Topic Highlight Open Access
Copyright ©The Author(s) 2016. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Gastroenterol. Feb 14, 2016; 22(6): 1943-1952
Published online Feb 14, 2016. doi: 10.3748/wjg.v22.i6.1943
Hepatocellular carcinoma and hepatitis B surface protein
Yong-Wei Li, Jiong-Shan Zhang, Department of Traditional Chinese Medicine, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510630, Guangdong Province, China
Feng-Cai Yang, Department of Medical Record Room, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510630, Guangdong Province, China
Hui-Qiong Lu, Medical Research Center, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510630, Guangdong Province, China
Author contributions: All authors equally contributed to the conception and design of the study, literature review, and analysis, critical revision, and editing of the paper; All authors approved the final version of the manuscript.
Supported by Science and Technology Planning Project of Guangdong Province, China, No. 2014A020212073.
Conflict-of-interest statement: The authors have no potential conflicts of interest.
Open-Access: This article is an open-access article which was selected by an in-house editor and fully peer-reviewed by external reviewers. It is distributed in accordance with the Creative Commons Attribution Non Commercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited and the use is non-commercial. See: http://creativecommons.org/licenses/by-nc/4.0/
Correspondence to: Dr. Yong-Wei Li, Department of Traditional Chinese Medicine, the Third Affiliated Hospital of Sun Yat-sen University, 600 Tianhe Road, Guangzhou 510630, Guangdong Province, China. liyongwei22@163.com
Telephone: +86-20-85253028
Received: April 11, 2015
Peer-review started: April 13, 2015
First decision: June 26, 2015
Revised: July 27, 2015
Accepted: November 9, 2015
Article in press: December 1, 2015
Published online: February 14, 2016

Abstract

The tumorigenesis of hepatitis B virus (HBV)-associated hepatocellular carcinoma (HCC) has been widely studied. HBV envelope proteins are important for the structure and life cycle of HBV, and these proteins are useful for judging the natural disease course and guiding treatment. Truncated and mutated preS/S are produced by integrated viral sequences that are defective for replication. The preS/S mutants are considered “precursor lesions” of HCC. Different preS/S mutants induce various mechanisms of tumorigenesis, such as transactivation of transcription factors and an immune inflammatory response, thereby contributing to HCC. The preS2 mutants and type II “Ground Glass” hepatocytes represent novel biomarkers of HBV-associated HCC. The preS mutants may induce the unfolded protein response and endoplasmic reticulum stress-dependent and stress-independent pathways. Treatments to inhibit hepatitis B surface antigen (HBsAg) and damage secondary to HBsAg or the preS/S mutants include antivirals and antioxidants, such as silymarin, resveratrol, and glycyrrhizin acid. Methods for the prevention and treatment of HCC should be comprehensive.

Key Words: Hepatitis B surface protein, Hepatocellular carcinoma, PreS/S mutants, Endoplasmic reticulum stress, “Ground Glass” hepatocytes

Core tip: The tumorigenesis of hepatitis B virus-associated hepatocellular carcinoma (HCC) has been widely studied. The preS/S mutants are considered “precursor lesions” of HCC. Different preS/S mutants induce various mechanisms of tumorigenesis, such as transactivation and an inflammatory response. The preS2 mutants and type II “Ground Glass” hepatocytes represent novel biomarkers of HCC. The preS mutants may induce the unfolded protein response and endoplasmic reticulum stress-dependent and stress-independent pathways. Treatments to inhibit hepatitis B surface antigen (HBsAg) and damage secondary to HBsAg or the preS/S mutants include antivirals and antioxidants. Methods for the prevention and treatment of HCC should be comprehensive.



INTRODUCTION

There are approximately 387 million carriers of hepatitis B virus (HBV) worldwide, and one-quarter of them will develop hepatocellular carcinoma (HCC)[1]. In HBV-endemic regions, chronic hepatitis B (CHB) is a primary risk for HCC[2]. Epidemiological studies have provided overwhelming evidence for a causal role of CHB infection in HCC development[3]. The risk for developing HCC increases from 6 to 37 times in patients with different statuses of HBV infection compared with control subjects[4-8]. In one study, the relative risk of HCC was increased by approximately 100-fold in HBV carriers compared with noncarriers[9]. Most of the HCC cases occur at the advanced stage or the anti-HBe-positive phase, with the peak incidence occurring in the sixth decade[10]. However, studies of HBV-induced tumorigenesis are widely debated[3].

STRUCTURE AND ROLE OF SURFACE PROTEINS IN HBV

The HBV genome of hepadnaviruses is a relaxed circular, partially double-stranded DNA (RC-DNA) structure[11]. The RC-DNA is converted into a template for the transcription of viral RNAs - covalently closed circular molecules - in the nucleus[12-14]. Three major unspliced transcripts, with sizes of 3.5 kb, 2.4 kb, and 2.1 kb, and a less abundant 0.8 kb transcript with a common polyadenylation site, all code for the viral proteins; the 3.5 kb pregenomic RNA also serves as a template for viral replication through a reverse transcription mechanism[14]. The 3.2 kb HBV genome has four overlapping open reading frames (ORFs)[3]: the preC/C ORF encodes the e antigen (HBeAg) and core antigen; the P ORF encodes the terminal protein (TP) and viral polymerase that possesses DNA polymerase, reverse transcriptase, and RNase H activities; and the X gene encodes hepatitis B x protein (HBx) for virus replication. The spliced viral transcript encodes a viral protein termed the hepatitis B spliced protein (HBSP)[15]. The preS/S ORF overlapping the HBV polymerase ORF encodes the three viral surface proteins. Three co-terminal envelope proteins termed large (LHBs, including the preS1 + preS2 + S domain), middle (MHBs, including the preS2 + S domain), and small (SHBs, the S domain alone) surface proteins, respectively[16]. Additionally, truncated and mutated preS2/S (the LHBs and truncated MHBs) or HBx proteins are produced by integrated viral sequences defective for replication[17,18]. The viral surface proteins are important for the HBV structure and life cycle because HBsAg reflects the transcriptional activity of the cccDNA[19]. The small surface proteins (SHBs) are major components of the virion envelope and the nucleocapsid-free subviral particles[20]. The HBV envelop proteins and capsids (with the HBV genome) are assembled on the viral particles in the endoplasmic reticulum (ER) and are discharged from the cell[21,22]. The viral surface proteins may interact with a host cell receptor to initiate the infection[3]. However, more than a dozen host-binding proteins to the preS1, preS2, or S domain have been identified[22]. PreS1, but not preS2, which is myristoylated at the glycine residue in position 2, is essential for virus infection[23,24]. The preS1 domain has a receptor binding site that contains the essential aa residues 9-18 and recognizes the asialoglycoprotein receptor on the surface of human hepatocytes or HCC cells[25-27]. HBV initially combines with heparan sulfate proteoglycans (HSPGs) that are trapped within the liver in the space of Dissé[20]. Sodium taurocholate cotransporting polypeptide (NTCP) is a binding partner for the myristoylated peptide 2-48 of the preS1 domain. Additionally, hepatitis delta virus uses HBV envelope proteins for its transmission[16]. However, the initial phases of HBV infecting hepatocytes, namely virion attachment, uncoating, and entry, are not completely identified[3]. Another important role of HBV surface proteins is in tumorigenesis, particularly in HCC, which has complex and heterogeneous features[3,28].

HBV PROTEINS AND GENOME IN HEPATOCARCINOGENESIS

HBV tumorigenesis involves inflammation and liver regeneration, HBV gene mutations, and viral (mutant) oncoproteins[4,13,29,30]. HCC-associated signaling pathways include the Wnt/b-catenin signaling[31], the p14ARF/p53 pathway[32], transforming growth factor alpha (TGF-α) signaling, Ras/mitogen-activated protein kinase (MAPK) signaling, and phosphatase and tensin homolog/Akt and mammalian target of rapamycin (mTOR) pathways[33].

Both HBeAg and HBV genotype C infections are considered risk factors of HCC[3,34]. Because reverse transcriptase lacks proofreading activity, HBV gene mutations occur more frequently than in other DNA viruses[35]. Various mutations can predict HBV-associated HCC during long-term infection[35]. HCC-associated mutations include the preS region in HBV genotype C, C1653T in enhancer II, as well as T1753V and A1762T/G1764A in the basal core promoter in HBV genotype C. These mutations alone or in combination can predict HCC development in 80% of all cases[36-38]. HBSP can promote carcinogenesis[39] as well as hepatoma cell motility and invasion[40]. Occult HBV infection (OBI, positive for HBV-DNA, negative for serum HBsAg) may produce proteins with transforming properties that contribute to hepatocellular transformation[41,42]. HBV DNA integration may frequently regulate key cellular pathways of carcinogenesis[42], contribute to cis- or trans-activation[3,43-47], and influence gene families that are involved in cell survival, proliferation, and immortalization[42].

