Hepatitis B virus preS/S gene mutations and their clinical implications
Review Article

Hepatitis B virus preS/S gene mutations and their clinical implications

Junzhong Wang1, Bin Zhu1, Mengji Lu2, Dongliang Yang1

1Department of Infectious Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; 2Institute of Virology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany

Contributions: (I) Conception and design: J Wang, D Yang; (II) Administrative support: M Lu, D Yang; (III) Provision of study materials or patients: M Lu, D Yang; (IV) Collection and assembly of data: M Lu, D Yang; (V) Data analysis and interpretation: J Wang, B Zhu; (VI) Manuscript writing: All authors; (VII) Final approval of manuscript: All authors.

Correspondence to: Prof. Dr. Dongliang Yang. Department of Infectious Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China. Email: dlyang@hust.edu.cn.

Abstract: Hepatitis B virus (HBV) infection remains a major health problem worldwide. HBV is one of the smallest enveloped DNA viruses, and also one of the principal pathogens causing acute and chronic hepatitis. Hepatitis B surface antigen (HBsAg) is a hallmark for the diagnosis of HBV infection, and the quantification of serum HBsAg is regarded as a reliable marker of disease progression and predictor of the outcome. However, the mutations in the preS/S genomic region can occur in HBV DNA sequences from the patients with chronic HBV infection, including genetic recombination, base pair deletion, and point mutations. These variants carrying the modified surface antigen may induce immune escape or occult HBV infection (OBI). Furthermore, the preS/S variants may be the cause of fulminant hepatitis (FH) and fibrosing cholestatic hepatitis (FCH). The mutations in the preS/S region would increase the difficulty of preventing and treatment of HBV infection. In this review, we summarized the prevalence of the mutations in the preS/S region and their effect for the diagnosis or progression of HBV infection.

Keywords: Hepatitis B virus (HBV); mutations in preS/S region; immune escape; occult HBV infection (OBI)


Received: 13 April 2017; Accepted: 01 September 2017; Published: 06 November 2017.

doi: 10.21037/aob.2017.10.01


Introduction

The human hepatitis B virus (HBV) is one of the smallest enveloped DNA viruses, and one of the principal pathogens causing acute and chronic hepatitis. Although prophylactic vaccine can prevent the HBV infection and reduce the incidence effectively, about 2% of the vaccinated people in some areas can develop chronic HBV infection. There are about 240 million people suffering from chronic HBV infection worldwide, and nearly 25% of whom have chronic liver disease and/or cirrhosis, which could progress to hepatocellular carcinoma. So HBV infection remains a major health problem worldwide (1,2). Hepatitis B surface antigen (HBsAg) is a hallmark for the diagnosis of HBV infection, and the quantification of serum HBsAg is regarded as a reliable marker of disease progression and predictor of the outcome. In addition, the clearance of HBsAg and the HBsAg-seroconversion are believed to be the ultimate goals of antiviral therapy, as they represent that host immune system can control the active HBV replication successfully (3). A large amount of evidence indicated that the emergence of mutations in the PreS/S genomic region, including amid acids substitution, insertion and deletion, is a frequent event that may be occur as the consequence of antiviral treatment or immunoprophylaxis, or occur spontaneously (4). In this review, we will describe the prevalence of the preS/S gene mutations and their clinic implications.


HBV virology and the feature of preS/S

HBV is a member of the Hepadnaviridae family, and characterized by a high liver tropism and species-specificity. Only the human been and chimpanzees are fully susceptible to infection with HBV, and the liver is thought to be the exclusive target organ for HBV virus, although a small amount of HBV has been found in other tissues such as mononuclear cells, pancreas, and kidney. HBV virion is an approximately 42 nM particle in diameter and its genome is a partially double-stranded relaxed, circular DNA (rcDNA) which comprised by approximately 3.2 kb base pairs (5). HBV genome is located in the nucleocapsid (core) which is surrounded by the envelop proteins. There are four partially overlapping open-reading frames (ORF) in the HBV genome: the preS/S, preCore/Core, Pol and X, and they encode the viral envelope proteins, the preCore and Core protein, viral polymerase, and the regulatory X protein respectively. Particularly, the preS/S ORF can be translated from three different starting codons and encode three different envelope proteins respectively (Larger, Middle, and Small protein). So the C-terminal domains of the three proteins are the same, but the N-terminal extensions are different. The Larger protein (L) consists of 389 to 400 aa depending on the different serotypes, and Middle (M) protein consists of 281 aa and Small (S) protein consists of 226 aa. The 55 hydrophilic aa at the N-terminal domain of the M protein is named preS2, and the extension of hydrophilic residues containing 108 or 119 aa (depending on the genotype) at the N-terminal domain L protein is called preS1 (6,7).

