+ All Categories
Home > Documents > The life cycle of hepatitis B virus and antiviral targets

The life cycle of hepatitis B virus and antiviral targets

Date post: 19-Dec-2016
Category:
Upload: fabien
View: 254 times
Download: 39 times
Share this document with a friend
16
599 10.2217/FVL.11.29 © 2011 Future Medicine Ltd ISSN 1746-0794 part of Future Virology Future Virol. (2011) 6(5), 599–614 Hepatitis B virus (HBV) is one of the most dangerous human pathogens. Indeed, accord- ing to the WHO, approximately 2 billion people worldwide have been infected and more than 350 million are chronically infected. Chronic HBV infection predisposes to severe liver dis- ease, including liver cirrhosis and hepatocellular carcinoma. Approximately 600,000 people die each year due to the acute or chronic conse- quences of hepatitis B (WHO estimations in 2008) [201] . Our knowledge of the molecular biology of HBV has increased considerably over the past decades, leading to the development of very effective prophylactic vaccines and to the development of therapeutic approaches against HBV. Among these, two are currently approved to treat chronically HBV-infected patients. Pegylated IFN- a is used as an antiviral as well as to enhance the host’s immune defence sys- tem. However, only 30% of pegylated-IFN- a- treated patients achieve a sustained antiviral response [1] . Alternatively, oral administration of nucleoside/nucleotide analogs (NUCs), which specifically inhibit viral polymerase (VP) activity and thus suppress HBV repli- cation, significantly improves liver histology and the clinical outcomes of the disease after 1 year of treatment [2] . Unfortunately, NUCs act at a late stage of the HBV life cycle and do not prevent the formation and activity of the HBV transcription template, the so-called cir- cular covalently closed DNA (cccDNA). Since cccDNA has a long half-life, long-term treat- ments with NUCs are necessary to cure HBV- infected cells, but also lead to the inevitable selection of HBV drug-resistant strains [3] . New therapeutic approaches that target other viral proteins besides VP are required to decrease viral drug resistance and improve treatments against HBV. HBV structure & proteins Hepatitis B virus is a small, enveloped DNA virus that replicates via an RNA intermedi- ate and belongs to the Hepadnaviridae fam- ily. HBV particles, commonly termed Dane particles, are spherical lipid-containing struc- tures with a diameter of approximately 42 nm (FIGURE 1) . The inner shell of the virus consists of an icosahedral nucleocapsid, which is assem- bled from 120 dimers of the core protein. The nucleocapsid is covered with a membrane made up of three forms of the viral envelope protein: large (L), middle (M) and small (S), which are acquired together with the host’s lipids dur- ing budding into the endoplasmic reticulum (ER). The three surface proteins are commonly defined as hepatitis B surface antigens. They are translated from their own start codons but share the same C-terminal amino acids, called the S domain. As a consequence, the The life cycle of hepatitis B virus and antiviral targets Julie Lucifora 1 & Fabien Zoulim †2,3,4,5 1 Institute of Virology, Technische Universität München/Helmholtz Zentrum München, Trogerstrasse, 30, 81675 Munich, Germany 2 INSERM, U1052, 151 Cours Albert Thomas, 69003 Lyon, France 3 Université de Lyon, 69003 Lyon, France 4 Hepatology Department, Hospices Civils de Lyon, 69004 Lyon, France 5 Institut Universitaire de France, 103, bd Saint-Michel, 75005 Paris, France Author for correspondence: [email protected] Hepatitis B virus (HBV) remains a major public health issue with more than 350 million people chronically infected worldwide. Therapies using IFN- a or nucleos(t)ide analogs, which are currently approved for the treatment of chronic HBV infection, have failed to completely eliminate the virus. By describing the HBV life cycle, this article focuses on potential HBV targets and recent advances in the development of alternative antiviral strategies to fight chronic HBV infection. The targets that are currently under investigation to develop inhibitors are viral entry, circular covalently closed DNA formation and its regulation, nucleocapsid assembly and viral morphogenesis, as well as the innate response of infected cells against HBV infection. Other steps in the viral life cycle are potential targets; however, the development of novel antivirals remains challenging. The development of new HBV inhibitors represents a major advance in the field of antiviral therapy, to prevent antiviral drug resistance and to achieve long-term control of viral replication. Keywords n antiviral targets n hepatitis B virus n inhibitory drugs n life cycle Review For reprint orders, please contact: [email protected]
Transcript
Page 1: The life cycle of hepatitis B virus and antiviral targets

59910.2217/FVL.11.29 © 2011 Future Medicine Ltd ISSN 1746-0794

part of

Futu

re V

irolo

gy

Future Virol. (2011) 6(5), 599–614

Hepatitis B virus (HBV) is one of the most dangerous human pathogens. Indeed, accord-ing to the WHO, approximately 2 billion people worldwide have been infected and more than 350 million are chronically infected. Chronic HBV infection predisposes to severe liver dis-ease, including liver cirrhosis and hepatocellular carcinoma. Approximately 600,000 people die each year due to the acute or chronic conse-quences of hepatitis B (WHO estimations in 2008) [201].

Our knowledge of the molecular biology of HBV has increased considerably over the past decades, leading to the development of very effective prophylactic vaccines and to the development of therapeutic approaches against HBV. Among these, two are currently approved to treat chronically HBV-infected patients. Pegylated IFN-a is used as an antiviral as well as to enhance the host’s immune defence sys-tem. However, only 30% of pegylated-IFN-a-treated patients achieve a sustained antiviral response [1]. Alternatively, oral administration of nucleoside/nucleotide analogs (NUCs), which specif ically inhibit viral polymerase (VP) activity and thus suppress HBV repli-cation, significantly improves liver histology and the clinical outcomes of the disease after 1 year of treatment [2]. Unfortunately, NUCs act at a late stage of the HBV life cycle and do not prevent the formation and activity of the

HBV transcription template, the so-called cir-cular covalently closed DNA (cccDNA). Since cccDNA has a long half-life, long-term treat-ments with NUCs are necessary to cure HBV-infected cells, but also lead to the inevitable selection of HBV drug-resistant strains [3]. New therapeutic approaches that target other viral proteins besides VP are required to decrease viral drug resistance and improve treatments against HBV.

HBV structure & proteins Hepatitis B virus is a small, enveloped DNA virus that replicates via an RNA intermedi-ate and belongs to the Hepadnaviridae fam-ily. HBV particles, commonly termed Dane particles, are spherical lipid-containing struc-tures with a diameter of approximately 42 nm (Figure 1). The inner shell of the virus consists of an icosahedral nucleocapsid, which is assem-bled from 120 dimers of the core protein. The nucleocapsid is covered with a membrane made up of three forms of the viral envelope protein: large (L), middle (M) and small (S), which are acquired together with the host’s lipids dur-ing budding into the endoplasmic reticulum (ER). The three surface proteins are commonly defined as hepatitis B surface antigens. They are translated from their own start codons but share the same C-terminal amino acids, called the S domain. As a consequence, the

The life cycle of hepatitis B virus and antiviral targets

Julie Lucifora1 & Fabien Zoulim†2,3,4,5

1Institute of Virology, Technische Universität München/Helmholtz Zentrum München, Trogerstrasse, 30, 81675 Munich, Germany 2INSERM, U1052, 151 Cours Albert Thomas, 69003 Lyon, France3Université de Lyon, 69003 Lyon, France4Hepatology Department, Hospices Civils de Lyon, 69004 Lyon, France5Institut Universitaire de France, 103, bd Saint-Michel, 75005 Paris, France†Author for correspondence: [email protected]

Hepatitis B virus (HBV) remains a major public health issue with more than 350 million people chronically infected worldwide. Therapies using IFN-a or nucleos(t)ide analogs, which are currently approved for the treatment of chronic HBV infection, have failed to completely eliminate the virus. By describing the HBV life cycle, this article focuses on potential HBV targets and recent advances in the development of alternative antiviral strategies to fight chronic HBV infection. The targets that are currently under investigation to develop inhibitors are viral entry, circular covalently closed DNA formation and its regulation, nucleocapsid assembly and viral morphogenesis, as well as the innate response of infected cells against HBV infection. Other steps in the viral life cycle are potential targets; however, the development of novel antivirals remains challenging. The development of new HBV inhibitors represents a major advance in the field of antiviral therapy, to prevent antiviral drug resistance and to achieve long-term control of viral replication.

Keywords

n antiviral targets n hepatitis B virus n inhibitory drugs n life cycle

Revie

wFor reprint orders, please contact: [email protected]

Page 2: The life cycle of hepatitis B virus and antiviral targets

Future Virol. (2011) 6(5)600 future science group

Review Lucifora & Zoulim

M protein contains an extra domain, terned the preS2 domain, compared with the S pro-tein, and the L protein contains two extra domains compared with the S protein: the preS2 and preS1 domains. Nucleocapsids con-tain a single copy of the HBV genome con-sisting of a 3.2-kb partially double-stranded relaxed circular (rc) DNA molecule. This rcDNA is covalently linked to the VP at the 5´ end of the complete strand, also called viral (-) strand DNA. Besides the Dane particles, HBV infection also leads to the secretion of subviral particles, which consist of empty viral envelopes with filamentous or spheri-cal shapes (Figure 1). Subviral particles are the most abundant HBV structures released into the blood, and are thought to facilitate virus spread and persistence in the host by adsorbing virus-neutralizing antibodies.

In addition to polymerase and the structural proteins, the HBV genome also encodes for two nonstructural proteins, which, currently, have less-well defined functions. Secreted HBeAg may have immunoregulatory functions [4–7], whereas the X protein (HBx) seems to have multiple functions. HBx interacts with vari-ous cellular partners and modifies several cel-lular processes, including transcription, cell-cycle progression, DNA-damage repair and apoptosis [8,9]. Using in vitro HBV infection

models, we recently demonstrated that HBx is essential to initiating and maintaining HBV transcription [10].