Both HBx and HBs (mutant) proteins are designated “viral oncoproteins”. The HBx protein has been studied extensively[3]. Tumorigenesis of the HBx protein may influence the regulation of the cell cycle, signaling pathways, DNA repair[1,48-50], chromosomal instability[51], cell transcription[52,53], proliferation, and inflammation and immune responses[54-56]. The HBs (mutant) proteins, primarily preS mutants, are recognized as “precursor lesions of HCC”[57] and a risk factor for the post-operative recurrence of HCC[58]. The tumorigenesis of HBs (mutant) proteins has also been widely studied.

CLINICAL ASSOCIATION BETWEEN HEPATITIS B SURFACE PROTEINS AND HCC

HBsAg has primarily been considered a marker of HBV infection. Since the two methods for HBsAg quantification, the Architect HBsAg assay (Abbott Diagnostics, Abbott Park, IL, United States)[59] and Elecsys HBsAg II quant assay[60], first became available, serum HBsAg levels have been found to reflect the activity of intrahepatic cccDNA[61]. These methods may evaluate HBV replication more accurately.

HBsAg levels vary significantly according to the different courses of HBV infection[62]. They have proven useful in judging the natural disease course and guiding treatment in addition to HBV DNA and HBV envelope antigen and antibody[61]. CHB is classified into five phases: the ‘‘immune tolerant phase”, ‘‘immune active phase”, ‘‘inactive HBV carrier state”, ‘‘HBeAg-negative CHB phase”, and ‘‘HBsAg-negative phase”[63,64]. In HBeAg-positive patients, the HBsAg levels are associated with fibrosis and immune tolerance; lower HBsAg levels are associated with moderate to severe fibrosis[65,66]. In HBeAg-negative patients, an HBsAg level less than 1000 IU/mL was associated with a lower risk of HCC[67]. The absolute lowest risk with a cumulative risk for HCC decreased less than 1% over 9 years after the patients had anti-HBs and anti-HBe seroconversion[68]. However, the prediction[68] may not apply to preS mutants because these mutants may account for the different levels of HBsAg[61] and have a close association with HCC.

EPIDEMIOLOGY OF PRES MUTANTS

PreS mutants evolve in the long course of CHB, possibly for immune pressure, or antivirals[3,7]. The frequency of preS mutants increases successively in the different stages of CHB infection. A meta-analysis showed that it was approximately 10%, 20%, 35%, and 50% in asymptomatic HBsAg carriers, CHB patients, patients with liver cirrhosis, and HCC patients, respectively[36]. Su et al[69] reported that these mutants are present in up to 63% of HCC patients. The prevalence of the preS mutants varied in different countries and HBV genotypes; there was a higher prevalence in genotype B and C than in the other genotypes[70]. The mutants are located in both the preS1 and preS2 regions[69,71]. The preS2 mutants occur more frequently than the preS1 mutants[36], possibly because preS1 is essential for virus infection. The preS2 mutants often coincide with changes in human immune cell epitopes[71].

DIFFERENT PRES MUTANTS INDUCE VARIOUS MECHANISMS THAT CONTRIBUTE TO HCC

The preS1/preS2/S sequence encodes a transcriptional activator with potentially transforming properties[17,72,73]. These transcriptional effects can activate the protein kinase C-dependent c-Raf-1/MAP1-kinase signal pathway, thus promoting transcription factors to increase the proliferation rate of hepatocytes[17]. Only the carboxy-terminal truncation of LHBs or MHBs has transactivating properties[1]. The truncated LHB protein expressed in transgenic mice resulted in the development of HCC[74]. The transactivating effects of MHBs are mediated via sequence-specific binding to DNA[75,76], thereby stimulating promoter sequences of the c-myc, c-fos, and c-Ha-ras oncogenes[77,78]. Both LHBs and MHBs proteins have the same transcriptional effects[73,79].

Subsequent studies have reported tumorigenesis in several mutants. An HBV polymerase rtA181T/surface truncation mutant in a patient with advanced HCC transactivated the simian virus 40 and human c-Myc promoters; the tumorigenic effects of the mutant were identified in nude mice[80]. Three mutations, sL95*, sW182*, and sL216*, activated cell proliferation and transformational abilities; the sW182* mutant demonstrated potent tumorigenic activity. However, the three mutants could not promote ER stress[81]. A W4P/R mutation in the LHB region of HBV genotype C may contribute to HCC development in an interleukin (IL)-6-dependent manner only in male patients[82]. In vitro, the MHBst167 mutants interacted with the proteins related to tumor development/progression[83]. Because the preS mutants varied in different research groups, it is difficult to investigate the corresponding management.

PRES MUTANTS INDUCE ER STRESS THAT CONTRIBUTES TO HCC

HBV proteins utilize the ER protein folding machinery and cellular secretory pathway[84]. Therefore, the underlying mechanisms of preS mutants that contribute to HCC are involved in ER stress. Both preS1 and preS2 mutants activate ER stress in hepatocytes[85]. Different preS mutants activate differential activities of ER stress in hepatoma cells with accumulated LHB proteins[86].

PreS1 mutants in HBV tumorigenesis

A previous study showed that HBV preSl mutants demonstrate hepatocarcinogenesis effects through the transactivation of the TGF-α gene[87]. PreS1 mutant activated higher levels of ER chaperones (Grp78 and 94), calcium release, cyclooxygenase-2 (COX-2), inflammatory cytokines, and oxidative stress intermediates, which tend to result in apoptosis[85]. The HBV preS2 mutant proteins play a more important role in ER stress.

“Ground glass” hepatocytes accumulated with preS mutants

“Ground glass” hepatocytes (GGHs) comprise abundant particles of surface antigens that accumulate in the ER during CHB infection[88-90]. Su et al[85] found that type II GGHs distributed in large clusters express marginal HBs proteins that harbored preS2 mutants and usually emerge at the late nonreplicative stage or in cirrhotic liver; type I GGHs, which accumulate with inclusion-like HBs (small surface protein) proteins and harbor preS1 mutants, are usually scattered sporadically during the replicative phases[86]. Type II GGHs with preS2 mutants are suggested biomarkers of HCC and can predict recurrence and survival in HBV-infected HCC patients[91].

The mutated HBV surface proteins cannot be properly folded in the ER, possibly leading to the induction of the unfolded protein response (UPR)[90]. Both HBV surface proteins and HBx protein can trigger the UPR[92,93]. HBV SHBs activate the UPR and host autophagy to enhance HBV envelopment and replication[94]. However, HBV also activates the ER degradation-enhancing mannosidase-like proteins (EDEMs) to enhance the degradation of HBV surface proteins (terminally misfolded glycoproteins), thus relieving ER stress during the UPR[95]. The mechanisms might maintain the balance between viral loads and host cells to facilitate the persistence of HBV infection. This mechanism may direct us to a new treatment strategy. However, the pathogenic role of the UPR in HBV infection and HCC appears to be complex. A drug (similar to EDEM) that relieves ER stress to prevent HCC might facilitate persistent HBV infection and vice versa. These mechanisms require further study.

Pres2 mutants induce ER stress-dependent and stress-independent pathways

The preS2 mutant protein accumulated in the ER can trigger the ER stress-dependent vascular epithelial growth factor/Akt/mTOR and nuclear factor kappa B/COX-2 signal pathway[96]. Through this signal pathway, HBx and envelope proteins that accumulate simultaneously in GGHs can enhance the oncogenic effects in transgenic and human HCCs[97]. mTOR can suppress autophagy and regulate cellular metabolism[98]. The suppression of autophagy by mTOR[98] is in contrast to the induction of autophagy during UPR[94]. This might result in contradictory treatment for HCC and HBV infection[57].

The LHBs protein with preS2 mutants may also activate an ER stress-independent pathway, ultimately leading to a growth advantage in type II GGHs. The pathway includes c-Jun activation domain binding protein 1 nuclear translocation and activation of p27/retinoblastoma/Cdk2/cyclin A, D pathways, which results in cell cycle progression, cell proliferation, and centrosome over-duplication[99-101].

Together, the data show that the preS2 mutant protein is a promising gene transactivator and an ER stress inducer, resulting in host genomic instability[85] and HCC development.

