan J, Lu L, et al. Long term outcome of prevention of liver cancer by hepatitis B vaccine: results from an RCT with 37 years. Cancer Lett 2022;536:215652.

3. Chiang CJ, Yang YW, You SL, Lai MS, Chen CJ. Thirty-year outcomes of the national hepatitis B immunization program in Taiwan. JAMA 2013;310:974-6.

4. Qu C, Chen T, Fan C, et al. Efficacy of neonatal HBV vaccination on liver cancer and other liver diseases over 30-year follow-up of the Qidong hepatitis B intervention study: a cluster randomized controlled trial. PLoS Med 2014;11:e1001774.

5. Sun Z, Chen T, Thorgeirsson SS, et al. Dramatic reduction of liver cancer incidence in young adults: 28 year follow-up of etiological interventions in an endemic area of China. Carcinogenesis 2013;34:1800-5.

6. Tseng CH, Tseng CM, Wu JL, Hsu YC, El-Serag HB. Magnitude of and prediction for risk of hepatocellular carcinoma in patients with chronic hepatitis B taking entecavir or tenofovir therapy: a systematic review. J Gastroenterol Hepatol 2020;35:1684-93.

7. Lockart I, Yeo MGH, Hajarizadeh B, Dore GJ, Danta M. HCC incidence after hepatitis C cure among patients with advanced fibrosis or cirrhosis: a meta-analysis. Hepatology 2022;76:139-54.

8. Zhang W, Xiang YB, Li HL, et al. Vegetable-based dietary pattern and liver cancer risk: results from the Shanghai women’s and men’s health studies. Cancer Sci 2013;104:1353-61.

9. Shen QM, Tuo JY, Li ZY, et al. Sex-specific impact of dietary patterns on liver cancer incidence: updated results from two population-based cohort studies in China. Eur J Nutr 2024;63:1113-24.

10. Zhao S, Breivik K, Jones KC, Sweetman AJ. Modeling the time-variant dietary exposure of PCBs in China over the period 1930 to 2100. Environ Sci Technol 2018;52:7371-9.

11. Eslam M, Sanyal AJ, George J; International Consensus Panel. MAFLD: a consensus-driven proposed nomenclature for metabolic associated fatty liver disease. Gastroenterology 2020;158:1999-2014.e1.

12. Makarova-Rusher OV, Altekruse SF, McNeel TS, et al. Population attributable fractions of risk factors for hepatocellular carcinoma in the United States. Cancer 2016;122:1757-65.

13. Vitale A, Svegliati-Baroni G, Ortolani A, et al; Italian Liver Cancer (ITA.LI.CA) group. Epidemiological trends and trajectories of MAFLD-associated hepatocellular carcinoma 2002-2033: the ITA.LI.CA database. Gut 2023;72:141-52.

14. Crane H, Eslick GD, Gofton C, et al. Global prevalence of metabolic dysfunction-associated fatty liver disease-related hepatocellular carcinoma: A systematic review and meta-analysis. Clin Mol Hepatol 2024;30:436-48.

15. George J, Lau G, Kawaguchi T, et al. Furthering research on MAFLD: the APASL Metabolic fAtty lIver DiseasE coNsortium (MAIDEN). Hepatol Int 2023;17:546-9.

16. Lin J, Zhang H, Yu H, et al. Epidemiological characteristics of primary liver cancer in mainland China from 2003 to 2020: a representative multicenter study. Front Oncol 2022;12:906778.

17. Tang X, Shi Y, Du J, et al. Clinical outcome of non-alcoholic fatty liver disease: an 11-year follow-up study. BMJ Open 2022;12:e054891.

18. Seeger C, Mason WS. Molecular biology of hepatitis B virus infection. Virology 2015;479-80:672-86.

19. Polaris Observatory Collaborators. Global prevalence, cascade of care, and prophylaxis coverage of hepatitis B in 2022: a modelling study. Lancet Gastroenterol Hepatol 2023;8:879-907.

20. Wang J, Huang H, Liu Y, et al. HBV genome and life cycle. Adv Exp Med Biol 2020;1179:17-37.

21. Lamontagne RJ, Bagga S, Bouchard MJ. Hepatitis B virus molecular biology and pathogenesis. Hepatoma Res 2016;2:163-86.

22. Yan H, Zhong G, Xu G, et al. Sodium taurocholate cotransporting polypeptide is a functional receptor for human hepatitis B and D virus. eLife 2012;1:e00049.