Overview of the HBV life cycle Hepatitis B virus is thought to be internalized into cells through receptor-mediated endo-cytosis [11]. Proteolytic cleavage of the surface protein occurs within the endosomal compart-ment, resulting in a conformational change that exposes some translocation motifs at the surface of the viral particle. The high density of translocation motifs allows endosomal escape of the nucleocapsid into the cytosol [12]. The naked nucleocapsid is then directed towards the nucleus, and the HBV genome is translocated to the nucleus [13]. At this stage, the rcDNA genome is converted into a cccDNA, the template for viral transcription. The 3.5-kb pregenomic RNA (pgRNA) serves as mRNA for the synthesis of polymerase and core proteins. Additionally, it is used as a template for reverse transcription. The 2.4/2.1-kb subgenomic RNAs encode for three viral envelope proteins. The pgRNA is reverse transcribed within the nucleocapsid in the cyto-plasm into new rcDNA. Mature nucleocapsids are then either directed to the secretory pathway for envelopment or are redirected towards the nucleus to establish a cccDNA pool [14]. An over-view of the HBV life cycle is depicted in Figure 2.

Dane particle

rcDNA

Core

Pol

M S L

FilamentSphere

HBeAg

HBsAg

HBV proteins

S Small surface protein

M Middle surface protein

L Large surface protein

Core Capsid protein

HBeAg Secreted e antigen

Pol Polymerase

HBx X protein (non-secreted)

Figure 1. HBV proteins and hepatitis B virus virion structure. (A) List of all HBV proteins. (B) Viral particles present in the serum of HBV-infected individuals. Dane particles consist of full infectious viral particles that contain the viral genome. Spheres and filaments consist of an empty viral envelope. Together, Dane particles, spheres and filaments are recognized as HBsAg. The precore protein is secreted as HBeAg. HBsAg: Hepatitis B surface antigen; HBV: Hepatitis B virus; rcDNA: Relaxed circular DNA.

Page 3: The life cycle of hepatitis B virus and antiviral targets

www.futuremedicine.com 601future science group

The life cycle of hepatitis B virus & antiviral targets Review

D

F

G

J

Polymerase

pgRNA

Core

HBeAg

HBsAg

Hepatocyte

Surface proteins

Entry

Endosome

Trafficking

rcDNA

cccDNA

Integration into the host DNA

I

Transcription of viral RNA

Trafficking

H Morphogenesisand secretion

DNA synthesis

Nucleocapsid formation and pgRNA packaging

Nucleus

PreC/pgRNAPre-S1Pre-S2

X

AAAAAA

AAAAAA

cccDNA formation

HBx

Translation

I

Figure 2. Overview of the key steps of the hepatitis B virus life cycle. (A) Once internalized in hepatocytes by endocytosis, (B) the naked HBV nucleocapsid is directed towards the nucleus and the HBV genome is translocated into the nucleus. (C) The rcDNA genome is then converted into cccDNA, (D) the template for viral transcription. (E) After HBV-RNA transcription and subsequent HBV-protein synthesis, (F) the pgRNA is encapsidated and (G) reverse transcribed to result in a new rcDNA molecule. (H) Mature nucleocapsids are then either directed to the secretory pathway for envelopment and new virions are released, (I) or they are redirected towards the nucleus to amplify the cccDNA pool. (J) As an optional step, HBV can be integrated into the cellular host genome.cccDNA: Circular covalently closed DNA; HBeAg: Hepatitis B e antigen; HBsAg: Hepatitis B surface antigen; HBx: Hepatitis B virus X protein; pgRNA: Pregenomic RNA; rcDNA: Relaxed circular DNA.

Page 4: The life cycle of hepatitis B virus and antiviral targets

Future Virol. (2011) 6(5)602 future science group

Review Lucifora & Zoulim

HBV entryHepatitis B virus infection is restricted to hepato cytes, but the early steps of the HBV life cycle, including virus attachment to the cell surface, are poorly understood. However, recent developments from cell culture (HepaRG cells [15], primary tupaia hepatocytes [16]) and animal (severe combined immuno-deficiency–uPA mice, [17]) models, to study HBV infection, have significantly improved our understanding of mechanisms leading to HBV entry into hepatocytes.

The S domain of the surface protein crosses the lipid bilayer several times. The pre-S domain is located in the cytoplasmic side during the initial steps of assembly, and is located both outside and inside the virus after budding of the viral particle [18]. The L protein plays a major role in the HBV infection process. Indeed, the first clues to its involvement came from stud-ies that used synthetic peptides spanning the pre-S1 domain and from antibodies against this domain that strongly inhibited HBV binding to HepG2 cells [19,20]. Later mapping experiments showed that a domain that includes amino acids 2–48 of the L protein mediates attachment of the virus to hepatocytes, whereas the S pro-tein seems to be involved in other steps [21]. Moreover, a myristic acid residue, covalently attached to glycine 2 of the L protein dur-ing translation, appears to determine HBV infectivity [22,23].

In vitro studies conducted either in HepaRG cells or in primary tupaia hepatocytes have shown that peptides, consisting of authen-tically myristoylated amino acids of the N-terminal part of the L protein, specifically inhibit HBV infection [15,21,24]. These peptides offer new perspectives in the development of strategies to block an essential step in the HBV life cycle. The prototype of these peptides (HBVpreS2/2–48myr) inhibits HBV infection, with an IC

50 of approximately 8 nM [24]. Their

ability to prevent HBV infection in vivo has been shown using a severe combined immuno-deficiency–uPA murine model [25]. Indeed, accumulation of peptides in the liver and spe-cific inhibitory potency after subcutaneous delivery at low doses have both been demon-strated [25]. These peptides are now entering the first phases of clinical evaluation for the treatment of chronic hepatitis B.

Immunoadhesins that contain the pre-S1 domain have also been noted for their inhibitory effect on HBV entry into primary human hepa-tocytes [26]. They do not seem to be as efficient

as myristoylated peptides in vitro, but might offer the advantage of having higher stability in vivo [26].

Hepatitis B virus entry into hepatocytes is now thought to be a multistep process. Dane particles are the first to be trapped at the surface of the cell by heparan sulfate proteo-glycans [27], and can then bind to a high-affinity receptor that confers uptake of the virus into the cells [28]. The identification of the high-affinity receptor(s) has remained one of the most puzzling questions in the field of HBV research. Several potential HBV-binding factors (e.g., immunoglobulin A receptor, asialoglyco-protein receptor, apoH and lipoprotein lipase) have been described over the past few years, but none have been convincingly shown to be essen-tial in HBV entry [29–42]. The development of molecules that can compete with HBV for bind-ing to heparan sulfate, or to the yet unknown, high-affinity receptor(s), would lead to very promising antiviral strategies.

HBV capsid transport towards the nucleus & genome release

Hepatitis B virus uses the cellular machinery located within the nucleus for transcription of its RNA. As a consequence, the HBV genome within the capsid has to be transported across the cytoplasm to reach and enter the nucleus.

As in vitro infection models are rather in efficient, most likely because of a defect(s) in the entry process, lipofection assays have been performed to study the behavior of nucleo capsids after their release from the endosomal compart-ment into the cytoplasm. Capsid lipofection into HuH7 cells, consisting of the replacement of the HBV envelope by a lipid shell, has led to highly productive HBV replication and has demon-strated capsid accumulation at the nuclear envelope within 15 min [13]. Further experi-ments using nocodazole – a depolymerizing microtubule drug – together with binding and co-immunoprecipitation assays, have revealed active microtubule-dependent capsid transfer towards the nucleus [13]. Upon phosphorylation within the cytoplasm, capsids undergo struc-tural changes that lead to exposure of increasing numbers of the C-terminal part of the core pro-teins, which contain a nuclear localization sig-nal [43,44]. Exposure of the nuclear localization signal increases the probability of an interaction with the importin a/b proteins and binding to the nuclear-pore complexes [43]. Using permea-bilized cells, it has been further demonstrated that nucleocapsids are imported in an intact

Page 5: The life cycle of hepatitis B virus and antiviral targets

www.futuremedicine.com 603future science group

The life cycle of hepatitis B virus & antiviral targets Review

form through the nuclear-pore complexes into the nuclear basket where they interact with nucleoporin 153. Once inside the nuclear-pore complex, mature capsids (containing dsDNA and, thus, less stable than immature capsids containing RNA) disassemble and release HBV genomes within the karyoplasm [44,45].

Viral capsids released from the endosomal pathway and newly synthesized progeny cap-sids have the same structure and, thus, can both be directed to the nucleus via the same mecha-nisms. It was shown that immature capsids (from newly synthesized progeny capsids) are also transported through the nuclear-pore com-plexes, but remain arrested within the nuclear basket and do not show any release of the imma-ture genome [44,46], thus preventing disruption of the viral life cycle. Immature capsids may complete genome maturation while they are arrested in the nuclear basket and then lead to HBV genome release within the karyoplasm to increase the number of cccDNA molecules.

So far, no inhibitor of HBV nucleocapsid transport toward the nucleus has been described, and efforts should be made in this direction as it could prevent the establishment of HBV rep-lication in de novo infected cells, as well as the amplification of the cccDNA pool in cells that are already infected with HBV.

cccDNA formation As shown in Figure 3, conversion of rcDNA into cccDNA requires several steps: removal of the VP linked to the 5́ end of the (-) strand DNA as well as one of the redundant sequences (either in the 3´ or in 5́ end); removal of the RNA primer linked to the 5´ end of the (+) strand; comple-tion of the viral (+) strand DNA; and ligations of DNA extremities for both (+) and (-) strands.