HBV SURFACE PROTEINS INDUCE AN IMMUNE INFLAMMATORY RESPONSE CONTRIBUTING TO HCC

HBV-induced chronic necroinflammation plays an indirect role in hepatocarcinogenicity in the preS/S transgenic mice[74]. The LHBs proteins in the ER triggered inflammation, abnormal regeneration and transcription, resulting in cancer development[102]. This mechanism is similar to the contribution of the W4P/R mutation to HCC development in an IL-6-dependent manner[84]. Another immune-associated carcinogenesis mechanism is viral immune escape induced by HBV mutants[103]. HBV escape from the host’s immune surveillance may favor the clonal proliferation of hepatocytes with the preS mutants[104].

HBV surface proteins may be detected in immune cells. In dendritic cells, both HBV and HBsAg abrogated the CpG-A/TLR9-induced signal pathway and decreased the levels of co-stimulatory molecules and cytokines; this mechanism may contribute to HBV persistence[105].

In addition, microRNAs may contribute to carcinogenesis in HBV-induced HCC. Both HBx and HBs proteins can increase microR499a expression, which might play an oncogenic role by targeting MAPK6[106].

The duration of time between when HBV first enters a host and HCC development is long. A large number of factors participate in carcinogenesis (Table 1). What are the key and special factors? For example, not all inflammation results in cancer. Additionally, are there any other special factors that promote HCC development in an IL-6-dependent manner?

Table 1 PreS1/S2/S and hepatocellular carcinoma carcinogenesis.
FunctionPreS1/S2/S mutationRef.
A trans-activator functionIntegration of HBV preS/S sequencesCaselmann et al[72] 1990,
Kekulé et al[75] 1990
Transactivation of NF-κB, or AP and other transcription factors for transactivation, stimulating promoter sequences of the c-myc, c-fos, and c-Ha-ras oncogenesMHBstMeyer et al[77] 1992,
Lauer et al[78] 1994
A transcriptional activator, activated tumor promoter pathways via the PKC-dependent c-Raf-1/ Erk2, MAP1- kinase signal pathway, increase the proliferation rate of hepatocytesLHBs, the preS2Hildt et al[17,73,79] 1996, 1998, 2002
Transactivator with DNA-binding propertiesHBV surface (S)Alka et al[76] 2000
Transactivated the simian virus 40 and human c-Myc promoters truncation mutantPolymerase rtA181T/surfaceLai et al[80] 2008
Activated cell proliferation and transformational abilitiessL95*, sW182*, and sL216*Huang et al[81] 2014
Interacted with the proteins related to tumor development/progression in vitroMHBst167Li et al[83] 2014
Biomarkers of HCC and predictor of recurrence survivalGGHs harbored preS2 mutants in the ERMalhi et al[90] 2011,
Su et al[85] 2014
Trigger the ER stress-dependent pathway
VEGF/Akt/mTOR and NF-κB/COX-2 signal pathway in GGHsYang et al[96] 2009
Enhance the oncogenic effects in transgenic and human HCCsCo-expressing hepatitis B virus X protein and surface antigensWu et al[97] 2014
An ER stress-independent pathway
Activate JAB1 nuclear translocation and activation of p27/retinoblastoma/Cdk2/cyclin A, D pathways, results in cell cycle progression, cell proliferation, and centrosome over-duplication, a growth advantage in type II GGsLHBs with preS2 mutantsWang et al[99] 2005,
Hsieh et al[101] 2011,
Wang et al[100] 2012
Immune-associated carcinogenesis
HBV-induced chronic necroinflammationPre S/S transgenic miceChisari et al[102] 1989
The clonal proliferation of hepatocytes with the preS mutantsThe HBV mutants escape from the host’s immune surveillanceZhong et al[104] 1999
Contributing to HCC in an IL-6-dependent manner only in male patientsA W4P/R mutation in the LHB region of HBV genotype CLee et al[82] 2015
Other
Increase microR499a expression, which might play an oncogenic role by targeting MAPK6HBs proteinsXiang et al[106] 2014
TREATMENTS TO INHIBIT HBSAG AND DAMAGE SECONDARY TO HBSAG ANTIVIRALS

HBV-induced HCC was the first tumor to be prevented by universal immunization against the responsible virus[1]. The host’s immune system can control HBsAg levels[61]. However, spontaneous HBsAg seroclearance is rarely achieved (an annual incidence of 1%-2% in CHB[107,108]). At an advanced stage of HCC, the levels of HBsAg are still high, while that of HBV DNA may be negative. This tumor type with a poor prognosis is refractory to chemotherapeutic regimens[109,110]. In chronic HBV infection, the prophylactic measures or therapy for HCC include suppression of HBV replication and recovery from molecular abnormalities due to HBV infection. Achieving HBsAg loss and anti-HBs positivity is the final aim of antivirals[61]. Nucleos(t)ide analog (NA) therapy has antiviral effects that may reduce HCC development and the post-operative recurrence of HCC[111]. NA treatment affects the reverse transcription of pregenomic RNA but does not affect cccDNA and subgenomic RNA that have translational activity associated with HBsAg levels[112]. Thus, the current NA therapy hardly clears HBsAg. With NA treatment, it may take 36 to 52 years to achieve HBsAg loss[113,114]. Lamivudine was the first licensed NA introduced to treat HBV, and it reduced the incidence of HCC when compared to no treatment. However, the development of HCC may increase in cases with lamivudine resistance. The current first-line NAs, entecavir and tenofovir, may also reduce HCC development, but the risk of HCC is not eliminated, even in the patients who remain in virological remission with this therapy[115]. NA resistance suggests antiviral failure. Additionally, the drug-resistant HBV strains may have mutant surface proteins with oncogenic effects[116]. In our study, substitutions in the S region, but not the drug-resistant mutations, possibly resulted in the poor effects of adefovir[117]. Subsequent studies also showed that preS2 mutants induced resistance to NAs and predicted HCC development[118]. Treatment with interferon inhibits HBsAg or preS mutant proteins more than NAs[119-121].

Thus, due to cccDNA, HBV and HBsAg cannot be eliminated[61]. There are also no measures to manage the integrative HBV DNA. It is impossible to develop sequence-specific antivirals for so many envelope mutants. The rtA181T/surface mutant may occur spontaneously in the absence of antiviral therapy. Its oncogenic potential warrants careful re-evaluation of the current strategy of prolonged antiviral therapy[80].

However, there are many compounds in development for CHB up to June 22, 2015, such as Myrcludex B (entry inhibitor targeting NTCP), Rep 2139 (REP 9AC, HBsAg release inhibitor), TKM-HBV (HBsAg inhibitor), and BSBI-25 (cccDNA inhibitor)[122]. These compounds should improve the effects of antivirals and reduce the burden of drug resistance and HCC development[123].

TREATMENTS BASED ON ER STRESS

Drugs based on the theory of ER stress vary in the prophylaxis and treatment of HCC. The UPR is targeted as primary or adjuvant chemotherapy for HCC, e.g., bortezomib for treating cancer via ER stress-induced apoptosis[124] and mTOR inhibition as a promising strategy for the clinical management of HCC[125]. However, mTOR inhibition may activate HBV replication in HBV-induced HCC[57]. mTOR activation may recruit the YY1-HDAC1 complex to feedback suppress transcription from the preS1 promoter (nucleotides 2812-2816)[57], thus partially explaining the low or negative HBV replication in the HCC stage. Therefore, mTOR inhibitors should be used in combination with antivirals.

To prevent HCC, targets to HBV-induced ER stress provide a strategy in high-risk CHB. Antioxidants may be such ideal agents because they reduce ER stress, thereby improving protein folding[126]. Natural products, such as silymarin and resveratrol, have been used in HCC. The two drugs can target the ER stress-associated signal pathways[85]. However, these findings require further verification.

Glycyrrhizin acid (GA) has multiple functions, such as effective hepato-protection and the reduction of elevated transaminases. Glycyrrhizin can suppress ER stress in acute liver injury[127]. Long-term treatment with glycyrrhizin prevents HCC development in chronic hepatitis C infection[128]. Glycyrrhizin treatment suppressed the sialylation of HBsAg and secretion of HBsAg in PLC/PRF/5 cells[129,130]. Therefore, it is also widely administered in CHB infection. A study to determine whether drugs such as GA and extracts from other herbs would influence the preS mutants is required.

Comprehensive prevention and treatment also include avoiding other risk factors such as aflatoxin B1 and alcohol intake. A prolonged battle against the damage induced by this virus is necessary.