The HBV envelope proteins are typical membrane proteins and have a relatively complex topology. The envelope protein synthesis is performed at the endoplasmic reticulum (ER) via a distinct mechanism that is different from viral replication. Three distinct particles containing envelop proteins can be found in the serum of HBV infected patients, spherical particles, filamentous particles, and Dane particles. The first two particles are formed by the HBV envelope proteins but don’t contain the HBV genome, so they lost the capacity of infection. The Dane particles are infectious virions containing complete construction and HBV genome. The spherical and filamentous particles are termed as subviral particles (SVP) and their amount is 1,000 to 10,000 times as the infectious virions. The SVPs can form the immune complexes circulating the host body and induce small vessel vasculitis; also, the SVPs can promote the host immune tolerance (5,7). Meanwhile, most of the envelope proteins present both in SVP and virions are S protein, only 20% of the envelope proteins are constituted by M and L protein. Notably, nearly all of the commercial HBsAg quantification assays use the antibodies which target the epitopes in the S protein, so these quantitative immunoassays can detect all of circulating envelope proteins including the immune complexes, but not distinguish the HBsAg in the virions or in the SVPs (8-10).

Three highly conserved areas in HBsAg are important for the HBV genotyping and sub-genotyping which located on the residues 25–43, 69–109 and 144–157. Genotype-specific substitutions reside in between these locations in a bare major hydrophilic region (MHR) which extend from 110–155 residues and encompasses the “a” determinant, and this determinant is the crucial site for the attachment of anti-HBs antibody (5,11). The mutations in “a” determinant are common and can reduce its ability of binding to anti-HBs polyclonal and/or monoclonal antibodies which used in commercial diagnostic assays, and finally result in failure to detect HBsAg. In addition to the point mutations have been found in the HBsAg, nucleotides insertion or deletion can also occur and induce frame shift mutations. The cysteines are conserved in the HBsAg aa sequence and are very important for the formation of the loops in the “a” determinant. The mutations can lead the replacement of cysteine residues with serine, and consequently result in the changes of the molecule conformation and antigenicity lost. The lysine/arginine substitutions at residues 122 or 160 can determine the variations of the d/y or w/r subtype. The hypervariability of the HBV genome can help HBV to escape from the selection pressures caused by the host immune system, vaccination, and antiviral treatments. The genome of HBV strains is diversity worldwide and we can separate these strains into different genotypes, subgenotypes and subtypes according to the sequence of HBsAg. Mutations within HBsAg may decrease its ability recognizing and binding by neutralizing antibody. Detection of HBsAg is affected by several factors, including the HBV genotypes or subtypes, the sensitivity and specificity of the immunoassay, the capture and detection antibodies, and the conditions of immunoassay (4,12,13).


The preS/S mutation prevalence and immune escape

The preS/S sequences exhibit the highest heterogeneity of the HBV genome. Many studies have confirmed that genetic recombination, base pair deletion, and point mutations in the preS/S regions can occur in HBV DNA sequences from the patients with chronic HBV infection. In some areas, the mutations in the preS/S are up to 10.8–11.2% among the patients who did not receive vaccine or HBIg (14,15). The plasma-derived vaccines can induce significantly higher prevalence of the “a” determinant mutations in children than the recombinant vaccines. The variants and the wild type virus can co-exist in the host. The vaccine-induced anti-HBs antibodies will be waning gradually, and it targets the variants initially. So the variants will be taken over, and the wild type virus will eventually become the predominant quasispecies (9,16). Several studies from the countries that implemented the Expanded Program on Immunization have confirmed that, people can be infected by variants with the G145R mutation or some other mutations in HBsAg “a” determinant, even those people have vaccinated and developed the anti-HBs antibodies successfully. But the prevalence of the preS/S mutations is different among these studies. Many factors may contribute to the discrepancies between these results, including the regional differences, the vaccination strategies and lasting time of the immunization program. The study in Singapore shows that the prevalence of “a” determinant variants was 39%, but in Taiwan it was 21.22%, in UK it was 22.12%, and in South Africa it was 0%. Interestingly, “a” determinant variants are rarely found in the area with high HBV prevalence, even from the individuals who have accepted the vaccination and were borne to HBV infected mothers. Moreover, the truth of prevalence of the “a” determinant variants can decrease the efficacy of vaccination has not caught people’s attention, because vaccine-induced variants are hardly to induce HBV infection breakthrough. The key factors contribute to the vaccination failure include the host gene background, babies infected with HBV before delivery, babies born by the mothers with high viral load, improper vaccination strategy and so on. Taken together, the results from these different studies illuminated that the prevalence of vaccine escape variants were negligible during the practice of the vaccination program worldwide (9,17-20).