23. Königer C, Wingert I, Marsmann M, Rösler C, Beck J, Nassal M. Involvement of the host DNA-repair enzyme TDP2 in formation of the covalently closed circular DNA persistence reservoir of hepatitis B viruses. Proc Natl Acad Sci U S A 2014;111:E4244-53.

24. Qi Y, Gao Z, Xu G, et al. DNA polymerase κ is a key cellular factor for the formation of covalently closed circular DNA of hepatitis B virus. PLoS Pathog 2016;12:e1005893.

25. Huovila AP, Eder AM, Fuller SD. Hepatitis B surface antigen assembles in a post-ER, pre-Golgi compartment. J Cell Biol 1992;118:1305-20.

26. Bruss V, Ganem D. The role of envelope proteins in hepatitis B virus assembly. Proc Natl Acad Sci U S A 1991;88:1059-63.

27. Liu L, Li H, Zhang Y, Zhang J, Cao Z. Hepatitis B virus infection combined with nonalcoholic fatty liver disease: interaction and prognosis. Heliyon 2023;9:e13113.

28. Joo EJ, Chang Y, Yeom JS, Ryu S. Hepatitis B virus infection and decreased risk of nonalcoholic fatty liver disease: a cohort study. Hepatology 2017;65:828-35.

29. Hu D, Wang H, Wang H, et al. Non-alcoholic hepatic steatosis attenuates hepatitis B virus replication in an HBV-immunocompetent mouse model. Hepatol Int 2018;12:438-46.

30. Adams LA, Lymp JF, St Sauver J, et al. The natural history of nonalcoholic fatty liver disease: a population-based cohort study. Gastroenterology 2005;129:113-21.

31. Yang HI, Lu SN, Liaw YF, et al; Taiwan Community-Based Cancer Screening Project Group. Hepatitis B e antigen and the risk of hepatocellular carcinoma. N Engl J Med 2002;347:168-74.

32. Wang MM, Wang GS, Shen F, Chen GY, Pan Q, Fan JG. Hepatic steatosis is highly prevalent in hepatitis B patients and negatively associated with virological factors. Dig Dis Sci 2014;59:2571-9.

33. Zheng Q, Zou B, Wu Y, et al. Systematic review with meta-analysis: prevalence of hepatic steatosis, fibrosis and associated factors in chronic hepatitis B. Aliment Pharmacol Ther 2021;54:1100-9.

34. Cheng YM, Hsieh TH, Wang CC, Kao JH. Impact of HBV infection on clinical outcomes in patients with metabolic dysfunction-associated fatty liver disease. JHEP Rep 2023;5:100836.

35. Mak LY, Hui RW, Fung J, et al. Reduced hepatic steatosis is associated with higher risk of hepatocellular carcinoma in chronic hepatitis B infection. Hepatol Int 2021;15:901-11.

36. 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.

37. Huang SC, Su TH, Tseng TC, et al. Distinct effects of hepatic steatosis and metabolic dysfunction on the risk of hepatocellular carcinoma in chronic hepatitis B. Hepatol Int 2023;17:1139-49.

38. 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;153:1006-17.e5.

39. Pollicino T, Squadrito G, Cerenzia G, et al. Hepatitis B virus maintains its pro-oncogenic properties in the case of occult HBV infection. Gastroenterology 2004;126:102-10.

40. Pollicino T, Cacciola I, Saffioti F, Raimondo G. Hepatitis B virus PreS/S gene variants: pathobiology and clinical implications. J Hepatol 2014;61:408-17.

41. Pollicino T, Amaddeo G, Restuccia A, et al. Impact of hepatitis B virus (HBV) preS/S genomic variability on HBV surface antigen and HBV DNA serum levels. Hepatology 2012;56:434-43.

42. Raimondo G, Locarnini S, Pollicino T, Levrero M, Zoulim F, Lok AS; Taormina Workshop on Occult HBV Infection Faculty Members. Update of the statements on biology and clinical impact of occult hepatitis B virus infection. J Hepatol 2019;71:397-408.

43. Xu L, Wei Y, Chen T, et al. Occult HBV infection in anti-HBs-positive young adults after neonatal HB vaccination. Vaccine 2010;28:5986-92.

44. Chaudhuri V, Tayal R, Nayak B, Acharya SK, Panda SK. Occult hepatitis B virus infection in chronic liver disease: full-length genome and analysis of mutant surface promoter. Gastroenterology 2004;127:1356-71.

