Topic Highlight Open Access
Copyright ©2014 Baishideng Publishing Group Inc. All rights reserved.
World J Gastroenterol. Jun 28, 2014; 20(24): 7707-7717
Published online Jun 28, 2014. doi: 10.3748/wjg.v20.i24.7707
Emerging antivirals for the treatment of hepatitis B
Xue-Yan Wang, Hong-Song Chen, Peking University Hepatology Institute, Peking University People’s Hospital, Beijing 100044, China
Xue-Yan Wang, Hong-Song Chen, Beijing Key Laboratory of Hepatitis C and Immunotherapy for Liver Diseases, Beijing 100044, China
Author contributions: Wang XY drafted and revised the manuscript; Chen HS revised the manuscript for intellectual content and gave final approval of the version to be published.
Supported by National Natural Science Foundation of China, No. 31340023, No. 91029741, No. 81001072 and No. 81171550; National Key Sci-Tech Special Project of China, No. 2012ZX10002011-006; Specialized Research Fund for the Doctoral Program of Higher Education, No. 20130001120075; and Peking University People’s Hospital Research and Development Funds, No. RDB2013-02
Correspondence to: Hong-Song Chen, MD, PhD, Professor, Peking University Hepatology Institute, Peking University People’s Hospital, Xizhimen South Road 11, Beijing 100044, China. chenhongsong@pkuph.edu.cn
Telephone: +86-10-88325726 Fax: +86-10-88325723
Received: October 29, 2013
Revised: February 13, 2014
Accepted: March 19, 2014
Published online: June 28, 2014

Abstract

Chronic infection with hepatitis B virus (HBV) constitutes a major global public health threat, causing substantial disease burdens such as liver cirrhosis and hepatocellular carcinoma, thus representing high unmet medical needs. Currently available therapies are safe, well tolerated, and highly effective in decreasing viremia and improving measured clinical outcomes with low rates of antiviral resistance. However, long-term management remains a clinical challenge, mainly due to the slow kinetics of HBV surface antigen clearance. In this article, we review emerging antivirals directed at novel targets derived from mechanisms of viral cellular entry, viral replication, viral assembly, and the host immune response, leading to preclinical and clinical trials for possible future therapeutic intervention. The recent therapeutic advances in the development of all categories of HBV inhibitors may pave the way for regimens of finite duration that result in long-lasting control of chronic hepatitis B infection.

Key Words: Hepatitis B, Antiviral, Nucleoside analogue, Nucleotide analogue, Non-nucleoside antivirals

Core tip: Despite the presence of an effective vaccine, chronic infection with hepatitis B virus remains a global public health problem. This review summary the emerging antivirals directed at novel targets derived from mechanisms of viral cellular entry, viral replication, viral assembly, and the host immune response, leading to preclinical and clinical trials for possible future therapeutic intervention.



INTRODUCTION

Despite the availability of an effective prophylactic vaccine, hepatitis B virus (HBV) infection remains a global public health problem. Worldwide, over 2 billion people have serological evidence of HBV, and approximately 360 million individuals are chronic HBV surface antigen (HBsAg) carriers[1]. The spectrum of disease and natural history of persistent HBV infection are diverse and variable, ranging from an inactive carrier state to progressive chronic hepatitis B (CHB), which is often associated with the development of cirrhosis and hepatocellular carcinoma (HCC)[2,3]. According to the Global Burden of Disease Study 2010, the total number of deaths in 2010 from HBV-related diseases was estimated to be 786000 worldwide[4].

In the last decade, therapeutic options for CHB have dramatically improved, which has resulted in more patients living with the disease in an inactive state. However, due to the variety of available drugs and the constantly changing guidelines, the management of HBV can be complex.

This article focuses on the current treatment options and various novel antiviral reagents for hepatitis B infection, including HBV life cycle inhibitors and host cellular target directed antiviral reagents.

HBV LIFE CYCLE AND POTENTIAL DRUG TARGETS

HBV, a member of the Hepadnaviridae family of viruses, is a small, enveloped, hepatotropic virus with an approximately 3.2-kb partially double-stranded, relaxed circular (rc) DNA genome[5]. The HBV life cycle is dependent on the reverse transcription of an RNA intermediate copy of the 3.2-kb DNA genome called pregenomic RNA (pgRNA). A simplified outline of the HBV replication cycle, the potential drug targets and the novel antiviral approaches are shown in Figure 1.

Figure 1
Figure 1 Hepatitis B virus replication cycle and potential drug targets. After hepatitis B virus (HBV) virions enter the hepatocyte, they are uncoated and transported to the nucleus, in which covalently closed circular DNA (cccDNA) is generated by repairing the partially double-stranded genome, followed by transcription of the cccDNA into the viral RNA gene products. Encapsidation of the pregenomic RNA occurs in the cytoplasm via a complex interaction among epsilon, core particles, and HBV polymerase. Reverse transcription leading to negative-strand and then positive-strand synthesis occurs within the viral nucleocapsid. After budding into the endoplasmic reticulum and Golgi apparatus, the nucleocapsid acquires an HBV surface antigen (HBsAg)-containing envelope and is released through the secretory pathway as progeny virions. Alternatively, nucleocapsids may also recycle their genomes into the nucleus for conversion to cccDNA. Potential inhibitors of the HBV life cycle include inhibitor of viral entry, small interfering double-stranded RNA (siRNA), inhibitor of nucleocapsid formation, inhibitor of HBV DNA polymerase, and inhibitor of HBsAg release.