For the antiviral therapy, nucleos(t)ide analogous (NA) can suppress HBV replication and reduce the alanine aminotransferase activity quickly. But NAs need to be administrated for long-term as they can’t eliminate the cccDNA in the nucleus of hepatocytes. Long-term antiviral treatment with NAs induces the polymerase gene mutate and leads to the emergence of the drug-resistance. Drug-resistance mutation is a common problem among the patients receiving NAs treatment. Because of the existence of the overlapping of the polymerase gene and the surface gene, the compensatory mutations can occur in the surface gene during the NAs treatment. Some of these mutations are located in the area of ‘a’ determinant and can help the HBV to escape from the host immunity (21).

The distal part of the A-B inter-domain had the overlapping areas accompanying with the B domain of RT (residues 80–236); and residues 72–228 of the RT and surface proteins, respectively. The aa residues of 208, 51, 52, 56, and 120 are notorious for various surface therapy-escape mutations of which the rtA181T/sW172 mutant has a dominant negative secretion effect which leads to the clearance of HBV antigen from the serum, where vaccine-escape-like mutants might be selected (21,22). Many studies have compared the replication capabilities of the wild type and surface mutants. The results showed that these mutants, including E164D, I195M, W196S, M198I, E164D, and I195M, which were selected during lamivudine treatment, had the minimal capabilities to bind to anti-HBs antibody, just as the mutant G145 which can escape from the vaccine immunity. So the variants selected by lamivudine may not be neutralized by the anti-HBs antibody which was induced by vaccine. For the other NAs, including adefovir, tenofovir, entecavir, and telbivudine, it’s not clear whether these antivirals have the potential to select HBV variants which reduce the capabilities of binding to the anti-HBs antibody (9,23) (Table 1).

Table 1

PreS/S mutations and their clinical implications

Mutations Clinical implications
E164D (24), I195M (24), M198I (24), W196S (24), T116N (25), P120S/E (26), I/T126A/N/I/S (27-29), Q129H/R (29-31), M133L/T (31,32), K141E (33), P142S (33), D144A/E (32,34), G145R/A (29,31,35,36), M103I (34), L109I (34), T118K (34), P120A (34), Y134H/N (34,37), S143L (34), S171F (34), C48G (34), V96A (34), L175S (34), G185E (34), V190A (34), L108V/I (31), T87I (31), I84T (31), A91T (31), T68V (31), S78N (31), V60A (31), T31I/T (31), P34N/T (31), G31E (31), Q2K/B (31), T140I (31,37), G130N (29), F134I (29) Immune escape
Y100S (38,39), Q101R (38), P105R (39,40), T115N/A (31,41), T116N (38), P120L (42), R122P (38), T123N/A (31,43), I126S/T (44), P127H/L (45), Q129P/R (29,31,46,47), M133T (48), S143L (38), S167L (38), R169H (48), G145R/A (31,48), C124Y (49), D144A (50), G119R (49), C124Y (49), I126S/A (31,49), C139R (51), S78N (31), L108I (31), A90V (31), Q118L (31), T87S (31), A90T (31), KL45F (31), P47T (31), N56T (31), G73E (31), P94S (31), I110L (31), S117N (31), S114T (52), P127S/T (52), M133T (52) OBI
A1762T (53), G1764A (53), G1862T (53), G1896A (53) Fulminant hepatitis

The preS/S mutation and occult HBV infection (OBI)

As the development of the high sensitive techniques, HBV DNA can be detected from the patients with HBsAg negative in the serum. We defined the individuals with HBV DNA positive persistently in the liver and HBsAg negative as OBI, whatever the HBV DNA in the serum is positive or negative (13). The mechanisms of OBI are not clear yet. Patients infected with HBV variants which had defective replication activity or cannot express S antigen may develop OBI. But most of the HBV quasispecies isolated from the OBI patients were replication-competent viruses. Patients infected with HBV variants producing surface antigen which cannot be recognized by the conventional detection assays may explain the development of OBI. But these explanations need to be confirmed further (8,13,54).

In recent years, OBI has attracted people’s attention worldwide because it may be a threat of the HBV infection in transfusion medicine. The HBV strains from OBI patients cannot be detected by the commercial assays, so these OBI patients may be selected as blood donors and their blood containing HBV may transfuse to the other patients. In the blood donors, the prevalence of OBI ranges from 0.0002% to 0.084% in Europe. China is one of the highest HBV endemic regions, and the prevalence of OBI in the blood donors is as high as 0.18% (38,55-58). Most of the studies about HBV mutation just focused on limited parts of the HBV genome, so we can’t get enough information to analyze relationship between mutation and OBI. A recent survey showed that the prevalence of specific mutation in OBI patients was 8.3–20.8%, but in the overt infected patients it ranged from 0% to 3.7%. It’s worth to mention that there is no unified standard to determine which mutations are associated with OBI. Some scholars considered the mutations located in “a” determinant as OBI-associated mutations, but others thought they may include the mutations in the whole MHR (13,38).