Using transient transfection of the enveloped deficient HBV genome or stable cell lines sup-porting replication of the enveloped deficient HBV genome in an inducible manner, it was demonstrated that surface proteins negatively regulate HBV cccDNA formation before the removal of the VP linked to the 5´ end of the rcDNA (-) strand [47,48]. Furthermore, this rcDNA deproteinization appears to occur within the capsid before translocation to the nucleus, by cleaving the phosphodiester bound between the tyrosine of the polymerase and the 5´ phosphoryl group of the (-) strand DNA [48]. It was also observed that deproteinized- rcDNA contains only full-length (+) strand DNA, and completion of this strand has to occur earlier [47,48]. These results and the recent finding that in vitro deproteinization is inhibited by viral DNA polymerase inhibi-tors [49] suggest that completion of (+) strand

-

+3´

3´3´

(+) strand

(-) strand(-) strand

CGG

Relaxed circular DNA cccDNA

Removal of the polymerase and one of the two redundant sequences

Removal of the RNA primer

Completion of the viral (+) DNA strand

Ligations of DNA extremities

Association with cellular proteins

HistonePolymerase Redundant sequence RNA primer

Figure 3. Representation of relaxed circular DNA conversion to circular covalently closed DNA. (A) Removal of viral polymerase linked to the 5’ end of the (-) DNA strand and of one of the redundant sequences, (B) removal of the RNA primer linked to the 5’ end of the (+) strand, (C) completion of the viral (+) DNA strand and (D) ligations of DNA extremities for both (+) and (-) strands. (E) Once formed, cccDNA associates with proteins, such as histones, resulting in being supercoiled and forming the so-called cccDNA minichromosome. cccDNA: Circular covalently closed DNA.

Page 6: The life cycle of hepatitis B virus and antiviral targets

Future Virol. (2011) 6(5)604 future science group

Review Lucifora & Zoulim

DNA and removal of polymerase linked to the (-) strand DNA are probably catalyzed by the VP itself and/or by cellular proteins packaged into the nucleocapsid. Two different groups have identified cellular tyrosyl phosphodieste-rase-2 as a molecule able to specifically cleave VP from rcDNA: its exact role in cccDNA formation is currently under investigation [50,51]. Moreover, consistent with the aforemen-tioned mechanisms for DNA entry into the nucleus, it has been shown that deproteiniza-tion of rcDNA is tightly linked with nucleo-capsid disassembly and HBV DNA transloca-tion into the karyoplasm [49]. A recent study that precisely analyzed rcDNA and cccDNA sequences showed that, once in the nucleus, the redundant sequence at the 5´ end of the (-) strand is removed and that both DNA strands then undergo DNA-repair reactions, which are probably carried out by cellular DNA-repair machinery [52]. The hypothesis that the initial cccDNA formation may at least partially depend on cellular enzymes is consistent with data showing that the initial formation of cccDNA from incoming virions cannot be prevented using potent NUCs that inhibit VP or (+) strand DNA synthesis [53–55]. However, a recent study reported that genera-tion of cccDNA via intracellular recycling is also probably regulated in a virus-specific man-ner. Indeed, it was shown that, whereas HBV and duck HBV (DHBV) cccDNA formation were not cell-type dependent, DHBV cccDNA conversion was much more efficient than that of HBV [56].

The recycling of the neo-formed nucleo-capsids, together with the long half-life of the cccDNA molecule, ensures HBV persistence in cells. Thus, the discovery of compounds that specifically inhibit cccDNA formation remains one of the major challenges to cur-ing chronic HBV infections. Recent data on cccDNA stability in liver regeneration suggest that therapeutic strategies that aim to induce a certain amount of cell injury and encourage compensatory hepatocyte regeneration may be necessary to significantly reduce cccDNA loads in HBV chronically infected patients and for the possible long-term immunological control of HBV infection [57]. Furthermore, the observation that efficient DHBV cccDNA for-mation is taking place in human hepatocytes [56] could facilitate experimental identification of the human cell ular factors involved in the cccDNA process, which may then suggest a potential target for drug development.

Control of HBV RNA transcription: HBV transcripts

Once formed, the cccDNA serves as a tran-scriptional template for viral RNA synthesis by cellular RNA polymerase II (the enzyme also responsible for cellular mRNA synthesis).

Histones and nonhistone proteins are bound to cccDNA, resulting in a chromatin-like structure named viral minichromosome [58,59]. Epigenetic modification of the cccDNA mol-ecule, which leads to the control of HBV RNA transcription, has been recently reported. For instance, it was shown that HBV transcrip-tion is regulated by the acetylation status of the cccDNA-bound H3/H4 histones [60]. Recent studies have highlighted HBx as a key regulator of transcription from cccDNA, either by favor-ing histone acetylation or by preventing histone deacetylation [10,61]. As HBx plays a central role in HBV infection [10], it would be a very interest-ing target for new therapies that prevent HBV viral replication. By contrast, methylation of the cccDNA CpG island was shown to inhibit HBV transcription in vitro [62], and the ratios of rcDNA to cccDNA molecules in HBeAg-positive individuals have revealed that cccDNA methylation correlates with impaired virion pro-ductivity [63]. Theoretically, finding drugs that could specifically increase cccDNA methylation would also help to inhibit HBV transcription.

Efficient transcription of HBV genes also requires a number of ubiquitous and hepato-cyte-specific transcription factors (for a review see [64]). Several studies have described com-pounds that can inhibit HBV transcription by targeting transcription factors. For instance, it has been shown that cytokine IL-6 controls expression of HNF1a and HNF4a (the two major transcription factors that allow HBV transcription [65]) by activating JNK and ERK kinases [66]. The helioxanthin analog 8–1 and the plant Phyllanthus amarus can also sup-press HBV RNA transcription by, respectively, decreasing levels of HNF4a and HNF3b [67] and repressing the HBV enhancer I [68]. Zinc-finger proteins have also shown activity against HBV, likely owing to competition for DNA binding sites with transcription factors, thus interfering with read-through transcription by RNA polymerase across the zinc-finger protein-binding region [69].

Other compounds have been described in the past few months that can inhibit HBV tran-scription such as Curcuma longa extract [70], lutein [71], cinobufacini [72] and ethanol extracts of Hypericum perforatum [73]. Although these

Page 7: The life cycle of hepatitis B virus and antiviral targets

www.futuremedicine.com 605future science group

The life cycle of hepatitis B virus & antiviral targets Review

data are preliminary and the exact mechanisms of their actions remain to be determined, these compounds could represent novel candidates to be further evaluated as therapies against HBV.

Several other antiviral approaches, such as RNA interference, have been evaluated to target the viral transcripts themselves [74,75]. However, some challenges, including poor siRNA stabil-ity, inefficient cellular uptake, widespread bio-distribution and nonspecific effects, need to be overcome. Different strategies to improve the therapeutic efficacy of siRNAs, while avoiding their off-target effects, are under investigation (for a review, see [76,77] and more recently [78–82]). Antisense oligo nucleotides [83,84] and ribozymes [85–89] have also been described for their efficient targeting of HBV RNA. However, their use in therapeutic strategies against HBV also requires that their delivery to infected cells is improved. Finally, IFN-a, via MxA activity, was shown to inhibit the nucleocytoplasmic export of viral mRNA [90].

Capsid formation & maturationOnce synthesized, pgRNA is encapsidated together with VP. The HBV capsid spontane-ously self-assembles from many copies of core dimers present in the cytoplasm. It has been shown that the formation of a trimeric nucleus and the subsequent elongation reactions occur by adding one dimeric subunit at a time until it is complete [91]. Three components are involved in specific packaging of pgRNA into the capsid: VP, the nucleic acid-binding domain of the core protein and the e stem–loop in the 5´ region of pgRNA [92–95]. A recent study has reported that core protein dimers can bind and encapsidate both pgRNA and heterologous RNA molecules with a high level of cooperation, irrespective of the phosphorylation status [96]. This strongly suggests that VP is probably the factor required for specific packaging of pgRNA.

Phenylpropenamide derivatives [97–99], includ-ing compounds named AT-61 and AT-130, and oxymatrine [100], a type of alkaloid extracted from the herb Sophora alopecuroides, have been shown to inhibit pgRNA packaging. A recent study suggested that phenylpropen amides are, in fact, accelerators of HBV capsid assem-bly, and their actions result in the formation of empty capsids [101]. These very interesting results illustrate the importance of the kinetic pathway in successful virus assembly.

Heteroaryldihydropyrimidines, including compounds named Bay 41–4109, Bay 38–7690 and Bay 39–5493, were discovered to prevent

the proper formation of viral core particles and to increase degradation of the core protein [102–105]. It was also shown that peptide aptam-ers, which bind to the HBV core protein, inhibit capsid formation by specific sequestration of bound proteins into aggresomes [106] and bis-ANS, a small molecule that acts as a molecular ‘wedge’ and interferes with normal capsid–protein geometry and capsid formation [107]. Stability of the capsid may also be disrupted by cytokines, such as IFN-a [108] and TNF-a [109].

Maturation of the nucleocapsid consists of synthesis of the (-) DNA strand by reverse-transcriptase activity of VP, followed by pgRNA degradation by RNase H activity of VP and synthesis of the (+) DNA strand by the DNA polymerase activity of VP. In viral DNA synthe-sis, the corresponding inhibitors, such as NUCs, and our understanding of the mechanisms of resistance to NUCs, have been extensively stud-ied over the past few years (for review, see [110]). Of note, different NUCs, such as lamivudine, adefovir, entecavir, telbivudine and tenofovir, have been approved to treat chronic HBV infec-tion, and continuing efforts are being made to maximize their therapeutic benefits and to minimize their resistance rates. For instance, after 5 years of treatment, 80–90% of patients receiving lamivudine (which was the first NUC available on the market) showed the emergence of drug-resistant mutants, whereas no resistance has been observed with tenofovir, so far [110].

All stages of viral DNA synthesis can be found within the cells, but only mature cap-sids, containing rcDNA, are secreted [111,112]. This suggests that the RNA-containing capsids cannot be incorporated into virions and that DNA synthesis is associated with morphological changes. Structural differences between capsids containing RNA or DNA have, indeed, been observed [113], as well as differences in the phos-phorylation state of the arginine-rich domain of the core protein [114]. It has also been shown that capsids from various sources encapsidated active PKC and inhibition of PKC phospho-rylation does not affect genome maturation, but results in capsid accumulation and inhibits virion release [115].