The HBV lifecycle has recently been fully described[6-8]. Early events of the viral life cycle include attachment, penetration and uncoating. HBV infection of target cells begins when the virion attaches to the cell surface through interactions between viral envelope proteins and cellular receptors. Evidence suggests that a peptide derived from the pre-S1 region plays a crucial role in HBV binding and is an example of a possible therapeutic target[9]. The host receptor has long remained elusive although a recent report suggests sodium taurocholate cotransporting polypeptide, a multiple transmembrane transporter as a candidate receptor[10]. Following binding, the viral particle is internalized into the cytoplasm, where it undergoes a series of transformations involving the disassembly of the lipid bilayer and dissolution of the nucleocapsid[11,12]. Genomic DNA is transported to the cell nucleus as a result of a nuclear localization signal on the capsid protein. Inside the nucleus, the capsid dissociates and the rcDNA is converted into covalently closed circular DNA (cccDNA)[13]. HBV cccDNA is a unique episomal replicative intermediate that is responsible for the persistent infection of hepatocytes and that acts as the template for the transcription of 4 co-terminal mRNAs. These mRNAs are as follows: 3.5-kb pre-core mRNA (pre-C) and pgRNA; a 2.4-kb large surface mRNA; a 2.1-kb middle and small surface mRNA; and a 0.7-kb X mRNA[14,15]. The slightly longer pre-core 3.5-kb mRNA pre-C mRNA is translated to produce a precore protein that is further proteolytically processed into the HBV e antigen (HBeAg). In addition to serving as mRNA for viral capsid proteins and DNA polymerase, pgRNA serves as the template for the reverse transcriptional synthesis of viral DNA.

The late events in viral replication include viral assembly and release. Following transcription, viral mRNAs are transported to the cytoplasm where translation of the viral proteins, self-assembly of core protein molecules into nucleocapsids, and progeny genome replication occurs. After being transported to the cytoplasm, the pgRNA encodes the viral capsid protein (also termed core protein) and the HBV polymerase. Replication requires the encapsidation of the pgRNA by the core particle. This process is initiated by the interaction of the HBV polymerase with the pgRNA, which triggers encapsidation by the core protein to form the viral nucleocapsid[16]. Inside the lumen of the capsids, the viral polymerase serves as a protein primer to initiate negative-strand DNA synthesis with the pgRNA serving as the template, followed by synthesis of an incomplete plus-strand DNA. At this point the mature nucleocapsid is functionally equivalent to the capsid that entered the cell during primary infection, and has two potential fates. After budding into the endoplasmic reticulum and Golgi apparatus, the nucleocapsid acquires an HBsAg-containing envelope and is released through the secretory pathway as progeny virions[17,18]. Alternatively, nucleocapsids may also deliver their genomes back to the nucleus, causing amplification of the cccDNA pool (a process referred to as the recycling pathway)[19].

These multiple complex steps in the HBV life cycle of cellular entry, disassembly, replication, assembly, and release are all potential targets for novel therapies. Furthermore, complex host-immune responses to viral infection involving both innate and adaptive immunity offer immunological targets for viral control[20]. Combination therapies targeting multiple mechanisms are particularly attractive. The potential targets of representative new antiviral approaches currently undergoing evaluation in vitro, in vivo, or in clinical trials are shown in Figure 1.

CURRENT ANTIVIRAL REAGENTS

The goal of therapy for CHB is to improve quality of life and survival by preventing progression of the disease to cirrhosis, decompensated cirrhosis, end-stage liver disease, HCC and death[21]. This goal can be achieved if HBV replication can be suppressed with an effective treatment. The ideal end point is HBsAg loss, which however is infrequently achievable with the currently available anti-HBV agents. A more realistic endpoint is the induction of sustained or maintained virological remission.

Over the last 20 years, 2 interferon (IFN)-based therapies, both host cellular target-directed antiviral reagents, and 5 oral nucleoside/nucleotide analogues, all of which are HBV life cycle inhibitors, have been approved by the United States Food and Drug Administration for the treatment of CHB. These agents include IFN, pegylated interferon, lamivudine (LMV), adefovir dipivoxil (ADV), entecavir, telbivudine, and tenofovir disoproxil fumarate (TDF) (Table 1, Figure 2).

Table 1 Approved antivirals for hepatitis B.
Generic NamesBrand NameManufacturer NameOriginal approval or tentative approval date
Interferon alpha-2bIntron AScheringJuly, 1992
Pegylated interferonPegasysRoche13-May-05
LamivudineEpivir-HBVGlaxoSmithKline8-Dec-98
Adefovir dipivoxilhepseraGilead sciences20-Sep-02
EntecavirBaracludeBristol-myers squibb29-Mar-05
TelbivudineTyzekaNovartis25-Oct-06
Tenofovir disoproxil fumarateVireadGilead sciences8-Nov-08
Figure 2
Figure 2 Approval dates for chronic hepatitis B therapeutics. IFN: Interferon.
EMERGING ANTIVIRALS FOR HEPATITIS B
Inhibitor of HBV DNA polymerase

Clevudine: Clevudine [1-(2-deoxy-2-fluoro-β-L-arabinofuranosyl) thymine, L-FMAU], a thymidine nucleoside analogue, has potent antiviral activities against both HBV and Epstein-Barr virus but not human immunodeficiency virus (HIV). It acts as an inhibitor of the DNA-dependent DNA activity of HBV polymerase, as well as of reverse transcription and priming. Clevudine was approved for the treatment of CHB in South Korea and in the Philippines. However, the use of clevudine has not been endorsed by AASLD or EASL in their hepatitis B treatment guidelines[21,22] (Table 2).