Footnotes

P- Reviewer: Li WH S- Editor: Yu J L- Editor: Filipodia E- Editor: Ma S

References
1.  Arbuthnot P, Kew M. Hepatitis B virus and hepatocellular carcinoma. Int J Exp Pathol. 2001;82:77-100.  [PubMed]  [DOI]  [Cited in This Article: ]
2.  Baumert TF, Thimme R, von Weizsäcker F. Pathogenesis of hepatitis B virus infection. World J Gastroenterol. 2007;13:82-90.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 98]  [Cited by in F6Publishing: 88]  [Article Influence: 5.2]  [Reference Citation Analysis (1)]
3.  Neuveut C, Wei Y, Buendia MA. Mechanisms of HBV-related hepatocarcinogenesis. J Hepatol. 2010;52:594-604.  [PubMed]  [DOI]  [Cited in This Article: ]
4.  Yang HI, Lu SN, Liaw YF, You SL, Sun CA, Wang LY, Hsiao CK, Chen PJ, Chen DS, Chen CJ. Hepatitis B e antigen and the risk of hepatocellular carcinoma. N Engl J Med. 2002;347:168-174.  [PubMed]  [DOI]  [Cited in This Article: ]
5.  Paterlini P, Driss F, Nalpas B, Pisi E, Franco D, Berthelot P, Bréchot C. Persistence of hepatitis B and hepatitis C viral genomes in primary liver cancers from HBsAg-negative patients: a study of a low-endemic area. Hepatology. 1993;17:20-29.  [PubMed]  [DOI]  [Cited in This Article: ]
6.  Ikeda K, Kobayashi M, Someya T, Saitoh S, Hosaka T, Akuta N, Suzuki F, Suzuki Y, Arase Y, Kumada H. Occult hepatitis B virus infection increases hepatocellular carcinogenesis by eight times in patients with non-B, non-C liver cirrhosis: a cohort study. J Viral Hepat. 2009;16:437-443.  [PubMed]  [DOI]  [Cited in This Article: ]
7.  Hassan MM, Hwang LY, Hatten CJ, Swaim M, Li D, Abbruzzese JL, Beasley P, Patt YZ. Risk factors for hepatocellular carcinoma: synergism of alcohol with viral hepatitis and diabetes mellitus. Hepatology. 2002;36:1206-1213.  [PubMed]  [DOI]  [Cited in This Article: ]
8.  Sun CA, Wu DM, Lin CC, Lu SN, You SL, Wang LY, Wu MH, Chen CJ. Incidence and cofactors of hepatitis C virus-related hepatocellular carcinoma: a prospective study of 12,008 men in Taiwan. Am J Epidemiol. 2003;157:674-682.  [PubMed]  [DOI]  [Cited in This Article: ]
9.  Beasley RP, Hwang LY, Lin CC, Chien CS. Hepatocellular carcinoma and hepatitis B virus. A prospective study of 22 707 men in Taiwan. Lancet. 1981;2:1129-1133.  [PubMed]  [DOI]  [Cited in This Article: ]
10.  Liaw YF, Chu CM. Hepatitis B virus infection. Lancet. 2009;373:582-592.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 460]  [Reference Citation Analysis (1)]
11.  Summers J, Mason WS. Replication of the genome of a hepatitis B--like virus by reverse transcription of an RNA intermediate. Cell. 1982;29:403-415.  [PubMed]  [DOI]  [Cited in This Article: ]
12.  Beck J, Nassal M. Hepatitis B virus replication. World J Gastroenterol. 2007;13:48-64.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 331]  [Cited by in F6Publishing: 321]  [Article Influence: 18.9]  [Reference Citation Analysis (1)]
13.  Weiser B, Ganem D, Seeger C, Varmus HE. Closed circular viral DNA and asymmetrical heterogeneous forms in livers from animals infected with ground squirrel hepatitis virus. J Virol. 1983;48:1-9.  [PubMed]  [DOI]  [Cited in This Article: ]
14.  Ganem D, Varmus HE. The molecular biology of the hepatitis B viruses. Annu Rev Biochem. 1987;56:651-693.  [PubMed]  [DOI]  [Cited in This Article: ]
15.  Soussan P, Garreau F, Zylberberg H, Ferray C, Brechot C, Kremsdorf D. In vivo expression of a new hepatitis B virus protein encoded by a spliced RNA. J Clin Invest. 2000;105:55-60.  [PubMed]  [DOI]  [Cited in This Article: ]
16.  Tong S, Li J. Identification of NTCP as an HBV receptor: the beginning of the end or the end of the beginning? Gastroenterology. 2014;146:902-905.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 24]  [Cited by in F6Publishing: 27]  [Article Influence: 2.7]  [Reference Citation Analysis (0)]
17.  Hildt E, Hofschneider PH. The PreS2 activators of the hepatitis B virus: activators of tumour promoter pathways. Recent Results Cancer Res. 1998;154:315-329.  [PubMed]  [DOI]  [Cited in This Article: ]
18.  Schlüter V, Meyer M, Hofschneider PH, Koshy R, Caselmann WH. Integrated hepatitis B virus X and 3’ truncated preS/S sequences derived from human hepatomas encode functionally active transactivators. Oncogene. 1994;9:3335-3344.  [PubMed]  [DOI]  [Cited in This Article: ]
19.  Chan HL, Thompson A, Martinot-Peignoux M, Piratvisuth T, Cornberg M, Brunetto MR, Tillmann HL, Kao JH, Jia JD, Wedemeyer H. Hepatitis B surface antigen quantification: why and how to use it in 2011 - a core group report. J Hepatol. 2011;55:1121-1131.  [PubMed]  [DOI]  [Cited in This Article: ]
20.  Leistner CM, Gruen-Bernhard S, Glebe D. Role of glycosaminoglycans for binding and infection of hepatitis B virus. Cell Microbiol. 2008;10:122-133.  [PubMed]  [DOI]  [Cited in This Article: ]
21.  Ganem D. Hepadnaviridae: the viruses and their replication. Fields virology. Philadelphia: Lippincott-Raven 1996; 2703-2737.  [PubMed]  [DOI]  [Cited in This Article: ]
22.  Wei Y, Neuveut C, Tiollais P, Buendia MA. Molecular biology of the hepatitis B virus and role of the X gene. Pathol Biol (Paris). 2010;58:267-272.  [PubMed]  [DOI]  [Cited in This Article: ]
23.  Glebe D, Urban S. Viral and cellular determinants involved in hepadnaviral entry. World J Gastroenterol. 2007;13:22-38.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 207]  [Cited by in F6Publishing: 198]  [Article Influence: 11.6]  [Reference Citation Analysis (0)]
24.  Meier A, Mehrle S, Weiss TS, Mier W, Urban S. Myristoylated PreS1-domain of the hepatitis B virus L-protein mediates specific binding to differentiated hepatocytes. Hepatology. 2013;58:31-42.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 96]  [Cited by in F6Publishing: 103]  [Article Influence: 9.4]  [Reference Citation Analysis (0)]
25.  Engelke M, Mills K, Seitz S, Simon P, Gripon P, Schnölzer M, Urban S. Characterization of a hepatitis B and hepatitis delta virus receptor binding site. Hepatology. 2006;43:750-760.  [PubMed]  [DOI]  [Cited in This Article: ]
26.  Glebe D, Urban S, Knoop EV, Cag N, Krass P, Grün S, Bulavaite A, Sasnauskas K, Gerlich WH. Mapping of the hepatitis B virus attachment site by use of infection-inhibiting preS1 lipopeptides and tupaia hepatocytes. Gastroenterology. 2005;129:234-245.  [PubMed]  [DOI]  [Cited in This Article: ]
27.  Zhang X, Lin SM, Chen TY, Liu M, Ye F, Chen YR, Shi L, He YL, Wu LX, Zheng SQ. Asialoglycoprotein receptor interacts with the preS1 domain of hepatitis B virus in vivo and in vitro. Arch Virol. 2011;156:637-645.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 21]  [Cited by in F6Publishing: 22]  [Article Influence: 1.7]  [Reference Citation Analysis (0)]
28.  Thorgeirsson SS, Lee JS, Grisham JW. Functional genomics of hepatocellular carcinoma. Hepatology. 2006;43:S145-S150.  [PubMed]  [DOI]  [Cited in This Article: ]
29.  Su IJ, Hsieh WC, Tsai HW, Wu HC. Chemoprevention and novel therapy for hepatocellular carcinoma associated with chronic hepatitis B virus infection. Hepatobiliary Surg Nutr. 2013;2:37-39.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 5]  [Reference Citation Analysis (0)]