A study including more the 400,000 blood-donors found that twenty mutations in genotype D strains were associated with OBI strongly. Most of these mutations were located in “a” determinant, but can also be found in other area of HBsAg. Interestingly, these mutations reported in this study were quite district from those found in OBI patients who were infected with genotypes B or C. In the author’s opinion, the mutations associated with OBI may be unique among every genotype. Nevertheless, the mutation G145R/A was thought to be one of the major mutations associated with OBI, and it could be found both in genotypes B and C (49,59,60). On the other hand, the mutations in the MHR were found to be responsible for decreasing the surface antigen levels in serum. Huang et al and colleges found some mutations in the MHR can reduce the detection sensitivity of commercial assays, and others can influence the secretion of surface antigen and/or virion itself (49).

Cassini et al isolated HBV strain from liver samples and found C695T mutation which can lead to the appearance of stop codon in HBs gene at aa181. This additional stop codon can result in truncated S protein production, and decrease the levels of HBsAg both in the serum and in the liver. In this study, HBV DNA can be found in the peripheral blood mononuclear cells (PBMCs) and liver tissue gotten from the OBI patients. So the authors strongly suggested that testing the PBMCs and liver tissue can help for diagnosis of OBI (61). In addition, preS antigen also played a crucial role for the expression, synthesis, and secretion of the S protein. And the mutation in preS may have the potential association with the OBI development. The effect of preS region for the OBI development needs to be investigated further (60,62) (Table 1).


The preS/S mutation in acute and chronic hepatitis B

More and more evidence showed that the mutations in preS and S region can be found in severe forms of HBV related acute and chronic liver diseases, and these mutations can affect the development or even the outcome of diseases. Indeed, mutations within preS can be found in a majority of patients chronic infected with HBV. Several genetic defects may be responsible for the mutation emergence within preS, including deletions of aa in the preS1 and/or preS2 area, and point mutations. The point mutations at the start codon of preS2 also bad been found and it prevented M protein expression completely (63,64).

Mutations in preS, especially that abolish the expression of M protein, have been found in the patients with fulminant hepatitis (FH). Pollicino et al. and colleges analyzed the whole genome of HBV isolated from a surgeon and his mother, both of them suffered from FH and died of it. They found the viruses from these two patients had a double mutation in the preS2 start codon (ATG→ACA) which prevent the production of the M protein (65). Several other studies also found the preS2 defective viruses from the patients of FH, and their outcomes were similar with reports by Pollicino et al. (66,67). These studies showed that infection by preS2 defective virus is frequently associated with FH, indicating that this variant might play a pathogenetic role in cases of acute liver failure. Until now, people can’t confirm that preS2 defective viruses can induce acute liver failure in the animal models. In the HBV transgenic mouse model, HBV surface proteins accumulated in the hepatocytes, and lead the hepatocytes to enhance the sensitivity to IFN-γ produced by the cytotoxic T lymphocytes. The livers from these mice showed pan lobular necrosis and hepatic failure. According to it, we can get a hypothesis that preS2 defective viruses which can’t express M protein may overproduce L protein, which promote the L protein accumulated in the hepatocytes and induce hepatic failure occurred. In addition, the specific immune response for M protein, including the T cell response and B cell response, are very important during the early stage of HBV infection. The viruses without M protein may be targeted ineffectively by the specific T cells, which may result in more severe course of the infection (4). These hypotheses can make an explanation for the association between preS2 defective mutations and the FH, but they need to be confirmed by experimental studies.

Fibrosing cholestatic hepatitis (FCH) is a rapidly progressive and usually fatal form of viral hepatitis, and characterized by specific histologic manifestation of HBV infection consisting of periportal fibrosis, hepatocyte ballooning, cholestasis, a relatively scant inflammatory infiltrate, and marked overexpression of HBV antigens in hepatocytes. FCH had been reported in immunosuppressed patients, including liver, kidney, and bone-marrow transplanted patients. The accumulation of massive HBV antigens in the hepatocytes caused by the preS mutation can also lead to a direct cytopathic effect and result in the hepatocytes impairment, just as the FCH, and this hypothesis has been confirmed by Bock CT et al in the human hepatoma cell line. It’s worth to mention that treated the patients undergoing liver transplantation with NAs can not only inhibit HBV replication, but also prevent FCH development effectively (68,69).

The HBV virions with preS mutations infected the human liver and induce viral proteins and replicative intermediates retention in the hepatocytes. In this condition, the massive intracellular viral proteins may be cytotoxic and result in liver injury, which is considered to be the mechanism for preS mutant infection can induce worsen outcome of the CHB. Actually, the association between preS mutant infection and liver cirrhosis has been found in a series of studies, which has also been confirmed by some prospective researches. In addition, more and more evidence showed that the “complex HBV variants”, which contain mutations both in preS and BCP area, are more danger than the variants with mutations only in one area. The “complex HBV variants” have more potential to improve CHB progression toward worse outcomes, including liver failure and cirrhosis (4,70,71) (Table 1).