Table 2 Emerging antivirals for hepatitis B virus treatment.
Family/drug nameMechanismStatusCompany
Nucleoside/nucleotide analogs
ClevudineInhibits viral DNA polymeraseApproved in S. Korea and PhilippinesBukwang/Eisai
MIV-210 (lagociclovir valactate)Inhibits viral DNA polymerasePhase IIMedivir/Daewoong
Besifovir (LB80380)Inhibits viral DNA polymerasePhase IIbLG Life Sciences
Tenofovir alafenamide (GS-7340)Inhibits viral DNA polymerasePhase IbGilead
CMX157Inhibits viral DNA polymerasePhase IChimerix
AGX-1009Inhibits viral DNA polymerasePhase I, ChinaAgenix
Non-nucleoside antivirals
Myrcludex-BEntry inhibitorPhase Ia, GermanyMyr-GmbH
Bay 41-4109Inhibits viral nucleocapsidPhase I, GermanyAiCuris
GLS 4Inhibits viral nucleocapsidPhase I, ChinaSunshine Lake
PhenylpropenamidesInhibits viral encapsidationPreclinical
REP 9 ACHBsAg release inhibitorPhase IbREPLICor Inc.
Nitazoxanide (alinia)Small MoleculePreclinicalRomark Labs
dd-RNAi compoundGene silencingPreclinicalBenitec/Biomics
ARC-520RNAigene silencerPhase IArrowhead Research
Immune-based
Zadaxin (thymosin alpha 1)ImmunomodulatorOrphan drug approval in United States for liver cancerSciClone
NOV-205 (BAM 205)ImmunomodulatorApproved in RussiaNovelos
GS-9620TLR7-agonistPhase IGilead
GI-13020HBV antigenPreclinicalGlobal Immune
DV-601Therapeutic HBV vaccinePhase IbDynavax

Clevudine has demonstrated potent antiviral activity during therapy and induced a sustained post treatment antiviral effect for 6 mo after a 12-wk treatment period[23]. A clinical trial of 1-year of clevudine treatment demonstrated it to be effective in suppressing the serum HBV DNA level and in normalizing the ALT level. Viral breakthrough associated with the rtM204I mutation occurred in 1 patient after 9 mo of clevudine treatment[24]. The M204I mutant in HBV RT plays a major role in clevudine resistance and leads to viral breakthrough during long-term clevudine treatment. In vitro drug susceptibility assays have revealed clevudine-associated mutants to be all resistant to lamivudine and usually sensitive to adefovir, entecavir, and tenofovir[25].

In a double-blind study to evaluate the safety and antiviral activity of clevudine (30 mg daily) over a 48-wk period compared with lamivudine (100 mg daily) in 92 HBeAg-positive CHB patients, clevudine was shown to be superior to lamivudine by yielding a higher rate of HBV DNA suppression[26]. In contrast, in a 2-year observational study comparing the clinical efficacy and safety of clevudine and entecavir, clevudine not only was no more effective than entecavir but also was associated with the development of drug resistance and muscle-related problems[27,28].

Clinical trials of long-term therapy with clevudine in the United States were terminated due to safety issues, such as drug-induced myopathy[29-31]. Furthermore, clevudine is more potent than is lamivudine, but does not seem to be more potent than entecavir or tenofovir. It has no advantage over the more potent antiviral regimen in current use, and therefore, is unlikely to be more universally approved in most markets given the safety concerns and the availability of the more effective drugs entecavir and tenofovir.

MIV-210 (Lagociclovir valactate): is a prodrug of the nucleoside analogue 3’-fluoro-2’, 3’-dideoxyguanosine (FLG) and has high oral bioavailability in humans and potent activity against HBV[32].

Preclinical in vitro and in vivo data reveal MIV-210 to be a good candidate for further testing as an agent against HIV and HBV. In Huh7 cells transiently transfected with different HBV constructs, FLG exhibits a broad spectrum of anti-HBV activity profiles against wild-type, lamivudine-resistant, adefovir-resistant, and lamivudine-plus-adefovir-resistant HBV mutants[33]. A study to determine the dose-related antiviral efficacy and safety of MIV-210 in chronically infected woodchucks showed that oral administration of MIV-210 at 20 or 60 mg/kg of body weight/day induced a rapid virological response[34]. The clinical development of this agent is ongoing. Following favorable plasma levels of MIV-210 and good oral bioavailability in phase I studies, a phase IIa clinical trial has been initiated in South Korea.

Besifovir (LB80380): is an acyclic nucleotide phosphonate with chemical similarity to adefovir and tenofovir. LB80380 is the prodrug of LB80331, which in turn is metabolized to LB80317, the active metabolite with antiviral effects against HBV after further intracellular phosphorylation to the triphosphate form[35].

In a phase Ib dose-finding study of LB80380 in treatment-naïve HBeAg-positive CHB patients, the safety, antiviral activity and pharmacokinetics were assessed[36]. A 3- to 4.2-log reduction of HBV DNA was observed with doses ranging from 30 to 240 mg daily, and viral suppression reached the optimal level when besifovir was administered at dose above 60 mg/d. In a phase IIa clinical study, LB80380 was found to be effective in reducing the viral load in lamivudine-resistant HBeAg-positive CHB patients for a period of 12 wk[37]. In both studies, LB80380 was safe and well tolerated up to a dose of 240 mg daily. In a phase IIb study that compared the safety and antiviral activity of besifovir with entecavir in Asian CHB patients for 48 wk, the proportions of patients who achieved undetectable HBV DNA, HBeAg loss, and HBeAg seroconversion were similar in all 3 groups of patients (besifovir 90 mg daily, besifovir 150 mg daily or entecavir 0.5 mg daily). Two doses of besifovir (90 mg and 150 mg) were non-inferior to entecavir 0.5 mg daily in treatment-naive CHB patients[38]. No antiviral drug resistance was observed in this study. A phase III study (ClinicalTrials.gov identifier NCT01937806) to assess the antiviral activity and safety of besifovir 150 mg compared with tenofovir 300 mg in CHB patients for 48 wk is not yet open for participant recruitment.

Tenofovir Alafenamide (GS-7340): associated with acyclic nucleotide phosphonates is currently under clinical development, with the goal of increasing oral availability, improving antiviral activity, and decreasing the potential for nephrotoxicity. Tenofovir alafenamide (TAF; formerly GS-7340) is a prodrug of TDF, achieving higher active metabolite concentrations in peripheral blood mononuclear lymphocytes and lymphatic tissues[39].