30.  Sitia G, Aiolfi R, Di Lucia P, Mainetti M, Fiocchi A, Mingozzi F, Esposito A, Ruggeri ZM, Chisari FV, Iannacone M. Antiplatelet therapy prevents hepatocellular carcinoma and improves survival in a mouse model of chronic hepatitis B. Proc Natl Acad Sci USA. 2012;109:E2165-E2172.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 238]  [Cited by in F6Publishing: 206]  [Article Influence: 18.7]  [Reference Citation Analysis (0)]
31.  de La Coste A, Romagnolo B, Billuart P, Renard CA, Buendia MA, Soubrane O, Fabre M, Chelly J, Beldjord C, Kahn A. Somatic mutations of the beta-catenin gene are frequent in mouse and human hepatocellular carcinomas. Proc Natl Acad Sci USA. 1998;95:8847-8851.  [PubMed]  [DOI]  [Cited in This Article: ]
32.  Hussain SP, Schwank J, Staib F, Wang XW, Harris CC. TP53 mutations and hepatocellular carcinoma: insights into the etiology and pathogenesis of liver cancer. Oncogene. 2007;26:2166-2176.  [PubMed]  [DOI]  [Cited in This Article: ]
33.  Villanueva A, Chiang DY, Newell P, Peix J, Thung S, Alsinet C, Tovar V, Roayaie S, Minguez B, Sole M. Pivotal role of mTOR signaling in hepatocellular carcinoma. Gastroenterology. 2008;135:1972-183, 1972-183.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 520]  [Cited by in F6Publishing: 562]  [Article Influence: 35.1]  [Reference Citation Analysis (0)]
34.  Chan HL, Hui AY, Wong ML, Tse AM, Hung LC, Wong VW, Sung JJ. Genotype C hepatitis B virus infection is associated with an increased risk of hepatocellular carcinoma. Gut. 2004;53:1494-1498.  [PubMed]  [DOI]  [Cited in This Article: ]
35.  Zhu Y, Jin Y, Cai X, Bai X, Chen M, Chen T, Wang J, Qian G, Gu J, Li J. Hepatitis B virus core protein variations differ in tumor and adjacent nontumor tissues from patients with hepatocellular carcinoma. Intervirology. 2012;55:29-35.  [PubMed]  [DOI]  [Cited in This Article: ]
36.  Liu S, Zhang H, Gu C, Yin J, He Y, Xie J, Cao G. Associations between hepatitis B virus mutations and the risk of hepatocellular carcinoma: a meta-analysis. J Natl Cancer Inst. 2009;101:1066-1082.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 279]  [Cited by in F6Publishing: 314]  [Article Influence: 20.9]  [Reference Citation Analysis (0)]
37.  Kuang SY, Jackson PE, Wang JB, Lu PX, Muñoz A, Qian GS, Kensler TW, Groopman JD. Specific mutations of hepatitis B virus in plasma predict liver cancer development. Proc Natl Acad Sci USA. 2004;101:3575-3580.  [PubMed]  [DOI]  [Cited in This Article: ]
38.  Kao JH, Chen PJ, Lai MY, Chen DS. Basal core promoter mutations of hepatitis B virus increase the risk of hepatocellular carcinoma in hepatitis B carriers. Gastroenterology. 2003;124:327-334.  [PubMed]  [DOI]  [Cited in This Article: ]
39.  Chen JY, Chen WN, Jiao BY, Lin WS, Wu YL, Liu LL, Lin X. Hepatitis B spliced protein (HBSP) promotes the carcinogenic effects of benzo [alpha] pyrene by interacting with microsomal epoxide hydrolase and enhancing its hydrolysis activity. BMC Cancer. 2014;14:282.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 11]  [Cited by in F6Publishing: 11]  [Article Influence: 1.1]  [Reference Citation Analysis (0)]
40.  Chen WN, Chen JY, Jiao BY, Lin WS, Wu YL, Liu LL, Lin X. Interaction of the hepatitis B spliced protein with cathepsin B promotes hepatoma cell migration and invasion. J Virol. 2012;86:13533-13541.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 27]  [Cited by in F6Publishing: 30]  [Article Influence: 2.5]  [Reference Citation Analysis (0)]
41.  Bréchot C. Pathogenesis of hepatitis B virus-related hepatocellular carcinoma: old and new paradigms. Gastroenterology. 2004;127:S56-S61.  [PubMed]  [DOI]  [Cited in This Article: ]
42.  Pollicino T, Squadrito G, Cerenzia G, Cacciola I, Raffa G, Craxi A, Farinati F, Missale G, Smedile A, Tiribelli C. Hepatitis B virus maintains its pro-oncogenic properties in the case of occult HBV infection. Gastroenterology. 2004;126:102-110.  [PubMed]  [DOI]  [Cited in This Article: ]
43.  Garcia M, de Thé H, Tiollais P, Samarut J, Dejean A. A hepatitis B virus pre-S-retinoic acid receptor beta chimera transforms erythrocytic progenitor cells in vitro. Proc Natl Acad Sci USA. 1993;90:89-93.  [PubMed]  [DOI]  [Cited in This Article: ]
44.  Berasain C, Patil D, Perara E, Huang SM, Mouly H, Bréchot C. Oncogenic activation of a human cyclin A2 targeted to the endoplasmic reticulum upon hepatitis B virus genome insertion. Oncogene. 1998;16:1277-1288.  [PubMed]  [DOI]  [Cited in This Article: ]
45.  Horikawa I, Barrett JC. cis-Activation of the human telomerase gene (hTERT) by the hepatitis B virus genome. J Natl Cancer Inst. 2001;93:1171-1173.  [PubMed]  [DOI]  [Cited in This Article: ]
46.  Yaginuma K, Kobayashi M, Yoshida E, Koike K. Hepatitis B virus integration in hepatocellular carcinoma DNA: duplication of cellular flanking sequences at the integration site. Proc Natl Acad Sci USA. 1985;82:4458-4462.  [PubMed]  [DOI]  [Cited in This Article: ]
47.  Tsuei DJ, Chang MH, Chen PJ, Hsu TY, Ni YH. Characterization of integration patterns and flanking cellular sequences of hepatitis B virus in childhood hepatocellular carcinomas. J Med Virol. 2002;68:513-521.  [PubMed]  [DOI]  [Cited in This Article: ]
48.  Andrisani OM, Barnabas S. The transcriptional function of the hepatitis B virus X protein and its role in hepatocarcinogenesis (Review). Int J Oncol. 1999;15:373-379.  [PubMed]  [DOI]  [Cited in This Article: ]
49.  Bouchard MJ, Schneider RJ. The enigmatic X gene of hepatitis B virus. J Virol. 2004;78:12725-12734.  [PubMed]  [DOI]  [Cited in This Article: ]
50.  Tang H, Oishi N, Kaneko S, Murakami S. Molecular functions and biological roles of hepatitis B virus x protein. Cancer Sci. 2006;97:977-983.  [PubMed]  [DOI]  [Cited in This Article: ]
51.  Rakotomalala L, Studach L, Wang WH, Gregori G, Hullinger RL, Andrisani O. Hepatitis B virus X protein increases the Cdt1-to-geminin ratio inducing DNA re-replication and polyploidy. J Biol Chem. 2008;283:28729-28740.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 43]  [Cited by in F6Publishing: 47]  [Article Influence: 2.9]  [Reference Citation Analysis (0)]
52.  Cougot D, Wu Y, Cairo S, Caramel J, Renard CA, Lévy L, Buendia MA, Neuveut C. The hepatitis B virus X protein functionally interacts with CREB-binding protein/p300 in the regulation of CREB-mediated transcription. J Biol Chem. 2007;282:4277-4287.  [PubMed]  [DOI]  [Cited in This Article: ]
53.  Barnabas S, Hai T, Andrisani OM. The hepatitis B virus X protein enhances the DNA binding potential and transcription efficacy of bZip transcription factors. J Biol Chem. 1997;272:20684-20690.  [PubMed]  [DOI]  [Cited in This Article: ]
54.  Rossner MT. Review: hepatitis B virus X-gene product: a promiscuous transcriptional activator. J Med Virol. 1992;36:101-117.  [PubMed]  [DOI]  [Cited in This Article: ]
55.  Yen TS. Hepadnaviral X Protein: Review of Recent Progress. J Biomed Sci. 1996;3:20-30.  [PubMed]  [DOI]  [Cited in This Article: ]
56.  Cougot D, Buendia MA, Neuveut C. Carcinogenesis induced by hepatitis B virus. Translational research in biomedecine. Basel: Karger 2008; 108-136.  [PubMed]  [DOI]  [Cited in This Article: ]