Morphogenesis & virus releaseHepatitis B virus budding has been shown to be strictly dependent on the L protein [116], and when the ratio between L proteins and nucleo-capsids is not optimal, the latter are preferentially recycled to the nucleus to amplify the cccDNA molecule pool [117]. Moreover, the interaction of

Page 8: The life cycle of hepatitis B virus and antiviral targets

Future Virol. (2011) 6(5)606 future science group

Review Lucifora & Zoulim

L protein with capsids is critical for HBV assem-bly as it can be disrupted by peptides interact-ing with the core [118]. Based on data obtained recently in different studies, it has been proposed that HBV virions can bud into late endosomes or multivesicular bodies, and exit the cell via the exosome pathway (for a review, see [119]).

Strategies that interfere with the correct fold-ing of the surface protein decrease virus secre-tion and its resulting infectivity. For example, a-glucosidase inhibitors are potent inhibitors of HBV-particle secretion [120,121] and the use of N-butyldeoxynojirimycin has been shown to induce secretion of HBV particles that con-tain an envelope with an altered composition of disulfide-linked oligomers and no detectable M protein. As a consequence, HBV infectiv-ity is reduced by 80% [122]. As an alternative strategy to targeting the surface proteins, single-domain antibodies that recognize the S protein were shown to reduce HBsAg secretion when expressed and retained in the ER as intra bodies. Moreover, in a hydrodynamics-based HBV mouse model, these intrabodies caused a ten- to >100-fold reduction in the concentration of HBV virions in plasma [123]. It was also demon-strated that treatments of HBV replicating cells with two human monoclonal anti-HBs antibod-ies (e.g., HepeX-B™; XTL Biopharmaceuticals, Israel) resulted in cellular uptake of the anti-bodies and intracellular accumulation of HBV particles, and prolonged blocking of their secretion [124].

The presence of cholesterol within the viral envelope is indispensable for HBV entry into hepatocytes. Indeed, infectivity of HBV pro-duced from cell culture in the presence of inhibitors of cholesterol synthesis is severely impaired, whereas cholesterol extraction from cellular membranes showed no effect on HBV infection, excluding a role of lipid rafts [125]. Cholesterol biosynthesis within the infected cell is thus a promising target to interfere with HBV morphogenesis and to lower infectivity of neo-formed virions. Confirming this hypothesis, it was recently shown that polyunsaturated ER liposomes, which can lower cholesterol, led to an approximately 40% decrease in HBV secre-tion and to an approximately 70% decrease in the resulting infectivity [126].

Other different compounds can inhibit HBV secretion, such as HBF-0259 (an aromatically substituted tetrahydro-tetrazolo-[1,5-a]-pyri-midine) [127] or the root extract of Boehmeria nivea [128], although their exact mechanisms of action remain unclear.

Virus–host-cell interactionsAlthough it is not essential for the HBV life cycle, the viral genome may integrate into the host genome. This is probably a result of inte-gration of viral double-stranded linear DNA by cellular enzymes such as topoisomerase I [129]. As mentioned earlier, chronic HBV infection is a major risk factor for the development of hepatocellular carcinoma, and HBV genome insertion into the host genome may contrib-ute to mechanisms leading to tumorigenesis. Integration of HBV DNA into the genome of hepatocytes was found in 85–90% of hepato-cellular carcinoma related to HBV infection, and no specific genes have been identified in humans that are preferentially targeted [130]. The insertion is thought to induce general genomic alterations, which may result in the loss of tumor-suppressor gene functions and/or activation of tumor-promoting genes. Blocking HBV DNA replication may also interfere with viral genome integration, as the generation of double-stranded linear DNA is a result of defects caused during replication. In this regard, it was shown that lamivudine treatment was associated with a decrease in hepatocellu-lar carcinoma development [131]. Early antiviral intervention is thus needed not only for chroni-cally infected patients to decrease HBV spread, but also to reduce insertional mutagenesis that can lead to HBV pathogenesis.

Long-term expression of viral proteins has also been suggested to play an important role in hepatocarcinogenesis. For example, it was shown that a truncated form of the L protein or of the M protein increases hepatocyte prolifera-tion by upregulating expression of cyclin A or activating the c-Raf-1/Erk2 signaling pathway, respectively [132]. Moreover, accumulation of surface proteins in the ER induces ER stress, which leads to oxidative stress and DNA dam-age [133], thus predisposing cells to transforma-tion. In addition to its crucial role in the control of HBV transcription, HBx has been shown to interact with various cellular partners and to modify diverse cellular processes, including transcription, cell-cycle progression, DNA-damage repair, and apoptosis (for a review, see [134]). HBx and the surface proteins are, thus, very interesting targets for the develop-ment of new therapies against chronic hepati-tis B infection by preventing viral replication. Indeed, by interfering with their functions, novel antiviral strategies could be designed to prevent viral replication as well long-term con-sequences of HBV infection.

Page 9: The life cycle of hepatitis B virus and antiviral targets

www.futuremedicine.com 607future science group

The life cycle of hepatitis B virus & antiviral targets Review

Another interesting aspect of the interaction between the virus and the host cell is the capac-ity of the virus to induce an innate response in the hepatocyte and to then counteract this effect by developing a persistent infection. For many years, HBV was viewed as a stealth virus that did not induce an innate response when it spread throughout the liver [135,136]. However, recent observations have provided evidence that suggests that HBV can be sensed by liver cells and can elicit an antiviral response [66,137]. To reconcile these data, one can suggest that HBV may induce a transient innate response but may also rapidly inhibit it [138]. Characterization of cellular and viral factors involved in HBV rec-ognition, as well as in mechanisms employed to counteract the innate response, would pro-vide new insights into the design of therapies aimed at stimulating the antiviral response of infected cells.

Other interactions of the virus with the cellular machinery are also required in the viral life cycle and may represent potential new targets for drug discovery. Of note, it has been shown that chaperones are required for hepadna virus reverse-transcriptase bind-ing to e on pre genomic RNA and to activate viral-enzyme activity (for review see [139]). The specific inhibition of this interaction may rep-resent a novel target to prevent pgRNA encap-sidation and VP activity. The virus can also use the cellular machinery of the later steps during viral replication, as detailed earlier. It has also been shown that HBV exploits the multive-sicular machinery, with the aid of g2-adaptin, to assemble an egress [140].

Of note, full understanding of the inter-actions between HBV and the immune system is crucial for the development of new anti viral strategies. Indeed, patients who resolve an acute hepatitis B infection display strong poly-clonal and multispecific helper and cytotoxic T-cell responses against HBV core, polymerase and envelope proteins [141]. As these cellular responses are weak and often dysfunctional in patients with chronic HBV infection [141], the development of therapies aimed at boosting the immune system, such as therapeutic vac-cination [142] or adoptive T-cell transfer [143], is also very important.

ConclusionMajor advances in our understanding of HBV molecular biology have been made over the past few years. As a consequence, increasing num-bers of inhibitors that are involved in the HBV

life cycle are being investigated in experimen-tal models (Table 1). After being evaluated in relevant animal models and in clinical trials, these novel inhibitory molecules may provide new options to treat chronic HBV infection. In the meantime, efforts remain to be made to develop new inhibitory molecules that par-ticularly target the steps of the HBV life cycle without known inhibitors.

As described in this article, each step of the HBV life cycle has the potential to be developed into a therapeutic approach. However, inter-vention at specific steps may provide relative advantages. For instance, the development of molecules that inhibit cccDNA-molecule for-mation and stability is one of the most chal-lenging areas in the development of strategies against HBV. Such a therapeutic approach would, indeed, lead to an abortive HBV life cycle, could abolish HBV persistence and pre-vent the HBV rebound observed after the arrest of treatment with NUCs. The development of efficient HBV-attachment and -entry inhibi-tors is also a very attractive antiviral strategy from a therapeutic viewpoint, as it is the first opportunity to curtail the virus’ life cycle. This may lead to novel therapies that potentially pre-vent vertical transmission and inhibit reinfec-tion after liver transplant, and are extremely valuable for postexposure prophylaxis. By con-trast, although targeting the latter steps of the HBV life cycle, such as transcription, capsid formation and maturation, as well as assem-bly and release, has been proven to efficiently fight HBV infection, these approaches could be considered less attractive from a theoretical point of view as they do not cure infected cells of all HBV components. However, targeting these steps is still very valuable for the treat-ment of HBV chronic infection as it allows us to keep the viral load at a low level, avoids the spread of the virus and reduces liver damage. However, some issues remain to be addressed, such as the fate of pgRNA-containing nucleo-capsids, which may accumulate in the presence of NUC treatment (owing to the blockade of viral DNA synthesis).

It is important to keep in mind that antiviral molecules can be divided into two classes: those that directly target the viral components and those that target the host cell. The first class is likely to result in inhibitors with a high spe-cificity and potency, and with low toxicity, but potentially with a high risk of resistance devel-opment. By comparison, the latter approach may yield broad-spectrum antivirals that target

Page 10: The life cycle of hepatitis B virus and antiviral targets

Future Virol. (2011) 6(5)608 future science group

Review Lucifora & Zoulim

more than one virus and cause minimal or no resistance development, but have compa-rably lower potency and higher toxicity [144]. The optimal antiviral strategy would, thus, combine drugs with different mechanisms of action targeting both HBV and the cellular components necessary for a productive HBV life cycle. Such combinations could reduce the risk of selection of HBV drug-resistant strains. Strategies that combine specific molecules that target HBV and the molecules that induce an innate immune response have been recently described [145] [Ebert G et  al., Unpublished Data] and should also be evaluated in clinical trials in order to assess their restoration and dura-tion in the specific immune response against HBV. Combined strategies may then prove use-ful to eradicate the viral genome or to induce long-term control of viral replication, thereby preventing the complications caused by the HBV disease.

Future perspectiveMajor challenges remain regarding our under-standing of the HBV life cycle. Answers to these key questions may not only improve our basic knowledge of the mechanism of viral

replication and pathogenesis, but will also help us develop innovative therapies beyond viral suppression that are induced by NUCs.