Inside cells, TAF is initially hydrolyzed to the intermediate tenofovir-alanine conjugate (TFV-Ala). TFV-Ala is converted to the parent tenofovir, which then undergoes subsequent phosphorylations to yield the active tenofovir diphosphate (TFV-DP) metabolite[40]. TFV-DP inhibits the activity of HIV reverse transcriptase by competing with natural substrates and causing DNA chain termination after incorporation into viral DNA[41]. TAF is being studied for the treatment of HIV infection, and is a potent inhibitor of hepatitis B virus. A phase I study to evaluate the safety and efficacy of TAF compared with that of TDF in treatment-naive and experienced adult subjects with chronic hepatitis B virus infection is currently recruiting participants (ClinicalTrials.gov Identifier: NCT01940341).

CMX157: is another investigational prodrug of tenofovir that is currently undergoing a phase I clinical study. CMX157 is a hexadecyloxypropyl conjugate of tenofovir with antiviral activity against HIV and HBV. In vitro, CMX157 is 267-fold more active than tenofovir against HIV-1 and 4.5-fold more active against HBV. CMX157 is orally bioavailable and has no toxicity in rats treated for 7 d at doses ranging from 10 to 100 mg/kg per day[42].

A phase I study to evaluate the safety, tolerability, and pharmacokinetics of a single dose of CMX157 in healthy volunteers has been completed (ClinicalTrials.gov Identifier: NCT01080820). Both TAF and CMX157 represent second-generation tenofovir analogues which may have an improved clinical profile.

AGX-1009: is a novel patented prodrug of tenofovir but is activated by a different molecular side-chain. Ongoing preclinical studies demonstrate good efficacy in inhibiting viral replication, and phase I trials are planned for 2013 in China.

Inhibitor of viral entry

Myrcludex-B: Specific inhibition of virus entry represents a rather new and attractive therapeutic concept for controlling acute and chronic infections. The establishment of the HBV-susceptible cell line HepaRG[43] and systems based on primary human hepatocytes and primary Tupaia belangeri hepatocytes have facilitated investigations of the cellular and viral determinants involved in HBV entry and resulted in the discovery of envelope protein-derived entry inhibitors. Both genetic and functional examination identified one domain in the N-terminusof HBV preS1 (amino acids 1-47) that is essential for HBV and hepatitis delta virus (HDV) infectivity.

Myrcludex-B, a synthetic lipopeptide consisting of the authentically myristoylated N-terminal 47 amino acids of the preS1 domain of the large viral envelope protein (L protein), specifically targets hepatocytes and efficiently blocks de novo HBV infection both in vitro[9,44-46], and in vivo[47]. Humanized chimeric uPA mice were first injected with HBV to permit an initial infection establishment, followed by administration of Myrcludex-B for either 3 d, 3 wk or 8 wk post-HBV inoculation. Myrcludex-B not only prevented the spread of HBV from infected human hepatocytes in vivo; but also hindered amplification of the cccDNA pool in initially infected hepatocytes[48]. A phase 0/1 clinical study to evaluate the safety, tolerability, pharmacokinetics, and immunogenicity of single ascending doses of Myrcludex-B in healthy volunteers is ongoing[49]. The data to date have shown Myrcludex-B to be very well tolerated in healthy volunteers for all investigated doses of up to 5 mg intravenous (iv) injection and 0.8 mg subcutaneous (sc) injection.

The entry inhibitor Myrcludex-B has been shown to prevent de novo establishment of HDV infection in vitro[50]. Furthermore, in a successfully established uPA/SCID mice model of HBV/HDV coinfection and superinfection, preclinical antiviral evaluations of the efficacy of Myrcludex-B in inhibiting the establishment of de novo HDV infection in vivo were performed[51].

Currently, the potential role of viral peptide-derived lipoproteins is primarily preventative and limited to post-exposure prophylaxis, the prevention of vertical transmission, and graft reinfection after liver transplant.

Inhibitor of nucleocapsid formation

Heteroaryldihydropyrimidines: The HBV capsid plays an indispensable role in the virus life cycle, participating in genome packaging, reverse transcription, intracellular trafficking, and maintenance of a stable infection. HBcAg serves as the structural unit in the assembly of the viral nucleocapsid, leading to nucleocapsid formation through a process of self-assembly. Perturbing HBV assembly, and thereby altering either the timing or the geometry of capsid formation, has great potential as an antiviral strategy. Recently, heteroaryldihydropyrimidines (HAPs), a family of assembly effectors, have been identified as highly potent non-nucleosidic inhibitors of HBV replication in vitro and in vivo[52-55]. HAPs act as allosteric effectors to increase the kinetics of assembly and strengthen dimer-dimer association to prevent the proper formation of viral capsids. Additionally, at high concentrations, HAPs misdirect viral assembly[56].

Bay 41-4109, a member of the HAP family, inhibits HBV replication by inducing inappropriate assembly and an aberrant transition to yield virus particles no longer competent for reverse transcription, budding, and/or nuclear transport[57]. In a humanized uPA/SCID HBV mouse model, a decrease in the HBV viral load of approximately 1 log (10) copies/mL was observed after treatment with Bay41-4109 for 5 d. Viral rebound was observed within 5 d of treatment cessation. A phase I study is ongoing, but no clinical results have been reported to date.

GLS4, a small molecular compound that inhibits HBV replication, is also a member of the HAP family. It is a potent inhibitor of the replication of both wild-type and ADV-resistant HBV mutant strains in vitro[58]. GLS4 is currently undergoing a phase I study in China to evaluate its safety, tolerability, and pharmacokinetics, and it exhibits good pharmacological behavior and tolerance. These inhibitors have a potential role as part of future therapeutic regimens against chronic HBV due to their specific mechanisms of action.