57.  Teng CF, Wu HC, Tsai HW, Shiah HS, Huang W, Su IJ. Novel feedback inhibition of surface antigen synthesis by mammalian target of rapamycin (mTOR) signal and its implication for hepatitis B virus tumorigenesis and therapy. Hepatology. 2011;54:1199-1207.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 51]  [Cited by in F6Publishing: 54]  [Article Influence: 4.2]  [Reference Citation Analysis (0)]
58.  Su CW, Chiou YW, Tsai YH, Teng RD, Chau GY, Lei HJ, Hung HH, Huo TI, Wu JC. The Influence of Hepatitis B Viral Load and Pre-S Deletion Mutations on Post-Operative Recurrence of Hepatocellular Carcinoma and the Tertiary Preventive Effects by Anti-Viral Therapy. PLoS One. 2013;8:e66457.  [PubMed]  [DOI]  [Cited in This Article: ]
59.  Deguchi M, Yamashita N, Kagita M, Asari S, Iwatani Y, Tsuchida T, Iinuma K, Mushahwar IK. Quantitation of hepatitis B surface antigen by an automated chemiluminescent microparticle immunoassay. J Virol Methods. 2004;115:217-222.  [PubMed]  [DOI]  [Cited in This Article: ]
60.  Zacher BJ, Moriconi F, Bowden S, Hammond R, Louisirirotchanakul S, Phisalprapa P, Tanwandee T, Wursthorn K, Brunetto MR, Wedemeyer H. Multicenter evaluation of the Elecsys hepatitis B surface antigen quantitative assay. Clin Vaccine Immunol. 2011;18:1943-1950.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 29]  [Cited by in F6Publishing: 30]  [Article Influence: 2.3]  [Reference Citation Analysis (0)]
61.  Höner Zu Siederdissen C, Cornberg M. The role of HBsAg levels in the current management of chronic HBV infection. Ann Gastroenterol. 2014;27:105-112.  [PubMed]  [DOI]  [Cited in This Article: ]
62.  Jaroszewicz J, Calle Serrano B, Wursthorn K, Deterding K, Schlue J, Raupach R, Flisiak R, Bock CT, Manns MP, Wedemeyer H. Hepatitis B surface antigen (HBsAg) levels in the natural history of hepatitis B virus (HBV)-infection: a European perspective. J Hepatol. 2010;52:514-522.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 295]  [Cited by in F6Publishing: 294]  [Article Influence: 21.0]  [Reference Citation Analysis (0)]
63.  Hoofnagle JH, Doo E, Liang TJ, Fleischer R, Lok AS. Management of hepatitis B: summary of a clinical research workshop. Hepatology. 2007;45:1056-1075.  [PubMed]  [DOI]  [Cited in This Article: ]
64.  European Association For The Study Of The Liver. EASL Clinical Practice Guidelines: management of chronic hepatitis B. J Hepatol. 2009;50:227-242.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1152]  [Cited by in F6Publishing: 1122]  [Article Influence: 74.8]  [Reference Citation Analysis (0)]
65.  Seto WK, Wong DK, Fung J, Ip PP, Yuen JC, Hung IF, Lai CL, Yuen MF. High hepatitis B surface antigen levels predict insignificant fibrosis in hepatitis B e antigen positive chronic hepatitis B. PLoS One. 2012;7:e43087.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 57]  [Cited by in F6Publishing: 62]  [Article Influence: 5.2]  [Reference Citation Analysis (0)]
66.  Martinot-Peignoux M, Carvalho-Filho R, Lapalus M, Netto-Cardoso AC, Lada O, Batrla R, Krause F, Asselah T, Marcellin P. Hepatitis B surface antigen serum level is associated with fibrosis severity in treatment-naïve, e antigen-positive patients. J Hepatol. 2013;58:1089-1095.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 80]  [Cited by in F6Publishing: 81]  [Article Influence: 7.4]  [Reference Citation Analysis (0)]
67.  Chen CJ, Yang HI, Iloeje UH. Hepatitis B virus DNA levels and outcomes in chronic hepatitis B. Hepatology. 2009;49:S72-S84.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 236]  [Cited by in F6Publishing: 238]  [Article Influence: 15.9]  [Reference Citation Analysis (0)]
68.  Chen CJ, Yang HI, Su J, Jen CL, You SL, Lu SN, Huang GT, Iloeje UH. Risk of hepatocellular carcinoma across a biological gradient of serum hepatitis B virus DNA level. JAMA. 2006;295:65-73.  [PubMed]  [DOI]  [Cited in This Article: ]
69.  Su IJ, Wang HC, Wu HC, Huang WY. Ground glass hepatocytes contain pre-S mutants and represent preneoplastic lesions in chronic hepatitis B virus infection. J Gastroenterol Hepatol. 2008;23:1169-1174.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 95]  [Cited by in F6Publishing: 108]  [Article Influence: 6.8]  [Reference Citation Analysis (0)]
70.  Huy TT, Ushijima H, Win KM, Luengrojanakul P, Shrestha PK, Zhong ZH, Smirnov AV, Taltavull TC, Sata T, Abe K. High prevalence of hepatitis B virus pre-s mutant in countries where it is endemic and its relationship with genotype and chronicity. J Clin Microbiol. 2003;41:5449-5455.  [PubMed]  [DOI]  [Cited in This Article: ]
71.  Fan YF, Lu CC, Chen WC, Yao WJ, Wang HC, Chang TT, Lei HY, Shiau AL, Su IJ. Prevalence and significance of hepatitis B virus (HBV) pre-S mutants in serum and liver at different replicative stages of chronic HBV infection. Hepatology. 2001;33:277-286.  [PubMed]  [DOI]  [Cited in This Article: ]
72.  Caselmann WH, Meyer M, Kekulé AS, Lauer U, Hofschneider PH, Koshy R. A trans-activator function is generated by integration of hepatitis B virus preS/S sequences in human hepatocellular carcinoma DNA. Proc Natl Acad Sci USA. 1990;87:2970-2974.  [PubMed]  [DOI]  [Cited in This Article: ]
73.  Hildt E, Saher G, Bruss V, Hofschneider PH. The hepatitis B virus large surface protein (LHBs) is a transcriptional activator. Virology. 1996;225:235-239.  [PubMed]  [DOI]  [Cited in This Article: ]
74.  Chisari FV, Filippi P, Buras J, McLachlan A, Popper H, Pinkert CA, Palmiter RD, Brinster RL. Structural and pathological effects of synthesis of hepatitis B virus large envelope polypeptide in transgenic mice. Proc Natl Acad Sci USA. 1987;84:6909-6913.  [PubMed]  [DOI]  [Cited in This Article: ]
75.  Kekulé AS, Lauer U, Meyer M, Caselmann WH, Hofschneider PH, Koshy R. The preS2/S region of integrated hepatitis B virus DNA encodes a transcriptional transactivator. Nature. 1990;343:457-461.  [PubMed]  [DOI]  [Cited in This Article: ]
76.  Alka S, Hemlata D, Vaishali C, Shahid J, Kumar PS. Hepatitis B virus surface (S) transactivator with DNA-binding properties. J Med Virol. 2000;61:1-10.  [PubMed]  [DOI]  [Cited in This Article: ]
77.  Meyer M, Caselmann WH, Schlüter V, Schreck R, Hofschneider PH, Baeuerle PA. Hepatitis B virus transactivator MHBst: activation of NF-kappa B, selective inhibition by antioxidants and integral membrane localization. EMBO J. 1992;11:2991-3001.  [PubMed]  [DOI]  [Cited in This Article: ]
78.  Lauer U, Weiss L, Lipp M, Hofschneider PH, Kekulé AS. The hepatitis B virus preS2/St transactivator utilizes AP-1 and other transcription factors for transactivation. Hepatology. 1994;19:23-31.  [PubMed]  [DOI]  [Cited in This Article: ]
79.  Hildt E, Munz B, Saher G, Reifenberg K, Hofschneider PH. The PreS2 activator MHBs(t) of hepatitis B virus activates c-raf-1/Erk2 signaling in transgenic mice. EMBO J. 2002;21:525-535.  [PubMed]  [DOI]  [Cited in This Article: ]
80.  Lai MW, Yeh CT. The oncogenic potential of hepatitis B virus rtA181T/ surface truncation mutant. Antivir Ther. 2008;13:875-879.  [PubMed]  [DOI]  [Cited in This Article: ]