Deciphering viral entry should be achieved during the next decade, but may still be chal-lenging because of the difficulty in growing HBV in tissue culture when compared with other viruses such as HIV or HCV. Entry inhibitors, which represent a new treatment target, are currently being evaluated and may represent a new class of antivirals. Identifying the detailed molecular steps of cccDNA for-mation is also underway with major recent advances, which should pave the way for com-plete understanding of this process, as well as the epigenetic control of this DNA template for viral gene expression. This would represent a major breakthrough in studying the molecular biology of HBV as well as for drug develop-ment. Specific targeting of the formation and regulation of this viral genome species respon-sible for viral persistence may then become feasible. Obtaining a crystal structure of an enzymatically active VP represents one of the major challenges over the next decade. This could enable the development of novel inhibi-tors that could complement the classic NUCs.

Table 1. Hepatitis B virus potential targets and their main inhibitors.

Steps of the HBV life cycle

Targets Inhibitors Ref.

Entry L protein Peptides consisting of the authentically myristoylated amino acids of the N-terminal part of the L protein

[15,21,24,25]

Nucleocapsid trafficking and DNA entry into the nucleus

Not determined None

cccDNA formation Not determined NoneControl of HBV transcription transcripts

HBx NoneTranscription factors IL-6, helioxanthin analog 8–1, the plant Phyllanthus amarus

and zinc finger proteins [66–69]

Unknown Curcuma longa Linn extract, lutein and ethanol extracts of Hypericum perforatum L

[70–73]

siRNA siRNA, antisense oligonucleotides, ribozyme and IFN-a [74,75,83–90]

Capsid formation and maturation

pgRNA packaging Phenylpropenamide derivates and oxymatrine [97–100]

Capsid assembly and stability Heteroaryldihydropyrimidines, peptide aptamers that bind to the core protein, bis-ANS, TNF-a and IFN-a

[102–109]

DNA synthesis Nucleos(t)ide analogs [110]

Virus assembly and release Interaction between capsid and the L protein

Peptides interacting with core proteins [118]

Surface proteins Antibodies against surface proteins [123,124]

Folding of surface proteins a-glucosidase inhibitors [120–122]

Cholesterol insertion within the viral envelope

Polyunsaturated ER liposomes [126]

Virus secretion HBF-0259, the root extract of Boehmeria nivea [127,128]

Integration of the DNA into the host genome

Not determined None

cccDNA: Circular covalently closed DNA; ER: Endoplasmic reticulum; HBV: Hepatitis B virus; HBx: Hepatitis B X protein nonsecreted; pgRNA: Pregenomic RNA.

Page 11: The life cycle of hepatitis B virus and antiviral targets

www.futuremedicine.com 609future science group

The life cycle of hepatitis B virus & antiviral targets Review

The role of the X protein has been debated for 20 years. The availability of more effi-cient and relevant cell-culture models should help us to gain a precise understanding of its function in the replication cycle and the pathogenesis of virus-induced hepatocellular carcinoma. Greater understanding regarding the role of the innate and adaptive immune responses against HBV in controlling or eradi-cating infection is critical to developing new treatment- combination strategies based on direct antivirals and immune modulators.

Hepatitis B virus research in the next decade should result in major advances in our knowl-edge regarding the viral life cycle and the interactions between the virus and host cells. This new information will pave the way to the identification of new targets, to discoveries of

new drugs and to the development of effica-cious drug-combination strategies. These research efforts could help us reach the goal of finite-duration treatments, prevent resistance and prevent disease complications, including hepatocellular carcinoma.

Executive summary

Hepatitis B virus entry into hepatocytesnThe hepatitis B virus (HBV) L protein is the main viral factor involved in the process. Peptides consisting of authentically myristoylated

amino acids, with an N-terminal part, are potent inhibitors.nHBV particles are first trapped at the surface of the cell by heparan sulfate proteoglycans and then bind to a high-affinity receptor that

confers uptake of the virus into cells. No inhibitors have been described, so far, that can block the cellular factors involved.

Nucleocapsid trafficking & DNA entry into the nucleusnHBV nucleocapsids are transported across the cytoplasm toward the nucleus by an active microtubule-dependent mechanism. They are

then imported into the nuclear basket where they are disassembled and release HBV genomes within the karyoplasm. nNo inhibitors have been described, so far, to block these mechanisms.

Circular covalently closed DNA formationnCircular covalently closed DNA formation may depend on both viral and cellular enzymes.nNo inhibitors have been described, so far, to block this step.

Control of HBV transcription transcriptsnHBx controls epigenetic modification of the circular covalently closed DNA, which allows HBV-RNA transcription. No inhibitors of HBx

have been described so far.nEfficient transcription of HBV genes also requires a number of ubiquitous and hepatocyte-specific transcription factors. Different

molecules that inhibit these transcription factors have been described.nSeveral antiviral approaches, such as RNA interference, have been successfully evaluated for targeting viral transcripts.

Capsid formation & maturationnHBV capsids spontaneously self-assemble from many copies of core dimers present in the cytoplasm.nViral polymerase, the nucleic-acid-binding domain of the core protein, and the e stem–loop in the 5’ region of pregenomic RNA

(pgRNA) are involved in specific packaging of pgRNA into the capsid.nSeveral molecules inhibit capsid formation: pgRNA encapsidation, reverse transcription and DNA synthesis have been described.

Virus assembly & releasenInteraction of L protein with capsids is critical for HBV assembly. nStrategies that interfere with the correct folding of the surface protein decrease virus secretion and its resulting infectivity.nInhibition of cholesterol biosynthesis within the infected cell interferes with HBV morphogenesis, resulting in lower infectivity of

neo-formed virions.

Virus–host-cell interactionsnVirus genome integration into the host genome and the long-term expression of viral proteins contributes to the development of

hepatocellular carcinoma.nThese observations favor an early antiviral intervention for patients who are chronically infected with HBV.

ConclusionnThe optimal antiviral strategy would combine drugs with different mechanisms of action that inhibit the different steps within the

HBV life cycle.

Financial & competing interests disclosureJ Lucifora holds a stipend from the European Association for the Study of Liver disease (EASL): ‘Sheila Sherlock EASL Post-Doc Fellowship’. The authors have no other relevant affiliations or financial involvement with any organization or entity with a financial interest in or financial conflict with the sub-ject matter or materials discussed in the manuscript apart from those disclosed.

No writing assistance was utilized in the production of this manuscript.

Page 12: The life cycle of hepatitis B virus and antiviral targets

Future Virol. (2011) 6(5)610 future science group

Review Lucifora & Zoulim

BibliographyPapers of special note have been highlighted as:n of interestnn of considerable interest

1. Karayiannis P: Hepatitis B virus: old, new and future approaches to antiviral treatment. J. Antimicrob. Chemother. 51(4), 761–785 (2003).

2. Liaw YF, Sung JJ, Chow WC et al.: Lamivudine for patients with chronic hepatitis B and advanced liver disease. N. Engl. J. Med. 351(15), 1521–1531 (2004).

3. Zoulim F, Lucifora J: Hepatitis B virus and drug resistance: implications for treatment. Future Virol. 1, 361–376 (2006).

4. Bertoletti A, Gehring AJ: The immune response during hepatitis B virus infection. J. Gen. Virol. 87(Pt 6), 1439–1449 (2006).

5. Chen MT, Billaud JN, Sallberg M et al.: A function of the hepatitis B virus precore protein is to regulate the immune response to the core antigen. Proc. Natl Acad. Sci. USA 101(41), 14913–14918 (2004).

6. Chen M, Sallberg M, Hughes J et al.: Immune tolerance split between hepatitis B virus precore and core proteins. J. Virol. 79(5), 3016–3027 (2005).

7. Visvanathan K, Skinner NA, Thompson AJ et al.: Regulation of Toll-like receptor-2 expression in chronic hepatitis B by the precore protein. Hepatology 45(1), 102–110 (2007).

8. Murakami S: Hepatitis B virus X protein: a multifunctional viral regulator. J. Gastroenterol. 36(10), 651–660 (2001).

9. Bouchard MJ, Schneider RJ: The enigmatic X gene of hepatitis B virus. J. Virol. 78(23), 12725–12734 (2004).

10. Lucifora J, Arzberger S, Durantel D et al.: Hepatitis B virus X protein is essential to initiate and maintain virus replication after infection. J. Hepatol. DOI: 10.1016/j.jhep.2011.02.015 (2011) (In Press).

11. Kott N, König A, Glebe D: Hepatitis B virus (HBV) bypasses classical endocytic pathways to infect primary hepatocytes in vitro. J. Hepatol. 52(Suppl. 1), S48–S49 (2010).

12. Stoeckl L, Funk A, Kopitzki A et al.: Identification of a structural motif crucial for infectivity of hepatitis B viruses. Proc. Natl Acad. Sci. USA 103(17), 6730–6734 (2006).

13. Rabe B, Glebe D, Kann M: Lipid-mediated introduction of hepatitis B virus capsids into nonsusceptible cells allows highly efficient replication and facilitates the study of early infection events. J. Virol. 80(11), 5465–5473 (2006).

14. Seeger C, Mason B, Zoulim F: Hepadnaviruses. In: Fields Virology. Knipe DM, Howley PM (Eds). Lippincott Williams and Wilkins, PA, USA 2977–3029 (2006).

15. Gripon P, Rumin S, Urban S et al.: Infection of a human hepatoma cell line by hepatitis B virus. Proc. Natl Acad. Sci. USA 99(24), 15655–15660 (2002).

16. Kock J, Nassal M, Macnelly S, Baumert TF, Blum HE, Von Weizsacker F: Efficient infection of primary tupaia hepatocytes with purified human and woolly monkey hepatitis B virus. J. Virol. 75(11), 5084–5089 (2001).

17. Dandri M, Burda MR, Torok E et al.: Repopulation of mouse liver with human hepatocytes and in vivo infection with hepatitis B virus. Hepatology 33(4), 981–988 (2001).