Conclusions and perspective

HBV sequences from the OBI patients demonstrate numerous mutations that lead to immune escape, down-regulating the expression of HBsAg, or impaired HBV packaging. In addition, these mutations may be associated with changes in the core or polymerase gene, and may play an important role for the diseases progression and outcomes. The variants with mutations in the ‘‘a’’ determinant can produce modified surface antigen, whose antigenicity has decreased and can’t be recognized and targeted by the anti-HBs neutralizing antibody induced by the vaccine immunity. Its consequence is the occurring of immune-escape. It’s difficult to detect these variants by the commercially available immunoassays which target the HBsAg. And this is a stronger risk factor for the prevalence of OBI. It’s necessary to screen the preS/S mutation in the CHB patients. Identifying the patients infected with these specific HBV variants can help doctors to evaluate their diseases and give them appropriate treatment, and also it may be useful for excluding the OBI patients from the blood donors and preventing these specific HBV variants transmissions.


Acknowledgments

Funding: This work was supported by grants from National Major Science and Technology Project for Infectious Diseases of China (2008ZX10002-011, 2012ZX10004503, 2013ZX10002001-001-006, and 2013ZX10002002), the National Natural Science Foundation of China (30170889, 30271170, 30400412, 30571646, 30700701, 81001313, 81101248, 81371828, 81461130019, and 81501748), the International Science & Technology Cooperation Program of China (2011DFA31030) and Deutsche Forschungsgemeinschaft (Transregio TRR60).


Footnote

Conflicts of Interest: All authors have completed the ICMJE uniform disclosure form (available at http://dx.doi.org/10.21037/aob.2017.10.01). The authors have no conflicts of interest to declare.

Ethical Statement: The authors are accountable for all aspects of the work in ensuring that questions related to the accuracy or integrity of any part of the work are appropriately investigated and resolved.

Open Access Statement: This is an Open Access article distributed in accordance with the Creative Commons Attribution-NonCommercial-NoDerivs 4.0 International License (CC BY-NC-ND 4.0), which permits the non-commercial replication and distribution of the article with the strict proviso that no changes or edits are made and the original work is properly cited (including links to both the formal publication through the relevant DOI and the license). See: https://creativecommons.org/licenses/by-nc-nd/4.0/.