81.  Huang SF, Chen YT, Lee WC, Chang IC, Chiu YT, Chang Y, Tu HC, Yuh CH, Matsuura I, Shih LY. Identification of transforming hepatitis B virus S gene nonsense mutations derived from freely replicative viruses in hepatocellular carcinoma. PLoS One. 2014;9:e89753.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 16]  [Cited by in F6Publishing: 19]  [Article Influence: 1.9]  [Reference Citation Analysis (0)]
82.  Lee SA, Kim H, Won YS, Seok SH, Na Y, Shin HB, Inn KS, Kim BJ. Male-specific hepatitis B virus large surface protein variant W4P potentiates tumorigenicity and induces gender disparity. Mol Cancer. 2015;14:23.  [PubMed]  [DOI]  [Cited in This Article: ]
83.  Li ZQ, Linghu E, Jun W, Cheng J. Screening of hepatocyte proteins binding with C-terminally truncated surface antigen middle protein of hepatitis B virus (MHBst167) by a yeast two-hybrid system. Mol Med Rep. 2014;10:1259-1263.  [PubMed]  [DOI]  [Cited in This Article: ]
84.  Awe K, Lambert C, Prange R. Mammalian BiP controls posttranslational ER translocation of the hepatitis B virus large envelope protein. FEBS Lett. 2008;582:3179-3184.  [PubMed]  [DOI]  [Cited in This Article: ]
85.  Su IJ, Wang LH, Hsieh WC, Wu HC, Teng CF, Tsai HW, Huang W. The emerging role of hepatitis B virus pre-S2 deletion mutant proteins in HBV tumorigenesis. J Biomed Sci. 2014;21:98.  [PubMed]  [DOI]  [Cited in This Article: ]
86.  Wang HC, Wu HC, Chen CF, Fausto N, Lei HY, Su IJ. Different types of ground glass hepatocytes in chronic hepatitis B virus infection contain specific pre-S mutants that may induce endoplasmic reticulum stress. Am J Pathol. 2003;163:2441-2449.  [PubMed]  [DOI]  [Cited in This Article: ]
87.  Ono M, Morisawa K, Nie J, Ota K, Taniguchi T, Saibara T, Onishi S. Transactivation of transforming growth factor alpha gene by hepatitis B virus preS1. Cancer Res. 1998;58:1813-1816.  [PubMed]  [DOI]  [Cited in This Article: ]
88.  Hadziyannis S, Gerber MA, Vissoulis C, Popper H. Cytoplasmic hepatitis B antigen in “ground-glass” hepatocytes of carriers. Arch Pathol. 1973;96:327-330.  [PubMed]  [DOI]  [Cited in This Article: ]
89.  Shikata T. Australia antigen in liver tissue--an immunofluorescent and immunoelectron microscopic study. Jpn J Exp Med. 1973;43:231-245.  [PubMed]  [DOI]  [Cited in This Article: ]
90.  Malhi H, Kaufman RJ. Endoplasmic reticulum stress in liver disease. J Hepatol. 2011;54:795-809.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 811]  [Cited by in F6Publishing: 870]  [Article Influence: 66.9]  [Reference Citation Analysis (0)]
91.  Tsai HW, Lin YJ, Lin PW, Wu HC, Hsu KH, Yen CJ, Chan SH, Huang W, Su IJ. A clustered ground-glass hepatocyte pattern represents a new prognostic marker for the recurrence of hepatocellular carcinoma after surgery. Cancer. 2011;117:2951-2960.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 50]  [Cited by in F6Publishing: 51]  [Article Influence: 3.9]  [Reference Citation Analysis (0)]
92.  Li B, Gao B, Ye L, Han X, Wang W, Kong L, Fang X, Zeng Y, Zheng H, Li S. Hepatitis B virus X protein (HBx) activates ATF6 and IRE1-XBP1 pathways of unfolded protein response. Virus Res. 2007;124:44-49.  [PubMed]  [DOI]  [Cited in This Article: ]
93.  Lazar C, Uta M, Branza-Nichita N. Modulation of the unfolded protein response by the human hepatitis B virus. Front Microbiol. 2014;5:433.  [PubMed]  [DOI]  [Cited in This Article: ]
94.  Li J, Liu Y, Wang Z, Liu K, Wang Y, Liu J, Ding H, Yuan Z. Subversion of cellular autophagy machinery by hepatitis B virus for viral envelopment. J Virol. 2011;85:6319-6333.  [PubMed]  [DOI]  [Cited in This Article: ]
95.  Ehrly AM, Seebens H, Saeger-Lorenz K. [Effect of a 10% and 6% hydroxyethyl starch solution (molecular weight 200,000/0.62) in comparison with a 10% dextran solution (molecular weight 40,000) on flow properties of blood and tissue oxygen pressure in patients with intermittent claudication]. Infusionstherapie. 1988;15:181-187.  [PubMed]  [DOI]  [Cited in This Article: ]
96.  Yang JC, Teng CF, Wu HC, Tsai HW, Chuang HC, Tsai TF, Hsu YH, Huang W, Wu LW, Su IJ. Enhanced expression of vascular endothelial growth factor-A in ground glass hepatocytes and its implication in hepatitis B virus hepatocarcinogenesis. Hepatology. 2009;49:1962-1971.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 87]  [Cited by in F6Publishing: 96]  [Article Influence: 6.4]  [Reference Citation Analysis (0)]
97.  Wu HC, Tsai HW, Teng CF, Hsieh WC, Lin YJ, Wang LH, Yuan Q, Su IJ. Ground-glass hepatocytes co-expressing hepatitis B virus X protein and surface antigens exhibit enhanced oncogenic effects and tumorigenesis. Hum Pathol. 2014;45:1294-1301.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 25]  [Cited by in F6Publishing: 26]  [Article Influence: 2.6]  [Reference Citation Analysis (0)]
98.  Cornu M, Albert V, Hall MN. mTOR in aging, metabolism, and cancer. Curr Opin Genet Dev. 2013;23:53-62.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 333]  [Cited by in F6Publishing: 353]  [Article Influence: 32.1]  [Reference Citation Analysis (0)]
99.  Wang HC, Chang WT, Chang WW, Wu HC, Huang W, Lei HY, Lai MD, Fausto N, Su IJ. Hepatitis B virus pre-S2 mutant upregulates cyclin A expression and induces nodular proliferation of hepatocytes. Hepatology. 2005;41:761-770.  [PubMed]  [DOI]  [Cited in This Article: ]
100.  Wang LH, Huang W, Lai MD, Su IJ. Aberrant cyclin A expression and centrosome overduplication induced by hepatitis B virus pre-S2 mutants and its implication in hepatocarcinogenesis. Carcinogenesis. 2012;33:466-472.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 55]  [Cited by in F6Publishing: 57]  [Article Influence: 4.4]  [Reference Citation Analysis (0)]
101.  Hsieh YH, Hsu JL, Su IJ, Huang W. Genomic instability caused by hepatitis B virus: into the hepatoma inferno. Front Biosci (Landmark Ed). 2011;16:2586-2597.  [PubMed]  [DOI]  [Cited in This Article: ]
102.  Chisari FV, Klopchin K, Moriyama T, Pasquinelli C, Dunsford HA, Sell S, Pinkert CA, Brinster RL, Palmiter RD. Molecular pathogenesis of hepatocellular carcinoma in hepatitis B virus transgenic mice. Cell. 1989;59:1145-1156.  [PubMed]  [DOI]  [Cited in This Article: ]
103.  Günther S, Fischer L, Pult I, Sterneck M, Will H. Naturally occurring variants of hepatitis B virus. Adv Virus Res. 1999;52:25-137.  [PubMed]  [DOI]  [Cited in This Article: ]
104.  Zhong S, Chan JY, Yeo W, Tam JS, Johnson PJ. Hepatitis B envelope protein mutants in human hepatocellular carcinoma tissues. J Viral Hepat. 1999;6:195-202.  [PubMed]  [DOI]  [Cited in This Article: ]
105.  Woltman AM, Op den Brouw ML, Biesta PJ, Shi CC, Janssen HL. Hepatitis B virus lacks immune activating capacity, but actively inhibits plasmacytoid dendritic cell function. PLoS One. 2011;6:e15324.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 105]  [Cited by in F6Publishing: 116]  [Article Influence: 8.9]  [Reference Citation Analysis (2)]
106.  Xiang Z, Wang S, Xiang Y. Up-regulated microRNA499a by hepatitis B virus induced hepatocellular carcinogenesis via targeting MAPK6. PLoS One. 2014;9:e111410.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 15]  [Cited by in F6Publishing: 17]  [Article Influence: 1.7]  [Reference Citation Analysis (0)]