Phenylpropenamides: Molecules of the phenylpropenamide family of compounds, AT-61 and AT-130, have been shown to inhibit HBV replication in vitro[59,60]. These compounds are specific to HBV and have no activity against related viruses such as duck HBV, woodchuck HBV, and human immunodeficiency virus type 1 (HIV-1). Both AT-61 and AT-130 were found to be active against LMV-resistant HBV mutants. Studies with these compounds have shown that the phenylpropenamides appears to effectively inhibit HBV replication by interfering with the encapsidation process at the level of pregenomic RNA encapsidation, producing apparently normal capsids that lacked genetic material[61]. Subsequent studies revealed an assembly effector mechanism underlying the apparent blocking of RNA packaging[62]. Unlike HAPs, the effects of the phenylpropenamides are almost entirely kinetic, affecting only assembly reaction rate and timing while still producing normal capsids, with very little thermodynamic effects on capsid stability. Although this class of compounds has a favorable toxicity profile, clinical trials are still required.

Inhibitor of HBsAg release

REP 9 AC: Phosphorothioated oligonucleotides (PS-ONs), a novel class of compounds, constitute a promising microbicide approach. Early studies indicated the antiviral activity of PS-ONs to be independent of antisense activity but specifically dependent on their amphipathic characteristic[63]. Thus, PS-ONs with sequence-independent antiviral activity were describedas amphipathic DNA polymers (APs). APs possess broad spectrum antiviral activity, via multiple mechanisms, against a range of viruses, including human HIV-1[64], herpes simplex virus[65-67], arenavirus lymphocytic choriomeningitis virus[68], and hepatitis C virus (HCV)[69].

REP 9 AC is a 40-nucleotide polycytidine (alternating adenosine) amphipathic DNA polymer that inhibits the release of HBsAg from infected hepatocytes in HBV infected patients and allows patients to regain durable immunity by eliminating HBsAg-mediated immunosuppression. This agent is currently in a phase I/II clinical trial. The safety and efficacy of REP 9 AC in 8 HBsAg+ patients with HBV DNA levels of 6-12 log10 copies/ml were evaluated[70]. Interim clinical data demonstrated that REP 9AC led to rapid clearance of HBsAg from the serum and development of anti-HBs in as early as 7 d and in no more than 32 wk. Seven of the 8 treated patients exhibited residual to no levels of serum HBsAg and developed anti-HBs. Three of these 7 patients demonstrated durable immunological control over their infections, as evidenced by substantial reductions in serum HBV DNA within 20-27 wk of treatment. REP 9 AC, a modification of REP 9 AC to reduce pro-inflammatory activity and improve compound stability, shows similar efficacy with respect to HBsAg clearance and HBV DNA suppression with no pro-inflammatory side effects[71]. These results suggest that APs may become an important new tool in the treatment of CHB.

Small molecules

Nitazoxanide (alinia): Nitazoxanide, a thiazolide anti-infective agent, is active against anaerobic bacteria, protozoa, and a range of viruses in cell culture models, and it is currently in phase II clinical development for the treatment of chronic hepatitis C. Nitazoxanide was approved by the United States Food and Drug Administration in 2002 for treating diarrhea caused by Cryptosporidium spp. or Giardia lamblia in adults and children as young as 12 mo[72].

The antiviral activity of nitazoxanide was discovered by serendipity when patients with AIDS and HBV or HCV co-infection were treated for diarrhea. Nitazoxanide is potent inhibitor of HBV and displays synergistic interactions with LMV or ADV against HBV in vitro. Nitazoxanide is also effective against HBV mutants that are resistant to lamivudine and adefovir[73]. In preliminary open-label studies, nitazoxanide alone has shown evidence of efficacy in the treatment of CHB over 1-year course of therapy[74]. Nitazoxanide 500 mg twice daily resulted in a decrease in serum HBV DNA in all 4 HBeAg-positive patients, with undetectable HBV DNA in 2 of the 4 patients, loss of HBeAg in 3 patients, and loss of HBsAg in 1 patient. Nitazoxanide and related agents represent a class of small molecules that modulate host antiviral pathways via protein kinase activation, thereby acting as interferon immune enhancers[75].

Phase II clinical trials have demonstrated the efficacy and safety of nitazoxanide added to peginterferon with or without ribavirin in treating patients with chronic hepatitis C. Multiple clinical trials of nitazoxanide for treating chronic hepatitis C are underway or have been completed; however, no trials of this drug for treating CHB are currently registered with ClinicalTrials.gov.

Gene silencing

DNA-directed RNA Interference: RNA interference is an evolutionary conserved mechanism that employs short RNAs in association with an effector complex, referred to as the RNA-induced silencing complex, to regulate gene expression in a sequence-specific manner[76]. Two classes of small RNAs differing in origin and early processing pathways, mediate this process, namely, microRNAs (miRNAs) and small interfering RNAs (siRNAs)[77].

HBV is a promising target for an RNA interference approach because its compact genome lacks significant redundancy. siRNA has shown promise as a potential therapeutic agent by potently knocking down one or more viral transcripts (and consequently, antigens), for prolonged periods, both in vitro and in vivo[78-80].

ddRNAi technology involves inserting a DNA construct into a cell, triggering the production of double stranded RNA (dsRNA), which is then cleaved into siRNA as part of the RNA interference (RNAi) process, causing the destruction of the target mRNA and knocking-down or silencing the target gene expression. Although challenges remain in drug delivery, the most advanced nucleic acid-based therapeutics may enter the clinical realm in the near future.

ARC-520: A siRNA-based therapeutic agent called ARC-520 is designed to reduce the expression and release of new viral particles and the viral protein load by the mechanism of RNAi. The siRNAs in ARC-520 intervene at the point of DNA transcription, upstream of where nucleotide and nucleoside analogues act. In transient and transgenic mouse models of HBV infection, a single intravenous injection of ARC-520 targeting HBV sequences caused long-term, multi-log suppression of HBV RNA proteins, and viral DNA.