References

  1. Trepo C, Chan HL, Lok A. Hepatitis B virus infection. Lancet 2014;384:2053-63. [Crossref] [PubMed]
  2. European Association for the Study of the Liver. EASL 2017 Clinical Practice Guidelines on the management of hepatitis B virus infection. J Hepatol 2017;67:370-98. [Crossref] [PubMed]
  3. Martinot-Peignoux M, Lapalus M, Asselah T, et al. HBsAg quantification: useful for monitoring natural history and treatment outcome. Liver Int 2014;34:97-107. [Crossref] [PubMed]
  4. Pollicino T, Cacciola I, Saffioti F, et al. Hepatitis B virus PreS/S gene variants: pathobiology and clinical implications. J Hepatol 2014;61:408-17. [Crossref] [PubMed]
  5. Seeger C, Mason WS. Hepatitis B virus biology. Microbiol Mol Biol Rev 2000;64:51-68. [Crossref] [PubMed]
  6. Dandri M, Locarnini S. New insight in the pathobiology of hepatitis B virus infection. Gut 2012;61:i6-17. [Crossref] [PubMed]
  7. Glebe D, Bremer CM. The molecular virology of hepatitis B virus. Semin Liver Dis 2013;33:103-12. [Crossref] [PubMed]
  8. Yu DM, Li XH, Mom V, et al. N-glycosylation mutations within hepatitis B virus surface major hydrophilic region contribute mostly to immune escape. J Hepatol 2014;60:515-22. [Crossref] [PubMed]
  9. Alavian SM, Carman WF, Jazayeri SM. HBsAg variants: diagnostic-escape and diagnostic dilemma. J Clin Virol 2013;57:201-8. [Crossref] [PubMed]
  10. Peng J, Cheng L, Yin B, et al. Development of an economic and efficient strategy to detect HBsAg: application of "gray-zones" in ELISA and combined use of several detection assays. Clin Chim Acta 2011;412:2046-51. [Crossref] [PubMed]
  11. Kay A, Zoulim F. Hepatitis B virus genetic variability and evolution. Virus Res 2007;127:164-76. [Crossref] [PubMed]
  12. Brunetto MR. Chance and necessity of simultaneous HBsAg and anti-HBs detection in the serum of chronic HBsAg carriers. J Hepatol 2014;60:473-5. [Crossref] [PubMed]
  13. Lazarevic I. Clinical implications of hepatitis B virus mutations: recent advances. World J Gastroenterol 2014;20:7653-64. [Crossref] [PubMed]
  14. Locarnini S. Molecular virology of hepatitis B virus. Semin Liver Dis 2004;24:3-10. [Crossref] [PubMed]
  15. Locarnini S, Zoulim F. Molecular genetics of HBV infection. Antivir Ther 2010;15:3-14. [Crossref] [PubMed]
  16. Huang X, Lu D, Ji G, et al. Hepatitis B virus (HBV) vaccine-induced escape mutants of HBV S gene among children from Qidong area, China. Virus Res 2004;99:63-8. [Crossref] [PubMed]
  17. Ngui SL, O'Connell S, Eglin RP, et al. Low detection rate and maternal provenance of hepatitis B virus S gene mutants in cases of failed postnatal immunoprophylaxis in England and Wales. J Infect Dis 1997;176:1360-5. [Crossref] [PubMed]
  18. Oon CJ, Lim GK, Ye Z, et al. Molecular epidemiology of hepatitis B virus vaccine variants in Singapore. Vaccine 1995;13:699-702. [Crossref] [PubMed]
  19. Lee PI, Chang LY, Lee CY, et al. Detection of hepatitis B surface gene mutation in carrier children with or without immunoprophylaxis at birth. J Infect Dis 1997;176:427-30. [Crossref] [PubMed]
  20. Hino K, Katoh Y, Vardas E, et al. The effect of introduction of universal childhood hepatitis B immunization in South Africa on the prevalence of serologically negative hepatitis B virus infection and the selection of immune escape variants. Vaccine 2001;19:3912-8. [Crossref] [PubMed]
  21. Yeh CT. Development of HBV S gene mutants in chronic hepatitis B patients receiving nucleotide/nucleoside analogue therapy. Antivir Ther 2010;15:471-5. [Crossref] [PubMed]
  22. Warner N, Locarnini S. The antiviral drug selected hepatitis B virus rtA181T/sW172* mutant has a dominant negative secretion defect and alters the typical profile of viral rebound. Hepatology 2008;48:88-98. [Crossref] [PubMed]
  23. Torresi J, Earnest-Silveira L, Deliyannis G, et al. Reduced antigenicity of the hepatitis B virus HBsAg protein arising as a consequence of sequence changes in the overlapping polymerase gene that are selected by lamivudine therapy. Virology 2002;293:305-13. [Crossref] [PubMed]
  24. Torresi J, Earnest-Silveira L, Civitico G, et al. Restoration of replication phenotype of lamivudine-resistant hepatitis B virus mutants by compensatory changes in the "fingers" subdomain of the viral polymerase selected as a consequence of mutations in the overlapping S gene. Virology 2002;299:88-99. [Crossref] [PubMed]
  25. Chong-Jin O, Wei Ning C, Shiuan K, et al. Identification of hepatitis B surface antigen variants with alterations outside the "a" determinant in immunized Singapore infants. J Infect Dis 1999;179:259-63. [Crossref] [PubMed]
  26. Ireland JH, O'Donnell B, Basuni AA, et al. Reactivity of 13 in vitro expressed hepatitis B surface antigen variants in 7 commercial diagnostic assays. Hepatology 2000;31:1176-82. [Crossref] [PubMed]