45. Candotti D, Lin CK, Belkhiri D, et al. Occult hepatitis B infection in blood donors from South East Asia: molecular characterisation and potential mechanisms of occurrence. Gut 2012;61:1744-53.

46. Huang FY, Wong DK, Seto WK, et al. Sequence variations of full-length hepatitis B virus genomes in Chinese patients with HBsAg-negative hepatitis B infection. PLoS One 2014;9:e99028.

47. Loomba R, Liang TJ. Hepatitis B reactivation associated with immune suppressive and biological modifier therapies: current concepts, management strategies, and future directions. Gastroenterology 2017;152:1297-309.

48. Liu Y, Zeng W, Xi J, et al. Over-gap PCR amplification to identify presence of replication-competent HBV DNA from integrated HBV DNA: an updated occult HBV infection definition. J Hepatol 2019;70:557-9.

49. Liang X, Cui F, Hadler S, et al. Origins, design and implementation of the China GAVI project. Vaccine 2013;31:J8-14.

50. Wang FZ, Zhang GM, Shen LP, et al. [Comparative analyze on hepatitis B seroepidemiological surveys among population aged 1-29 years in different epidemic regions of China in 1992 and 2014]. Zhonghua Yu Fang Yi Xue Za Zhi 2017;51:462-8.

51. Ni YH, Huang LM, Chang MH, et al. Two decades of universal hepatitis B vaccination in taiwan: impact and implication for future strategies. Gastroenterology 2007;132:1287-93.

52. Wang Y, Chen T, Lu LL, et al. Adolescent booster with hepatitis B virus vaccines decreases HBV infection in high-risk adults. Vaccine 2017;35:1064-70.

53. Yang YT, Huang AL, Zhao Y. The prevalence of hepatitis B core antibody in vaccinated Chinese children: a hospital-based study. Vaccine 2019;37:458-63.

54. Hsu HY, Chang MH, Ni YH, et al. Chronologic changes in serum hepatitis B virus DNA, genotypes, surface antigen mutants and reverse transcriptase mutants during 25-year nationwide immunization in Taiwan. J Viral Hepat 2017;24:645-53.

55. Mu SC, Lin YM, Jow GM, Chen BF. Occult hepatitis B virus infection in hepatitis B vaccinated children in Taiwan. J Hepatol 2009;50:264-72.

56. Yan YP, Su HX, Ji ZH, Shao ZJ, Pu ZS. Epidemiology of hepatitis B virus infection in china: current status and challenges. J Clin Transl Hepatol 2014;2:15-22.

57. Wang R, Liu C, Chen T, et al. Neonatal hepatitis B vaccination protects mature adults from occult virus infection. Hepatol Int 2021;15:328-37.

58. Qu C, Duan Z, Chen K, Zou H. Reducing liver cancer risk beginning at birth: experiences of preventing chronic hepatitis B virus infection in China. HR 2017;3:228-40.

59. Romanò L, Carsetti R, Tozzi AE, Mele A, Zanetti AR. Chronic hepatitis B infection in adolescents vaccinated at birth: an alarm bell in favor of the need for a booster? Hepatology 2014;59:349.

60. Shi Y, Wu YH, Wu W, Zhang WJ, Yang J, Chen Z. Association between occult hepatitis B infection and the risk of hepatocellular carcinoma: a meta-analysis. Liver Int 2012;32:231-40.

61. Wong DK, Huang FY, Lai CL, et al. Occult hepatitis B infection and HBV replicative activity in patients with cryptogenic cause of hepatocellular carcinoma. Hepatology 2011;54:829-36.

62. Carman WF, Zanetti AR, Karayiannis P, et al. Vaccine-induced escape mutant of hepatitis B virus. Lancet 1990;336:325-9.

63. Fortuin M, Karthigesu V, Allison L, et al. Breakthrough infections and identification of a viral variant in Gambian children immunized with hepatitis B vaccine. J Infect Dis 1994;169:1374-6.

64. Zhu CL, Liu P, Chen T, et al. Presence of immune memory and immunity to hepatitis B virus in adults after neonatal hepatitis B vaccination. Vaccine 2011;29:7835-41.

65. Liu C, Chen K, Zhao F, et al. Occult infection with hepatitis B virus PreS variants synergistically promotes hepatocellular carcinoma development in a high-fat diet context by generating abnormal ceramides. BMC Med 2022;20:279.