In a chimpanzee chronically infected with HBV and a high viral titer, a single intravenous injection of 2 mg/kg ARC-520 was well-tolerated and resulted in decreases in serum levels of HBsAg, HBeAg, and HBV DNA[81].

A phase I study of ARC-520 administered intravenously to healthy adult volunteers is being conducted in Melbourne, Australia. Each dose cohort includes 6 subjects randomized at a ratio of 1:2 (placebo: active) to receive a single intravenous injection of either placebo or ARC-520. This phase I trial is expected to be completed in the fourth quarter of 2013, and a phase IIa trial in chronic HBV patients is expected to begin in 2014.

Immune-based therapies

Zadaxin: Zadaxin (thymosin alpha 1), a synthetic peptide of 28 amino acids, is a substance found naturally in the circulation and produced in the body’s thymus gland. Zadaxin has been evaluated for its immunomodulatory activities and related therapeutic potential in several conditions and diseases, including cancer (such as HCC, lung cancer, and melanoma) and infectious disease (sepsis, infections after bone marrow transplant, lung infections including chronic obstructive pulmonary disorder (COPD), SARS, hepatitis B or C, and HIV). Investigation of Zadaxin’s mechanism of action at the cellular level has revealed both immune-modulating and direct-acting effects.

Interest in using Zadaxin for treating CHB has been based on the drug’s immunomodulating effects, which can trigger lymphocyte maturation, augment T-cell function, and reconstitute immune defects.

A study revealed that Zadaxin added to lamivudine was not superior to lamivudine alone and did not prevent resistance to lamivudine despite increased HBeAg seroconversion[82]. The data from a meta-analysis showed that, compared with IFN-α, the benefit of thymosin alpha-1 was not immediately significant at the end of therapy but that the virological, biochemical, and complete response had a tendency to increase or accumulate gradually after the therapy[83]. No persuasive data have yet emerged to suggest a great potential for Zadaxin.

NOV-205 (BAM 205): NOV-205 acts as a hepatoprotective agent with immunomodulating and anti-inflammatory properties and, thus, likely requires treatment periods longer than 14 d to affect a clinical response. Its regulatory approval in the Russian Federation under the trade name Molixan® in 2001 was based on clinical studies in 178 Russian hepatitis B and C patients. When used as mono-therapy for 1 mo in hepatitis B and for 2 mo in hepatitis C patients, NOV-205 has been shown to greatly reduce/eliminate viral loads and to significantly improve liver function. NOV-205 was also well-tolerated in those studies.

GS 9620: Toll-like receptors (TLRs) are components of the innate immune system and serve as a first line of defense against invading pathogens. TLR activation could represent a powerful and novel therapeutic strategy for the treatment of chronic HBV infection[84]. GS-9620, a potent selective TLR-7 agonist, was designed to have rapid clearance and low systemic levels after oral administration. In chimpanzees chronically infected with HBV, the effects of immune activation have been investigated using GS-9620 administered 3 times each week for 4 wk at 1 mg/kg and a 1-wk rest period, for a second cycle of 4 wk at 2 mg/kg[85]. A detailed evaluation was performed that included an assessment of the pharmacokinetics of GS-9620, viral load, IFN-stimulated gene (ISG) expression, cytokine and chemokine levels, and lymphocyte and NK cell activation, as well as safety and tolerability parameters. Short term administration of GS-9620 provided long-term suppression of serum and liver HBV DNA. Serum levels of HBsAg and HBeAg, as well as numbers of HBV antigen-positive hepatocytes, were reduced as hepatocyte apoptosis increased. In parallel, GS-9620 administration induced the production of IFN-α and other cytokines and chemokines, up-regulated ISGs expression, and activated NK-cell and lymphocyte subsets, confirming the activation of TLR-7 signaling.

The safety, tolerability, pharmacokinetics and pharmacodynamics of GS-9620 in 75 healthy volunteers were evaluated with a single ascending-dose up to 12 mg. GS-9620 demonstrated safety and pharmacodynamic activity at doses up to 12 mg and induced an antiviral response before systemic adverse events were observed[86].

The novel approach inhibiting HBV replication and inducing infected cell clearance represents a step toward the development of a true combination therapy for CHB.

GI-13020: HBV-specific T cell responses have been shown to have a positive association with infectious status in patients with chronic HBV, with the weakest T cell responses observed in patients with untreated chronic active infection and the strongest T cell responses observed in patients who have achieved seroconversion or cure. GI-13020 is a recombinant yeast-based biological product engineered to express a chimera of HBV X, S, and C antigens. GI-13020 is immunogenic in murine models and has also been used to stimulate human immune cell samples ex vivo to elicit HBV specific T cell responses, which could predict the immune responses in patients dosed with GI-13020.

A phase Ia trial assessing the safety, tolerability, and ability to elicit HBV-specific T cell responses of GI-13020 at various doses and with various regimens in healthy adults is ongoing but is not recruiting participants (ClinicalTrials.gov Identifier: NCT01779505). In the future, GI-13020 could be used in combination with HBV antivirals in an attempt to improve HBsAg seroconversion rates in patients with chronic HBV infection.

DV-601: Therapeutic vaccines may promote the resolution of chronic HBV infection through the stimulation of specific cytotoxic T-lymphocyte and B-cell antibody responses against dominant HBV-antigens. DV-601, which comprises recombinant HBsAg and HBcAg, is an investigational therapeutic vaccine. A phase Ib dose-escalation study has been completed to determine whether DV-601 would be well-tolerated and induce HBV-specific virological and immunological responses in CHB patients undergoing concurrent treatment with a nucleoside analogue (ClinicalTrials.gov, Identifier: NCT01023230).