  27. Oon CJ, Chen WN, Goo KS, et al. Intra-familial evidence of horizontal transmission of hepatitis B virus surface antigen mutant G145R. J Infect 2000;41:260-4. [Crossref] [PubMed]
  28. Chakravarty R, Neogi M, Roychowdhury S, et al. Presence of hepatitis B surface antigen mutant G145R DNA in the peripheral blood leukocytes of the family members of an asymptomatic carrier and evidence of its horizontal transmission. Virus Res 2002;90:133-41. [Crossref] [PubMed]
  29. Ding F, Miao XL, Li YX, et al. Mutations in the S gene and in the overlapping reverse transcriptase region in chronic hepatitis B Chinese patients with coexistence of HBsAg and anti-HBs. Braz J Infect Dis 2016;20:1-7. [Crossref] [PubMed]
  30. Carman WF. The clinical significance of surface antigen variants of hepatitis B virus. J Viral Hepat 1997;4:11-20. [Crossref] [PubMed]
  31. Hsu HY, Chang MH, Ni YH, et al. Universal infant immunization and occult hepatitis B virus infection in children and adolescents: a population-based study. Hepatology 2015;61:1183-91. [Crossref] [PubMed]
  32. Oon CJ, Tan KL, Harrison T, et al. Natural history of hepatitis B surface antigen mutants in children. Lancet 1996;348:1524. [Crossref] [PubMed]
  33. Hou J, Wang Z, Cheng J, et al. Prevalence of naturally occurring surface gene variants of hepatitis B virus in nonimmunized surface antigen-negative Chinese carriers. Hepatology 2001;34:1027-34. [Crossref] [PubMed]
  34. Salpini R, Colagrossi L, Bellocchi MC, et al. Hepatitis B surface antigen genetic elements critical for immune escape correlate with hepatitis B virus reactivation upon immunosuppression. Hepatology 2015;61:823-33. [Crossref] [PubMed]
  35. Song BC, Kim SH, Kim H, et al. Prevalence of naturally occurring surface antigen variants of hepatitis B virus in Korean patients infected chronically. J Med Virol 2005;76:194-202. [Crossref] [PubMed]
  36. Liu Y, Zhang L, Zhou JY, et al. Clinical and Virological Characteristics of Chronic Hepatitis B Patients with Coexistence of HBsAg and Anti-HBs. PLoS One 2016;11:e0146980 [Crossref] [PubMed]
  37. de Campos Albuquerque I, Sousa MT, Santos MD, et al. Mutation in the S gene a determinant of the hepatitis B virus associated with concomitant HBsAg and anti-HBs in a population in Northeastern Brazil. J Med Virol 2017;89:458-62. [Crossref] [PubMed]
  38. Svicher V, Cento V, Bernassola M, et al. Novel HBsAg markers tightly correlate with occult HBV infection and strongly affect HBsAg detection. Antiviral Res 2012;93:86-93. [Crossref] [PubMed]
  39. Biswas S, Candotti D, Allain JP. Specific amino acid substitutions in the S protein prevent its excretion in vitro and may contribute to occult hepatitis B virus infection. J Virol 2013;87:7882-92. [Crossref] [PubMed]
  40. Scheiblauer H, El-Nageh M, Diaz S, et al. Performance evaluation of 70 hepatitis B virus (HBV) surface antigen (HBsAg) assays from around the world by a geographically diverse panel with an array of HBV genotypes and HBsAg subtypes. Vox Sang 2010;98:403-14. [Crossref] [PubMed]
  41. El Chaar M, Candotti D, Crowther RA, et al. Impact of hepatitis B virus surface protein mutations on the diagnosis of occult hepatitis B virus infection. Hepatology 2010;52:1600-10. [Crossref] [PubMed]
  42. Dunford L, Carr MJ, Dean J, et al. A multicentre molecular analysis of hepatitis B and blood-borne virus coinfections in Viet Nam. PLoS One 2012;7:e39027 [Crossref] [PubMed]
  43. Golsaz Shirazi F, Amiri MM, Mohammadi H, et al. Construction and expression of hepatitis B surface antigen escape variants within the "a" determinant by site directed mutagenesis. Iran J Immunol 2013;10:127-38. [PubMed]
  44. Ren F, Tsubota A, Hirokawa T, et al. A unique amino acid substitution, T126I, in human genotype C of hepatitis B virus S gene and its possible influence on antigenic structural change. Gene 2006;383:43-51. [Crossref] [PubMed]
  45. Yong-Lin Y, Qiang F, Ming-Shun Z, et al. Hepatitis B surface antigen variants in voluntary blood donors in Nanjing, China. Virol J 2012;9:82. [Crossref] [PubMed]
  46. Ueda Y, Marusawa H, Egawa H, et al. De novo activation of HBV with escape mutations from hepatitis B surface antibody after living donor liver transplantation. Antivir Ther 2011;16:479-87. [Crossref] [PubMed]
  47. Yao QQ, Dong XL, Wang XC, et al. Hepatitis B virus surface antigen (HBsAg)-positive and HBsAg-negative hepatitis B virus infection among mother-teenager pairs 13 years after neonatal hepatitis B virus vaccination. Clin Vaccine Immunol 2013;20:269-75. [Crossref] [PubMed]
  48. Ito K, Qin Y, Guarnieri M, et al. Impairment of hepatitis B virus virion secretion by single-amino-acid substitutions in the small envelope protein and rescue by a novel glycosylation site. J Virol 2010;84:12850-61. [Crossref] [PubMed]