18. Bruss V, Lu X, Thomssen R, Gerlich WH: Post-translational alterations in transmembrane topology of the hepatitis B virus large envelope protein. EMBO J. 13(10), 2273–2279 (1994).

19. Neurath AR, Kent SB, Strick N, Parker K: Identification and chemical synthesis of a host cell receptor binding site on hepatitis B virus. Cell 46(3), 429–436 (1986).

20. Petit MA, Dubanchet S, Capel F, Voet P, Dauguet C, Hauser P: HepG2 cell binding activities of different hepatitis B virus isolates: inhibitory effect of anti-HBs and anti-preS1(21–47). Virology 180(2), 483–491 (1991).

21. Glebe D, Urban S, Knoop EV et al.: Mapping of the hepatitis B virus attachment site by use of infection-inhibiting preS1 lipopeptides and tupaia hepatocytes. Gastroenterology 129(1), 234–245 (2005).

22. Gripon P, Le Seyec J, Rumin S, Guguen-Guillouzo C: Myristylation of the hepatitis B virus large surface protein is essential for viral infectivity. Virology 213(2), 292–299 (1995).

23. Bruss V, Hagelstein J, Gerhardt E, Galle PR: Myristylation of the large surface protein is required for hepatitis B virus in vitro infectivity. Virology 218(2), 396–399 (1996).

24. Gripon P, Cannie I, Urban S: Efficient inhibition of hepatitis B virus infection by acylated peptides derived from the large viral surface protein. J. Virol. 79(3), 1613–1622 (2005).

25. Petersen J, Dandri M, Mier W et al.: Prevention of hepatitis B virus infection in vivo by entry inhibitors derived from the large envelope protein. Nat. Biotechnol. 26(3), 335–341 (2008).

nn Important study showing in vivo applicability of acylated peptides derived from the large hepatitis B virus (HBV) envelope protein for HBV entry inhibition.

26. Chai N, Gudima S, Chang J, Taylor J: Immunoadhesins containing pre-S domains of hepatitis B virus large envelope protein are secreted and inhibit virus infection. J. Virol. 81(10), 4912–4918 (2007).

27. Schulze A, Gripon P, Urban S: Hepatitis B virus infection initiates with a large surface protein-dependent binding to heparan sulfate proteoglycans. Hepatology 46(6), 1759–1768 (2007).

28. Leistner CM, Gruen-Bernhard S, Glebe D: Role of glycosaminoglycans for binding and infection of hepatitis B virus. Cell Microbiol. 10(1), 122–133 (2008).

29. Neurath AR, Strick N: Antigenic mimicry of an immunoglobulin A epitope by a hepatitis B virus cell attachment site. Virology 178(2), 631–634 (1990).

30. Pontisso P, Ruvoletto MG, Tiribelli C, Gerlich WH, Ruol A, Alberti A: The preS1 domain of hepatitis B virus and IgA cross-react in their binding to the hepatocyte surface. J. Gen. Virol. 73(Pt 8), 2041–2045 (1992).

31. Dash S, Rao KV, Panda SK: Receptor for pre-S1(21–47) component of hepatitis B virus on the liver cell: role in virus cell interaction. J. Med. Virol. 37(2), 116–121 (1992).

32. Neurath AR, Strick N, Sproul P: Search for hepatitis B virus cell receptors reveals binding sites for interleukin 6 on the virus envelope protein. J. Exp. Med. 175(2), 461–469 (1992).

33. Neurath AR, Strick N, Li YY: Cells transfected with human interleukin 6 cDNA acquire binding sites for the hepatitis B virus envelope protein. J. Exp. Med. 176(6), 1561–1569 (1992).

34. Petit MA, Capel F, Dubanchet S, Mabit H: PreS1-specific binding proteins as potential receptors for hepatitis B virus in human hepatocytes. Virology 187(1), 211–222 (1992).

35. Treichel U, Meyer Zum Buschenfelde KH, Stockert RJ, Poralla T, Gerken G: The asialoglycoprotein receptor mediates hepatic binding and uptake of natural hepatitis B virus particles derived from viraemic carriers. J. Gen. Virol. 75(Pt 11), 3021–3029 (1994).

36. Mehdi H, Kaplan MJ, Anlar FY et al.: Hepatitis B virus surface antigen binds to apolipoprotein H. J. Virol. 68(4), 2415–2424 (1994).

37. Treichel U, Meyer Zum Buschenfelde KH, Dienes HP, Gerken G: Receptor-mediated entry of hepatitis B virus particles into liver cells. Arch. Virol. 142(3), 493–498 (1997).

Page 13: The life cycle of hepatitis B virus and antiviral targets

www.futuremedicine.com 611future science group

The life cycle of hepatitis B virus & antiviral targets Review

38. Duclos-Vallee JC, Capel F, Mabit H, Petit MA: Phosphorylation of the hepatitis B virus core protein by glyceraldehyde-3-phosphate dehydrogenase protein kinase activity. J. Gen. Virol. 79(Pt 7), 1665–1670 (1998).

39. Ryu CJ, Cho DY, Gripon P et al.: An 80-kilodalton protein that binds to the pre-S1 domain of hepatitis B virus. J. Virol. 74(1), 110–116 (2000).

40. Stefas I, Rucheton M, D’Angeac AD et al.: Hepatitis B virus Dane particles bind to human plasma apolipoprotein H. Hepatology 33(1), 207–217 (2001).

41. Owada T, Matsubayashi K, Sakata H et al.: Interaction between desialylated hepatitis B virus and asialoglycoprotein receptor on hepatocytes may be indispensable for viral binding and entry. J. Viral Hepat. 13(1), 11–18 (2006).

42. Deng Q, Zhai JW, Michel ML et al.: Identification and characterization of peptides that interact with hepatitis B virus via the putative receptor binding site. J. Virol. 81(8), 4244–4254 (2007).

43. Kann M, Sodeik B, Vlachou A, Gerlich WH, Helenius A: Phosphorylation-dependent binding of hepatitis B virus core particles to the nuclear pore complex. J. Cell Biol. 145(1), 45–55 (1999).

44. Rabe B, Vlachou A, Pante N, Helenius A, Kann M: Nuclear import of hepatitis B virus capsids and release of the viral genome. Proc. Natl Acad. Sci. USA 100(17), 9849–9854 (2003).

nn Major study solving mechanisms of HBV nucleocapsid import and genome release to the nucleus.

45. Rabe B, Delaleau M, Bischof A et al.: Nuclear entry of hepatitis B virus capsids involves disintegration to protein dimers followed by nuclear reassociation to capsids. PLoS Pathog. 5(8), E1000563 (2009).

46. Schmitz A, Schwarz A, Foss M et al.: Nucleoporin 153 arrests the nuclear import of hepatitis B virus capsids in the nuclear basket. PLoS Pathog. 6(1), e1000741 (2010).

47. Gao W, Hu J: Formation of hepatitis B virus covalently closed circular DNA: removal of genome-linked protein. J. Virol. 81(12), 6164–6174 (2007).

48. Guo H, Jiang D, Zhou T, Cuconati A, Block TM, Guo JT: Characterization of the intracellular deproteinized relaxed circular DNA of hepatitis B virus: an intermediate of covalently closed circular DNA formation. J. Virol. 81(22), 12472–12484 (2007).

49. Guo H, Mao R, Block TM, Guo JT: Production and function of the cytoplasmic deproteinized relaxed circular DNA of hepadnaviruses. J. Virol. 84(1), 387–396 (2010).

50. Mcallister R, Cortes-Ledesma F, Boregowda R, Caldecott K, Hu J: Characterizing the role of tyrosyl phosphodiesterase-2 in HBV cccDNA formation. Presented at: International Meeting on Molecular Biology of Hepatitis B Virus. Nankang, Taipei, Taiwan, October 9–13 2010.

51. Koeniger C, Nassal M, Beck J: Human tyrosyl-DNA-phosphodiesterase 2 can specifically remove polymerase from authentic hepadnaviral rc-DNA. Presented at: International Meeting on Molecular Biology of Hepatitis B Virus. Nankang, Taipei, Taiwan, October 9–13 2010.

52. Sohn JA, Litwin S, Seeger C: Mechanism for CCC DNA synthesis in hepadnaviruses. PLoS ONE 4(11), E8093 (2009).

53. Le Guerhier F, Pichoud C, Guerret S et al.: Characterization of the antiviral effect of 2 ,́3 -́dideoxy-2 ,́ 3 -́didehydro-b-l-5-fluorocytidine in the duck hepatitis B virus infection model. Antimicrob. Agents Chemother. 44(1), 111–122 (2000).

54. Kock J, Baumert TF, Delaney WE, Blum HE, Von Weizsacker F: Inhibitory effect of adefovir and lamivudine on the initiation of hepatitis B virus infection in primary tupaia hepatocytes. Hepatology 38(6), 1410–1418 (2003).

55. Delmas J, Schorr O, Jamard C et al.: Inhibitory effect of adefovir on viral DNA synthesis and covalently closed circular DNA formation in duck hepatitis B virus-infected hepatocytes in vivo and in vitro. Antimicrob. Agents Chemother. 46(2), 425–433 (2002).

56. Kock J, Rosler C, Zhang JJ, Blum HE, Nassal M, Thoma C: Generation of covalently closed circular DNA of hepatitis B viruses via intracellular recycling is regulated in a virus specific manner. PLoS Pathog. 6(9), E1001082 (2010).

n Study showing that efficient duck HBV circular covalently closed DNA (cccDNA) formation is taking place in human hepatocytes. In the future, this may facilitate experimental identification and possible inhibition of the human cellular factors involved in the cccDNA process.

57. Lutgehetmann M, Volz T, Kopke A et al.: In vivo proliferation of hepadnavirus-infected hepatocytes induces loss of covalently closed circular DNA in mice. Hepatology 52(1), 16–24 (2010).

58. Bock CT, Schranz P, Schroder CH, Zentgraf H: Hepatitis B virus genome is organized into nucleosomes in the nucleus of the infected cell. Virus Genes 8(3), 215–229 (1994).