In this initial dose-escalation study of DV-601, 6 injections of DV-601 were administered over a period of 12 wk. The primary objective was to assess the safety and tolerability of DV-601 by evaluating local and systemic adverse events on Day 99, including changes in laboratory analyses. Secondary objectives were to evaluate the virological response and immunogenicity of DV-601 based on viral load, and humoral and T cell immunological responses. DV-601 appears to be a safe and well-tolerated therapeutic vaccine for the treatment of CHB, and virological response is evident[87].

CONCLUSION

Currently available treatments offer major benefits to patients with chronic HBV infection and may prevent liver disease progression to liver cirrhosis, liver failure, and HCC; furthermore, these therapies may improve survival through robust and rapid viral suppression and the persistence of undetectable levels of serum HBV DNA. Currently, more effective and less resistance-prone antiviral agents are available to treat the HBV infection. The need for new antiviral agents that meet the ideal target product profile remains great. The emerging antivirals will provide additional and improved choices for optimized regimen development and will increase the confidence that many patients in more diverse settings will derive the full benefits of CHB treatment.

Footnotes

P- Reviewers: Chen C, Huang C, Lazar C S- Editor: Qi Y L- Editor: A E- Editor: Wang CH

References
1.  Hepatitis B vaccines Wkly Epidemiol Rec. 2009;84:405-419.  [PubMed]  [DOI]  [Cited in This Article: ]
2.  Fattovich G. Natural history and prognosis of hepatitis B. Semin Liver Dis. 2003;23:47-58.  [PubMed]  [DOI]  [Cited in This Article: ]
3.  Liaw YF, Chu CM. Hepatitis B virus infection. Lancet. 2009;373:582-592.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 929]  [Cited by in F6Publishing: 936]  [Article Influence: 62.4]  [Reference Citation Analysis (0)]
4.  Lozano R, Naghavi M, Foreman K, Lim S, Shibuya K, Aboyans V, Abraham J, Adair T, Aggarwal R, Ahn SY. Global and regional mortality from 235 causes of death for 20 age groups in 1990 and 2010: a systematic analysis for the Global Burden of Disease Study 2010. Lancet. 2012;380:2095-2128.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 9500]  [Cited by in F6Publishing: 9077]  [Article Influence: 756.4]  [Reference Citation Analysis (0)]
5.  Ganem D, Prince AM. Hepatitis B virus infection--natural history and clinical consequences. N Engl J Med. 2004;350:1118-1129.  [PubMed]  [DOI]  [Cited in This Article: ]
6.  Block TM, Guo H, Guo JT. Molecular virology of hepatitis B virus for clinicians. Clin Liver Dis. 2007;11:685-706, vii.  [PubMed]  [DOI]  [Cited in This Article: ]
7.  Locarnini S, Zoulim F. Molecular genetics of HBV infection. Antivir Ther. 2010;15 Suppl 3:3-14.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 117]  [Cited by in F6Publishing: 108]  [Article Influence: 8.3]  [Reference Citation Analysis (0)]
8.  Doo EC, Ghany MG. Hepatitis B virology for clinicians. Clin Liver Dis. 2010;14:397-408.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 13]  [Cited by in F6Publishing: 16]  [Article Influence: 1.1]  [Reference Citation Analysis (0)]
9.  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. 2005;79:1613-1622.  [PubMed]  [DOI]  [Cited in This Article: ]
10.  Yan H, Zhong G, Xu G, He W, Jing Z, Gao Z, Huang Y, Qi Y, Peng B, Wang H. Sodium taurocholate cotransporting polypeptide is a functional receptor for human hepatitis B and D virus. Elife. 2012;1:e00049.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1280]  [Cited by in F6Publishing: 1428]  [Article Influence: 119.0]  [Reference Citation Analysis (0)]
11.  Glebe D. Recent advances in hepatitis B virus research: a German point of view. World J Gastroenterol. 2007;13:8-13.  [PubMed]  [DOI]  [Cited in This Article: ]
12.  Kann M, Schmitz A, Rabe B. Intracellular transport of hepatitis B virus. World J Gastroenterol. 2007;13:39-47.  [PubMed]  [DOI]  [Cited in This Article: ]
13.  Sohn JA, Litwin S, Seeger C. Mechanism for CCC DNA synthesis in hepadnaviruses. PLoS One. 2009;4:e8093.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 33]  [Cited by in F6Publishing: 39]  [Article Influence: 2.6]  [Reference Citation Analysis (0)]
14.  Seeger C, Zoulim F, Mason WS. Hepadnaviridae. Fields virology. 5th ed. Philadelphia: Lippincott-Raven Publishers 2007; .  [PubMed]  [DOI]  [Cited in This Article: ]
15.  Seeger C, Mason WS. Hepatitis B virus biology. Microbiol Mol Biol Rev. 2000;64:51-68.  [PubMed]  [DOI]  [Cited in This Article: ]
16.  Pollack JR, Ganem D. An RNA stem-loop structure directs hepatitis B virus genomic RNA encapsidation. J Virol. 1993;67:3254-3263.  [PubMed]  [DOI]  [Cited in This Article: ]
17.  Kamimura T, Yoshikawa A, Ichida F, Sasaki H. Electron microscopic studies of Dane particles in hepatocytes with special reference to intracellular development of Dane particles and their relation with HBeAg in serum. Hepatology. 1981;1:392-397.  [PubMed]  [DOI]  [Cited in This Article: ]
18.  Roingeard P, Lu SL, Sureau C, Freschlin M, Arbeille B, Essex M, Romet-Lemonne JL. Immunocytochemical and electron microscopic study of hepatitis B virus antigen and complete particle production in hepatitis B virus DNA transfected HepG2 cells. Hepatology. 1990;11:277-285.  [PubMed]  [DOI]  [Cited in This Article: ]