  49. Huang CH, Yuan Q, Chen PJ, et al. Influence of mutations in hepatitis B virus surface protein on viral antigenicity and phenotype in occult HBV strains from blood donors. J Hepatol 2012;57:720-9. [Crossref] [PubMed]
  50. Mathet VL, Cuestas ML, Ruiz V, et al. Detection of hepatitis B virus (HBV) genotype E carried--even in the presence of high titers of anti-HBs antibodies--by an Argentinean patient of African descent who had received vaccination against HBV. J Clin Microbiol 2006;44:3435-9. [Crossref] [PubMed]
  51. Kim KH, Chang HY, Park JY, et al. Spontaneous HBsAg loss in Korean patients: relevance of viral genotypes, S gene mutations, and covalently closed circular DNA copy numbers. Clin Mol Hepatol 2014;20:251-60. [Crossref] [PubMed]
  52. Kim MH, Kang SY, Lee WI. Occult HBV among Anti-HBc Alone: Mutation Analysis of an HBV Surface Gene and Pre-S Gene. Yonsei Med J 2017;58:557-63. [Crossref] [PubMed]
  53. Inoue J, Ueno Y, Nagasaki F, et al. Enhanced intracellular retention of a hepatitis B virus strain associated with fulminant hepatitis. Virology 2009;395:202-9. [Crossref] [PubMed]
  54. Su H, Zhang Y, Xu D, et al. Occult hepatitis B virus infection in anti-HBs-positive infants born to HBsAg-positive mothers in China. PLoS One 2013;8:e70768 [Crossref] [PubMed]
  55. Dettori S, Candido A, Kondili LA, et al. Identification of low HBV-DNA levels by nucleic acid amplification test (NAT) in blood donors. J Infect 2009;59:128-33. [Crossref] [PubMed]
  56. Velati C, Romano L, Fomiatti L, et al. Impact of nucleic acid testing for hepatitis B virus, hepatitis C virus, and human immunodeficiency virus on the safety of blood supply in Italy: a 6-year survey. Transfusion 2008;48:2205-13. [Crossref] [PubMed]
  57. Brojer E, Grabarczyk P, Liszewski G, et al. Characterization of HBV DNA+/HBsAg- blood donors in Poland identified by triplex NAT. Hepatology 2006;44:1666-74. [Crossref] [PubMed]
  58. Katsoulidou A, Paraskevis D, Magiorkinis E, et al. Molecular characterization of occult hepatitis B cases in Greek blood donors. J Med Virol 2009;81:815-25. [Crossref] [PubMed]
  59. Yuan Q, Ou SH, Chen CR, et al. Molecular characteristics of occult hepatitis B virus from blood donors in southeast China. J Clin Microbiol 2010;48:357-62. [Crossref] [PubMed]
  60. Huang X, Qin Y, Li W, et al. Molecular analysis of the hepatitis B virus presurface and surface gene in patients from eastern China with occult hepatitis B. J Med Virol 2013;85:979-86. [Crossref] [PubMed]
  61. Cassini R, De Mitri MS, Gibellini D, et al. A novel stop codon mutation within the hepatitis B surface gene is detected in the liver but not in the peripheral blood mononuclear cells of HIV-infected individuals with occult HBV infection. J Viral Hepat 2013;20:42-9. [Crossref] [PubMed]
  62. Chen SJ, Zhao YX, Fang Y, et al. Viral deletions among healthy young Chinese adults with occult hepatitis B virus infection. Virus Res 2012;163:197-201. [Crossref] [PubMed]
  63. Fan YF, Lu CC, Chen WC, et al. 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;33:277-86. [Crossref] [PubMed]
  64. Chen BF, Liu CJ, Jow GM, et al. High prevalence and mapping of pre-S deletion in hepatitis B virus carriers with progressive liver diseases. Gastroenterology 2006;130:1153-68. [Crossref] [PubMed]
  65. Pollicino T, Zanetti AR, Cacciola I, et al. Pre-S2 defective hepatitis B virus infection in patients with fulminant hepatitis. Hepatology 1997;26:495-9. [Crossref] [PubMed]
  66. Sterneck M, Gunther S, Gerlach J, et al. Hepatitis B virus sequence changes evolving in liver transplant recipients with fulminant hepatitis. J Hepatol 1997;26:754-64. [Crossref] [PubMed]
  67. Kalinina T, Riu A, Fischer L, et al. A dominant hepatitis B virus population defective in virus secretion because of several S-gene mutations from a patient with fulminant hepatitis. Hepatology 2001;34:385-94. [Crossref] [PubMed]
  68. Bock CT, Tillmann HL, Maschek HJ, et al. A preS mutation isolated from a patient with chronic hepatitis B infection leads to virus retention and misassembly. Gastroenterology 1997;113:1976-82. [Crossref] [PubMed]
  69. Zanati SA, Locarnini SA, Dowling JP, et al. Hepatic failure due to fibrosing cholestatic hepatitis in a patient with pre-surface mutant hepatitis B virus and mixed connective tissue disease treated with prednisolone and chloroquine. J Clin Virol 2004;31:53-7. [Crossref] [PubMed]
  70. Bruss V. Revisiting the cytopathic effect of hepatitis B virus infection. Hepatology 2002;36:1327-9. [Crossref] [PubMed]
  71. Sugauchi F, Ohno T, Orito E, et al. Influence of hepatitis B virus genotypes on the development of preS deletions and advanced liver disease. J Med Virol 2003;70:537-44. [Crossref] [PubMed]
doi: 10.21037/aob.2017.10.01
Cite this article as: Wang J, Zhu B, Lu M, Yang D. Hepatitis B virus preS/S gene mutations and their clinical implications. Ann Blood 2017;2:17.

Download Citation