59. Newbold JE, Xin H, Tencza M et al.: The covalently closed duplex form of the hepadnavirus genome exists in situ as a heterogeneous population of viral minichromosomes. J. Virol. 69(6), 3350–3357 (1995).

60. Pollicino T, Belloni L, Raffa G et al.: Hepatitis B virus replication is regulated by the acetylation status of hepatitis B virus cccDNA-bound H3 and H4 histones. Gastroenterology 130(3), 823–837 (2006).

61. Belloni L, Pollicino T, De Nicola F et al.: Nuclear HBx binds the HBV minichromosome and modifies the epigenetic regulation of cccDNA function. Proc. Natl Acad. Sci. USA 106(47), 19975–19979 (2009).

nn Important study showing for the first time that the HBV X protein is involved in epigenetic regulation of the cccDNA.

62. Vivekanandan P, Thomas D, Torbenson M: Methylation regulates hepatitis B viral protein expression. J. Infect. Dis. 199(9), 1286–1291 (2009).

63. Guo Y, Li Y, Mu S, Zhang J, Yan Z: Evidence that methylation of hepatitis B virus covalently closed circular DNA in liver tissues of patients with chronic hepatitis B modulates HBV replication. J. Med. Virol. 81(7), 1177–1183 (2009).

64. Quasdorff M, Protzer U: Control of hepatitis B virus at the level of transcription. J. Viral Hepat. 17(8), 527–536 (2010).

65. Quasdorff M, Hosel M, Odenthal M et al.: A concerted action of HNF4a and HNF1a links hepatitis B virus replication to hepatocyte differentiation. Cell Microbiol. 10(7), 1478–1490 (2008).

66. Hosel M, Quasdorff M, Wiegmann K et al.: Not interferon, but interleukin-6 controls early gene expression in hepatitis B virus infection. Hepatology 50(6), 1773–1782 (2009).

67. Ying C, Li Y, Leung CH, Robek MD, Cheng YC: Unique antiviral mechanism discovered in anti-hepatitis B virus research with a natural product analogue. Proc. Natl Acad. Sci. USA 104(20), 8526–8531 (2007).

68. Ott M, Thyagarajan SP, Gupta S: Phyllanthus amarus suppresses hepatitis B virus by interrupting interactions between HBV enhancer I and cellular transcription factors. Eur. J. Clin. Invest. 27(11), 908–915 (1997).

69. Hoeksema KA, Tyrrell DL: Inhibition of viral transcription using designed zinc finger proteins. Methods Mol. Biol. 649, 97–116 (2010).

Page 14: The life cycle of hepatitis B virus and antiviral targets

Future Virol. (2011) 6(5)612 future science group

Review Lucifora & Zoulim

70. Kim HJ, Yoo HS, Kim JC et al.: Antiviral effect of Curcuma longa Linn extract against hepatitis B virus replication. J. Ethnopharmacol. 124(2), 189–196 (2009).

71. Pang R, Tao JY, Zhang SL et al.: In vitro antiviral activity of lutein against hepatitis B virus. Phytother. Res. 24(11), 1627–1630 (2010).

72. Cui X, Inagaki Y, Xu H et al.: Anti-hepatitis B virus activities of cinobufacini and its active components bufalin and cinobufagin in HepG2.2.15 cells. Biol. Pharm. Bull. 33(10), 1728–1732 (2010).

73. Pang R, Tao J, Zhang S et al.: In vitro anti-hepatitis B virus effect of Hypericum perforatum L. J. Huazhong Univ. Sci. Technolog. Med. Sci. 30(1), 98–102 (2010).

74. Klein C, Bock CT, Wedemeyer H et al.: Inhibition of hepatitis B virus replication in vivo by nucleoside analogues and siRNA. Gastroenterology 125(1), 9–18 (2003).

75. Shlomai A, Shaul Y: Inhibition of hepatitis B virus expression and replication by RNA interference. Hepatology 37(4), 764–770 (2003).

76. Chen Y, Cheng G, Mahato RI: RNAi for treating hepatitis B viral infection. Pharm. Res. 25(1), 72–86 (2008).

n Reviews RNA interference strategies for the treatment of chronic HBV infection.

77. Wilson R, Purcell D, Netter HJ, Revill PA: Does RNA interference provide new hope for control of chronic hepatitis B infection? Antivir. Ther. 14(7), 879–889 (2009).

n Reviews RNA interference strategies for the treatment of chronic HBV infection.

78. Wang J, Feng SS, Wang S, Chen ZY: Evaluation of cationic nanoparticles of biodegradable copolymers as siRNA delivery system for hepatitis B treatment. Int. J. Pharm. 400(1–2), 194–200 (2010).

79. Teng X, Xu WZ, Hao ML et al.: Differential inhibition of lamivudine-resistant hepatitis B virus by allele-specific RNAi. J. Virol. Methods 168(1–2), 6–12 (2010).

80. Li Z, He ML, Yao H et al.: Inhibition of HBV replication and gene expression in vitro and in vivo with a single AAV vector delivering two shRNA molecules. BMB Rep. 42(1), 59–64 (2009).

81. Crowther C, Ely A, Hornby J et al.: Efficient inhibition of hepatitis B virus replication in vivo, using polyethylene glycol-modified adenovirus vectors. Hum. Gene Ther. 19(11), 1325–1331 (2008).

82. Xin XM, Li GQ, Guan XR et al.: Combination therapy of siRNAs mediates greater suppression on hepatitis B virus

cccDNA in HepG2.2.15 cell. Hepatogastroenterology 55(88), 2178–2183 (2008).

83. Offensperger WB, Offensperger S, Walter E et al.: In vivo inhibition of duck hepatitis B virus replication and gene expression by phosphorothioate modified antisense oligodeoxynucleotides. EMBO J. 12(3), 1257–1262 (1993).

84. Robaczewska M, Guerret S, Remy JS et al.: Inhibition of hepadnaviral replication by polyethylenimine-based intravenous delivery of antisense phosphodiester oligodeoxynucleotides to the liver. Gene Ther. 8(11), 874–881 (2001).

85. Feng Y, Kong YY, Wang Y, Qi GR: Inhibition of hepatitis B virus by hammerhead ribozyme targeted to the poly(A) signal sequence in cultured cells. Biol. Chem. 382(4), 655–660 (2001).

86. Feng Y, Kong YY, Wang Y, Qi GR: Intracellular inhibition of the replication of hepatitis B virus by hammerhead ribozymes. J. Gastroenterol. Hepatol. 16(10), 1125–1130 (2001).

87. Nash KL, Alexander GJ, Lever AM: Inhibition of hepatitis B virus by lentiviral vector delivered antisense RNA and hammerhead ribozymes. J. Viral Hepat. 12(4), 346–356 (2005).

88. Li X, Kuang E, Dai W, Zhou B, Yang F: Efficient inhibition of hepatitis B virus replication by hammerhead ribozymes delivered by hepatitis delta virus. Virus Res. 114(1–2), 126–132 (2005).

89. Wang CX, Lu YQ, Qi P, Chen LH, Han JX: Efficient inhibition of hepatitis B virus replication by hepatitis delta virus ribozymes delivered by targeting retrovirus. Virol. J. 7, 61 (2010).

90. Gordien E, Rosmorduc O, Peltekian C, Garreau F, Brechot C, Kremsdorf D: Inhibition of hepatitis B virus replication by the interferon-inducible MxA protein. J. Virol. 75(6), 2684–2691 (2001).

91. Zlotnick A, Johnson JM, Wingfield PW, Stahl SJ, Endres D: A theoretical model successfully identifies features of hepatitis B virus capsid assembly. Biochemistry 38(44), 14644–14652 (1999).

92. Hirsch RC, Lavine JE, Chang LJ, Varmus HE, Ganem D: Polymerase gene products of hepatitis B viruses are required for genomic RNA packaging as wel as for reverse transcription. Nature 344(6266), 552–555 (1990).

93. Junker-Niepmann M, Bartenschlager R, Schaller H: A short cis-acting sequence is required for hepatitis B virus pregenome encapsidation and sufficient for packaging of foreign RNA. EMBO J. 9(10), 3389–3396 (1990).

94. Nassal M: The arginine-rich domain of the hepatitis B virus core protein is required for pregenome encapsidation and productive viral positive-strand DNA synthesis but not for virus assembly. J. Virol. 66(7), 4107–4116 (1992).

95. Knaus T, Nassal M: The encapsidation signal on the hepatitis B virus RNA pregenome forms a stem-loop structure that is critical for its function. Nucleic Acids Res. 21(17), 3967–3975 (1993).

96. Porterfield JZ, Dhason MS, Loeb DD, Nassal M, Stray SJ, Zlotnick A: Full-length hepatitis B virus core protein packages viral and heterologous RNA with similarly high levels of cooperativity. J. Virol. 84(14), 7174–7184 (2010).

97. King RW, Ladner SK, Miller TJ et al.: Inhibition of human hepatitis B virus replication by AT-61, a phenylpropenamide derivative, alone and in combination with (-)b-l-2 ,́3´-dideoxy-3´-thiacytidine. Antimicrob. Agents Chemother. 42(12), 3179–3186 (1998).

98. Delaney WE, Edwards R, Colledge D et al.: Phenylpropenamide derivatives AT-61 and AT-130 inhibit replication of wild-type and lamivudine-resistant strains of hepatitis B virus in vitro. Antimicrob. Agents Chemother. 46(9), 3057–3060 (2002).

99. Feld JJ, Colledge D, Sozzi V, Edwards R, Littlejohn M, Locarnini SA: The phenylpropenamide derivative AT-130 blocks HBV replication at the level of viral RNA packaging. Antiviral Res. 76(2), 168–177 (2007).

100. Xu WS, Zhao KK, Miao XH et al.: Effect of oxymatrine on the replication cycle of hepatitis B virus in vitro. World J. Gastroenterol. 16(16), 2028–2037 (2010).