107.  Liu J, Yang HI, Lee MH, Lu SN, Jen CL, Wang LY, You SL, Iloeje UH, Chen CJ. Incidence and determinants of spontaneous hepatitis B surface antigen seroclearance: a community-based follow-up study. Gastroenterology. 2010;139:474-482.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 192]  [Cited by in F6Publishing: 183]  [Article Influence: 13.1]  [Reference Citation Analysis (0)]
108.  Chu CM, Liaw YF. HBsAg seroclearance in asymptomatic carriers of high endemic areas: appreciably high rates during a long-term follow-up. Hepatology. 2007;45:1187-1192.  [PubMed]  [DOI]  [Cited in This Article: ]
109.  Colak D, Chishti MA, Al-Bakheet AB, Al-Qahtani A, Shoukri MM, Goyns MH, Ozand PT, Quackenbush J, Park BH, Kaya N. Integrative and comparative genomics analysis of early hepatocellular carcinoma differentiated from liver regeneration in young and old. Mol Cancer. 2010;9:146.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 27]  [Cited by in F6Publishing: 21]  [Article Influence: 1.5]  [Reference Citation Analysis (0)]
110.  Bruix J, Boix L, Sala M, Llovet JM. Focus on hepatocellular carcinoma. Cancer Cell. 2004;5:215-219.  [PubMed]  [DOI]  [Cited in This Article: ]
111.  Wu CY, Chen YJ, Ho HJ, Hsu YC, Kuo KN, Wu MS, Lin JT. Association between nucleoside analogues and risk of hepatitis B virus-related hepatocellular carcinoma recurrence following liver resection. JAMA. 2012;308:1906-1914.  [PubMed]  [DOI]  [Cited in This Article: ]
112.  Manesis EK, Papatheodoridis GV, Tiniakos DG, Hadziyannis ES, Agelopoulou OP, Syminelaki T, Papaioannou C, Nastos T, Karayiannis P. Hepatitis B surface antigen: relation to hepatitis B replication parameters in HBeAg-negative chronic hepatitis B. J Hepatol. 2011;55:61-68.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 49]  [Cited by in F6Publishing: 55]  [Article Influence: 4.2]  [Reference Citation Analysis (0)]
113.  Chevaliez S, Hézode C, Bahrami S, Grare M, Pawlotsky JM. Long-term hepatitis B surface antigen (HBsAg) kinetics during nucleoside/nucleotide analogue therapy: finite treatment duration unlikely. J Hepatol. 2013;58:676-683.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 178]  [Cited by in F6Publishing: 187]  [Article Influence: 17.0]  [Reference Citation Analysis (0)]
114.  Zoutendijk R, Hansen BE, van Vuuren AJ, Boucher CA, Janssen HL. Serum HBsAg decline during long-term potent nucleos(t)ide analogue therapy for chronic hepatitis B and prediction of HBsAg loss. J Infect Dis. 2011;204:415-418.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 105]  [Cited by in F6Publishing: 110]  [Article Influence: 8.5]  [Reference Citation Analysis (0)]
115.  Vlachogiannakos J, Papatheodoridis G. Hepatocellular carcinoma in chronic hepatitis B patients under antiviral therapy. World J Gastroenterol. 2013;19:8822-8830.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 32]  [Cited by in F6Publishing: 30]  [Article Influence: 2.7]  [Reference Citation Analysis (0)]
116.  Warner N, Locarnini S, Nguyen T. Anti-viral Medication to Prevent HCC Development: Where Are We Now? Cancer Forum. 2009;33:111-114.  [PubMed]  [DOI]  [Cited in This Article: ]
117.  Li Y, Zhu M, Guo Y, Chen W, Li G. Full-length hepatitis B virus sequences from naïve patients with fluctuation of viral load during ADV monotherapy. Virus Genes. 2010;40:155-162.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3]  [Cited by in F6Publishing: 2]  [Article Influence: 0.1]  [Reference Citation Analysis (0)]
118.  Pollicino T, Cacciola I, Saffioti F, Raimondo G. Hepatitis B virus PreS/S gene variants: pathobiology and clinical implications. J Hepatol. 2014;61:408-417.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 175]  [Cited by in F6Publishing: 186]  [Article Influence: 18.6]  [Reference Citation Analysis (0)]
119.  Zhang D, Dong P, Zhang K, Deng L, Bach C, Chen W, Li F, Protzer U, Ding H, Zeng C. Whole genome HBV deletion profiles and the accumulation of preS deletion mutant during antiviral treatment. BMC Microbiol. 2012;12:307.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 30]  [Cited by in F6Publishing: 32]  [Article Influence: 2.7]  [Reference Citation Analysis (0)]
120.  Reijnders JG, Rijckborst V, Sonneveld MJ, Scherbeijn SM, Boucher CA, Hansen BE, Janssen HL. Kinetics of hepatitis B surface antigen differ between treatment with peginterferon and entecavir. J Hepatol. 2011;54:449-454.  [PubMed]  [DOI]  [Cited in This Article: ]
121.  Jaroszewicz J, Ho H, Markova A, Deterding K, Wursthorn K, Schulz S, Bock CT, Tillmann HL, Manns MP, Wedemeyer H. Hepatitis B surface antigen (HBsAg) decrease and serum interferon-inducible protein-10 levels as predictive markers for HBsAg loss during treatment with nucleoside/nucleotide analogues. Antivir Ther. 2011;16:915-924.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 66]  [Cited by in F6Publishing: 73]  [Article Influence: 6.1]  [Reference Citation Analysis (0)]
122.  HBF Drug Watch.  Available from: http://www.hepb.org/professionals/hbf_drug_watch.htm.  [PubMed]  [DOI]  [Cited in This Article: ]
123.  Zoulim F, Durantel D. Antiviral therapies and prospects for a cure of chronic hepatitis B. Cold Spring Harb Perspect Med. 2015;5.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 104]  [Cited by in F6Publishing: 117]  [Article Influence: 13.0]  [Reference Citation Analysis (0)]
124.  Fribley A, Wang CY. Proteasome inhibitor induces apoptosis through induction of endoplasmic reticulum stress. Cancer Biol Ther. 2006;5:745-748.  [PubMed]  [DOI]  [Cited in This Article: ]
125.  Wang Z, Jin W, Jin H, Wang X. mTOR in viral hepatitis and hepatocellular carcinoma: function and treatment. Biomed Res Int. 2014;2014:735672.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 11]  [Cited by in F6Publishing: 18]  [Article Influence: 1.8]  [Reference Citation Analysis (0)]
126.  Malhotra JD, Miao H, Zhang K, Wolfson A, Pennathur S, Pipe SW, Kaufman RJ. Antioxidants reduce endoplasmic reticulum stress and improve protein secretion. Proc Natl Acad Sci USA. 2008;105:18525-18530.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 502]  [Cited by in F6Publishing: 529]  [Article Influence: 33.1]  [Reference Citation Analysis (0)]
127.  Tsai JJ, Kuo HC, Lee KF, Tsai TH. Glycyrrhizin represses total parenteral nutrition-associated acute liver injury in rats by suppressing endoplasmic reticulum stress. Int J Mol Sci. 2013;14:12563-12580.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 17]  [Cited by in F6Publishing: 17]  [Article Influence: 1.5]  [Reference Citation Analysis (0)]
128.  Kumada H. Long-term treatment of chronic hepatitis C with glycyrrhizin [stronger neo-minophagen C (SNMC)] for preventing liver cirrhosis and hepatocellular carcinoma. Oncology. 2002;62 Suppl 1:94-100.  [PubMed]  [DOI]  [Cited in This Article: ]
129.  Li JY, Cao HY, Liu P, Cheng GH, Sun MY. Glycyrrhizic acid in the treatment of liver diseases: literature review. Biomed Res Int. 2014;2014:872139.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 78]  [Cited by in F6Publishing: 117]  [Article Influence: 11.7]  [Reference Citation Analysis (0)]
130.  Sato H, Goto W, Yamamura J, Kurokawa M, Kageyama S, Takahara T, Watanabe A, Shiraki K. Therapeutic basis of glycyrrhizin on chronic hepatitis B. Antiviral Res. 1996;30:171-177.  [PubMed]  [DOI]  [Cited in This Article: ]