66. Lemmer IL, Willemsen N, Hilal N, Bartelt A. A guide to understanding endoplasmic reticulum stress in metabolic disorders. Mol Metab 2021;47:101169.

67. Turpin-Nolan SM, Brüning JC. The role of ceramides in metabolic disorders: when size and localization matters. Nat Rev Endocrinol 2020;16:224-33.

68. Vesselinovitch SD, Mihailovich N. Kinetics of diethylnitrosamine hepatocarcinogenesis in the infant mouse. Cancer Res 1983;43:4253-9.

69. Zang M, Li Y, He H, et al. IL-23 production of liver inflammatory macrophages to damaged hepatocytes promotes hepatocellular carcinoma development after chronic hepatitis B virus infection. Biochim Biophys Acta Mol Basis Dis 2018;1864:3759-70.

70. Grivennikov SI, Greten FR, Karin M. Immunity, inflammation, and cancer. Cell 2010;140:883-99.

71. Colotta F, Allavena P, Sica A, Garlanda C, Mantovani A. Cancer-related inflammation, the seventh hallmark of cancer: links to genetic instability. Carcinogenesis 2009;30:1073-81.

72. Mridha AR, Wree A, Robertson AAB, et al. NLRP3 inflammasome blockade reduces liver inflammation and fibrosis in experimental NASH in mice. J Hepatol 2017;66:1037-46.

73. Kubes P, Jenne C. Immune responses in the liver. Annu Rev Immunol 2018;36:247-77.

74. El-Serag HB, Tran T, Everhart JE. Diabetes increases the risk of chronic liver disease and hepatocellular carcinoma. Gastroenterology 2004;126:460-8.

75. Lauby-Secretan B, Scoccianti C, Loomis D, et al; International Agency for Research on Cancer Handbook Working Group. Body fatness and cancer - viewpoint of the IARC working group. N Engl J Med 2016;375:794-8.

76. Paradis V, Zalinski S, Chelbi E, et al. Hepatocellular carcinomas in patients with metabolic syndrome often develop without significant liver fibrosis: a pathological analysis. Hepatology 2009;49:851-9.

77. Yasui K, Hashimoto E, Komorizono Y, et al; Japan NASH Study Group, Ministry of Health, Labour, and Welfare of Japan. Characteristics of patients with nonalcoholic steatohepatitis who develop hepatocellular carcinoma. Clin Gastroenterol Hepatol 2011;9:428-33.

78. Hagström H, Tynelius P, Rasmussen F. High BMI in late adolescence predicts future severe liver disease and hepatocellular carcinoma: a national, population-based cohort study in 1.2 million men. Gut 2018;67:1536-42.

79. Chan AWH, Wong GLH, Chan HY, et al. Concurrent fatty liver increases risk of hepatocellular carcinoma among patients with chronic hepatitis B. J Gastroenterol Hepatol 2017;32:667-76.

80. El-Serag HB, Kanwal F. Epidemiology of hepatocellular carcinoma in the United States: where are we? Where do we go? Hepatology 2014;60:1767-75.

81. El-Serag HB. Epidemiology of viral hepatitis and hepatocellular carcinoma. Gastroenterology 2012;142:1264-73.e1.

82. Wu YL, Peng XE, Zhu YB, Yan XL, Chen WN, Lin X. Hepatitis B virus x protein induces hepatic steatosis by enhancing the expression of liver fatty acid binding protein. J Virol 2016;90:1729-40.

83. Kulik L, El-Serag HB. Epidemiology and management of hepatocellular carcinoma. Gastroenterology 2019;156:477-91.e1.

84. Omata M, Cheng AL, Kokudo N, et al. Asia-Pacific clinical practice guidelines on the management of hepatocellular carcinoma: a 2017 update. Hepatol Int 2017;11:317-70.

85. Marrero JA, Kulik LM, Sirlin CB, et al. Diagnosis, staging, and management of hepatocellular carcinoma: 2018 practice guidance by the American association for the study of liver diseases. Hepatology 2018;68:723-50.

Cite This Article

Review
Open Access
Impact of occult hepatitis B virus infection and high-fat diet on hepatocellular carcinoma development
Kun ChenKun Chen, Chunfeng QuChunfeng Qu

How to Cite

Chen, K.; Qu C. Impact of occult hepatitis B virus infection and high-fat diet on hepatocellular carcinoma development. Hepatoma. Res. 2024, 10, 38. http://dx.doi.org/10.20517/2394-5079.2024.85

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
78
Downloads
22
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/