101. Katen SP, Chirapu SR, Finn MG, Zlotnick A: Trapping of hepatitis B virus capsid assembly intermediates by phenylpropenamide assembly accelerators. ACS Chem. Biol. 5(12), 1125–1136 (2010).

102. Deres K, Schroder CH, Paessens A et al.: Inhibition of hepatitis B virus replication by drug-induced depletion of nucleocapsids. Science 299(5608), 893–896 (2003).

103. Bourne C, Lee S, Venkataiah B et al.: Small-molecule effectors of hepatitis B virus capsid assembly give insight into virus life cycle. J. Virol. 82(20), 10262–10270 (2008).

104. Wu GY, Zheng XJ, Yin CC et al.: Inhibition of hepatitis B virus replication by Bay 41-4109 and its association with nucleocapsid disassembly. J. Chemother. 20(4), 458–467 (2008).

Page 15: The life cycle of hepatitis B virus and antiviral targets

www.futuremedicine.com 613future science group

The life cycle of hepatitis B virus & antiviral targets Review

105. Zhu X, Zhao G, Zhou X et al.: 2,4-diaryl-4,6,7,8-tetrahydroquinazolin-5(1H)-one derivatives as anti-HBV agents targeting at capsid assembly. Bioorg. Med. Chem. Lett. 20(1), 299–301 (2010).

106. Tomai E, Butz K, Lohrey C, Von Weizsacker F, Zentgraf H, Hoppe-Seyler F: Peptide aptamer-mediated inhibition of target proteins by sequestration into aggresomes. J. Biol. Chem. 281(30), 21345–21352 (2006).

107. Zlotnick A, Ceres P, Singh S, Johnson JM: A small molecule inhibits and misdirects assembly of hepatitis B virus capsids. J. Virol. 76(10), 4848–4854 (2002).

108. Schultz U, Summers J, Staeheli P, Chisari FV: Elimination of duck hepatitis B virus RNA-containing capsids in duck interferon-a-treated hepatocytes. J. Virol. 73(7), 5459–5465 (1999).

109. Biermer M, Puro R, Schneider RJ: Tumor necrosis factor alpha inhibition of hepatitis B virus replication involves disruption of capsid Integrity through activation of NF-kB. J. Virol. 77(7), 4033–4042 (2003).

110. Zoulim F, Locarnini S: Hepatitis B virus resistance to nucleos(t)ide analogues. Gastroenterology 137(5), 1593–1608, E1–E2 (2009).

n Reviews nucleos(t)ide analogs, their use for the treatment of chronic HBV infection and the resistances that can be developed.

111. Mason WS, Aldrich C, Summers J, Taylor JM: Asymmetric replication of duck hepatitis B virus DNA in liver cells: free minus-strand DNA. Proc. Natl Acad. Sci. USA 79(13), 3997–4001 (1982).

112. 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. 48(1), 1–9 (1983).

113. Roseman AM, Berriman JA, Wynne SA, Butler PJ, Crowther RA: A structural model for maturation of the hepatitis B virus core. Proc. Natl Acad. Sci. USA 102(44), 15821–15826 (2005).

114. Perlman DH, Berg EA, O’Connor PB, Costello CE, Hu J: Reverse transcription-associated dephosphorylation of hepadnavirus nucleocapsids. Proc. Natl Acad. Sci. USA 102(25), 9020–9025 (2005).

115. Wittkop L, Schwarz A, Cassany A et al.: Inhibition of protein kinase C phosphorylation of hepatitis B virus capsids

inhibits virion formation and causes intracellular capsid accumulation. Cell Microbiol. 12(7), 962–975 (2010).

116. Bruss V, Vieluf K: Functions of the internal pre-S domain of the large surface protein in hepatitis B virus particle morphogenesis. J. Virol. 69(11), 6652–6657 (1995).

117. Summers J, Smith PM, Horwich AL: Hepadnavirus envelope proteins regulate covalently closed circular DNA amplification. J. Virol. 64(6), 2819–2824 (1990).

118. Bottcher B, Tsuji N, Takahashi H et al.: Peptides that block hepatitis B virus assembly: ana lysis by cryomicroscopy, mutagenesis and transfection. EMBO J. 17(23), 6839–6845 (1998).

119. Patient R, Hourioux C, Roingeard P: Morphogenesis of hepatitis B virus and its subviral envelope particles. Cell Microbiol. 11(11), 1561–1570 (2009).

n Recent review on later stages of the HBV life cycle including capsid maturation, HBV-particle assembly and secretion.

120. Block TM, Lu X, Mehta AS et al.: Treatment of chronic hepadnavirus infection in a woodchuck animal model with an inhibitor of protein folding and trafficking. Nat. Med. 4(5), 610–614 (1998).

121. Simsek E, Lu X, Ouzounov S, Block TM, Mehta AS: a-Glucosidase inhibitors have a prolonged antiviral effect against hepatitis B virus through the sustained inhibition of the large and middle envelope glycoproteins. Antivir. Chem. Chemother. 17(5), 259–267 (2006).

122. Lazar C, Durantel D, Macovei A et al.: Treatment of hepatitis B virus-infected cells with alpha-glucosidase inhibitors results in production of virions with altered molecular composition and infectivity. Antiviral Res. 76(1), 30–37 (2007).

123. Serruys B, Van Houtte F, Verbrugghe P, Leroux-Roels G, Vanlandschoot P: Llama-derived single-domain intrabodies inhibit secretion of hepatitis B virions in mice. Hepatology 49(1), 39–49 (2009).

124. Neumann AU, Phillips S, Levine I et al.: Novel mechanism of antibodies to hepatitis B virus in blocking viral particle release from cells. Hepatology 52(3), 875–885 (2010).

125. Bremer CM, Bung C, Kott N, Hardt M, Glebe D: Hepatitis B virus infection is dependent on cholesterol in the viral envelope. Cell Microbiol. 11(2), 249–260 (2009).

126. Pollock S, Nichita NB, Bohmer A, Radulescu C, Dwek RA, Zitzmann N: Polyunsaturated liposomes are antiviral against hepatitis B and C viruses and HIV by

decreasing cholesterol levels in infected cells. Proc. Natl Acad. Sci. USA 107(40), 17176–17181 (2010).

n Interesting study demonstrating that polyunsaturated endoplasmic reticulum liposomes, which can lower cholesterol, can lead to decreased HBV-virion secretion and so decrease their infectivity.

127. Dougherty AM, Guo H, Westby G et al.: A substituted tetrahydro-tetrazolo-pyrimidine is a specific and novel inhibitor of hepatitis B virus surface antigen secretion. Antimicrob. Agents Chemother. 51(12), 4427–4437 (2007).

128. Huang KL, Lai YK, Lin CC, Chang JM: Involvement of GRP78 in inhibition of HBV secretion by Boehmeria nivea extract in human HepG2 2.2.15 cells. J. Viral Hepat. 16(5), 367–375 (2009).

129. Wang HP, Rogler CE: Topoisomerase I-mediated integration of hepadnavirus DNA in vitro. J. Virol. 65(5), 2381–2392 (1991).

130. Chemin I, Zoulim F: Hepatitis B virus induced hepatocellular carcinoma. Cancer Lett. 286(1), 52–59 (2009).

131. Liaw YF: Hepatitis B virus replication and liver disease progression: the impact of antiviral therapy. Antivir. Ther. 11(6), 669–679 (2006).

132. 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. 21(4), 525–535 (2002).

133. Hsieh YH, Su IJ, Wang HC et al.: Pre-S mutant surface antigens in chronic hepatitis B virus infection induce oxidative stress and DNA damage. Carcinogenesis 25(10), 2023–2032 (2004).

134. Benhenda S, Cougot D, Buendia MA, Neuveut C: Hepatitis B virus X protein molecular functions and its role in virus life cycle and pathogenesis. Adv. Cancer Res. 103, 75–109 (2009).

135. Wieland S, Thimme R, Purcell RH, Chisari FV: Genomic ana lysis of the host response to hepatitis B virus infection. Proc. Natl Acad. Sci. USA 101(17), 6669–6674 (2004).

136. Wieland SF, Chisari FV: Stealth and cunning: hepatitis B and hepatitis C viruses. J. Virol. 79(15), 9369–9380 (2005).

137. Lucifora J, Durantel D, Testoni B, Hantz O, Levrero M, Zoulim F: Control of hepatitis B virus replication by innate response of HepaRG cells. Hepatology 51(1), 63–72 (2010).

138. Ait-Goughoute M, Lucifora J, Zoulim F, Durantel D: Innate antiviral immune response to hepatitis B virus. Viruses 2, 1394–1410 (2010).

Page 16: The life cycle of hepatitis B virus and antiviral targets

Future Virol. (2011) 6(5)614 future science group

Review Lucifora & Zoulim

139. Nassal M: Hepatitis B viruses: reverse transcription a different way. Virus Res. 134(1–2), 235–249 (2008).

140. Lambert C, Doring T, Prange R: Hepatitis B virus maturation is sensitive to functional inhibition of ESCRT-III, Vps4, and g2-adaptin. J. Virol. 81(17), 9050–9060 (2007).

141. Rehermann B, Nascimbeni M: Immunology of hepatitis B virus and hepatitis C virus infection. Nat. Rev. Immunol. 5(3), 215–229 (2005).

142. Michel ML, Tiollais P: Hepatitis B vaccines: protective efficacy and therapeutic potential. Pathol. Biol. (Paris) 58(4), 288–295 (2010).

143. Protzer U, Abken H: Can engineered ‘designer’ T cells outsmart chronic hepatitis B? Hepat. Res. Treat. 2010, 901216 (2010).

144. De Clercq E: Strategies in the design of antiviral drugs. Nat. Rev. Drug Discov. 1(1), 13–25 (2002).

145. Vincent IE, Lucifora J, Durantel D et al.: Inhibitory effect of the combination of CpG-induced cytokines with lamivudine against hepatitis B virus replication in vitro. Antivir. Ther. 14(1), 131–135 (2009).

Website201. WHO: Hepatitis B factsheet

www.who.int/mediacentre/factsheets/fs204/en


Recommended