19.  Tuttleman JS, Pourcel C, Summers J. Formation of the pool of covalently closed circular viral DNA in hepadnavirus-infected cells. Cell. 1986;47:451-460.  [PubMed]  [DOI]  [Cited in This Article: ]
20.  Bertoletti A, Maini MK, Ferrari C. The host-pathogen interaction during HBV infection: immunological controversies. Antivir Ther. 2010;15 Suppl 3:15-24.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 77]  [Cited by in F6Publishing: 80]  [Article Influence: 6.2]  [Reference Citation Analysis (0)]
21.  European Association For The Study Of The Liver. EASL clinical practice guidelines: Management of chronic hepatitis B virus infection. J Hepatol. 2012;57:167-185.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2323]  [Cited by in F6Publishing: 2338]  [Article Influence: 194.8]  [Reference Citation Analysis (0)]
22.  Lok AS, McMahon BJ. Chronic hepatitis B: update 2009. Hepatology. 2009;50:661-662.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2125]  [Cited by in F6Publishing: 2114]  [Article Influence: 140.9]  [Reference Citation Analysis (0)]
23.  Lee HS, Chung YH, Lee K, Byun KS, Paik SW, Han JY, Yoo K, Yoo HW, Lee JH, Yoo BC. A 12-week clevudine therapy showed potent and durable antiviral activity in HBeAg-positive chronic hepatitis B. Hepatology. 2006;43:982-988.  [PubMed]  [DOI]  [Cited in This Article: ]
24.  Ko SY, Kwon SY, Choe WH, Kim BK, Kim KH, Lee CH. Clinical and virological responses to clevudine therapy in chronic hepatitis B patients: results at 1 year of an open-labelled prospective study. Antivir Ther. 2009;14:585-590.  [PubMed]  [DOI]  [Cited in This Article: ]
25.  Kwon SY, Park YK, Ahn SH, Cho ES, Choe WH, Lee CH, Kim BK, Ko SY, Choi HS, Park ES. Identification and characterization of clevudine-resistant mutants of hepatitis B virus isolated from chronic hepatitis B patients. J Virol. 2010;84:4494-4503.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 35]  [Cited by in F6Publishing: 40]  [Article Influence: 2.9]  [Reference Citation Analysis (0)]
26.  Lau GK, Leung N. Forty-eight weeks treatment with clevudine 30 mg qd versus lamivudine 100 mg qd for chronic hepatitis B infection: a double-blind randomized study. Korean J Hepatol. 2010;16:315-320.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 9]  [Cited by in F6Publishing: 12]  [Article Influence: 0.9]  [Reference Citation Analysis (0)]
27.  Shin SR, Yoo BC, Choi MS, Lee DH, Song SM, Lee JH, Koh KC, Paik SW. A comparison of 48-week treatment efficacy between clevudine and entecavir in treatment-naïve patients with chronic hepatitis B. Hepatol Int. 2011;5:664-670.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 14]  [Cited by in F6Publishing: 18]  [Article Influence: 1.4]  [Reference Citation Analysis (0)]
28.  Yoon EL, Yim HJ, Lee HJ, Lee YS, Kim JH, Jung ES, Kim JH, Seo YS, Yeon JE, Lee HS. Comparison of clevudine and entecavir for treatment-naive patients with chronic hepatitis B virus infection: two-year follow-up data. J Clin Gastroenterol. 2011;45:893-899.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 15]  [Cited by in F6Publishing: 19]  [Article Influence: 1.5]  [Reference Citation Analysis (0)]
29.  Seok JI, Lee DK, Lee CH, Park MS, Kim SY, Kim HS, Jo HY, Lee CH, Kim DS. Long-term therapy with clevudine for chronic hepatitis B can be associated with myopathy characterized by depletion of mitochondrial DNA. Hepatology. 2009;49:2080-2086.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 88]  [Cited by in F6Publishing: 91]  [Article Influence: 6.1]  [Reference Citation Analysis (0)]
30.  Tak WY, Park SY, Jung MK, Jeon SW, Cho CM, Kweon YO, Kim SK, Choi YH. Mitochondrial myopathy caused by clevudine therapy in chronic hepatitis B patients. Hepatol Res. 2009;39:944-947.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 21]  [Cited by in F6Publishing: 23]  [Article Influence: 1.5]  [Reference Citation Analysis (0)]
31.  Kim BK, Oh J, Kwon SY, Choe WH, Ko SY, Rhee KH, Seo TH, Lim SD, Lee CH. Clevudine myopathy in patients with chronic hepatitis B. J Hepatol. 2009;51:829-834.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 49]  [Cited by in F6Publishing: 51]  [Article Influence: 3.4]  [Reference Citation Analysis (0)]
32.  Harmenberg J, LarssonT , Böttiger D, Augustsson E, Mardh G, Öberg B. Pharmacokinetic evaluation of the hepatitis B nucleoside analogue MIV-210 in human volunteers, abstr 150. Antivir Res. 2002;53:3.  [PubMed]  [DOI]  [Cited in This Article: ]
33.  Jacquard AC, Brunelle MN, Pichoud C, Durantel D, Carrouée-Durantel S, Trepo C, Zoulim F. In vitro characterization of the anti-hepatitis B virus activity and cross-resistance profile of 2’,3’-dideoxy-3’-fluoroguanosine. Antimicrob Agents Chemother. 2006;50:955-961.  [PubMed]  [DOI]  [Cited in This Article: ]
34.  Michalak TI, Zhang H, Churchill ND, Larsson T, Johansson NG, Oberg B. Profound antiviral effect of oral administration of MIV-210 on chronic hepadnaviral infection in a woodchuck model of hepatitis B. Antimicrob Agents Chemother. 2009;53:3803-3814.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 19]  [Cited by in F6Publishing: 20]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
35.  Fung J, Lai CL, Yuen MF. LB80380: a promising new drug for the treatment of chronic hepatitis B. Expert Opin Investig Drugs. 2008;17:1581-1588.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 19]  [Cited by in F6Publishing: 21]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
36.  Yuen MF, Kim J, Kim CR, Ngai V, Yuen JC, Min C, Kang HM, Shin BS, Yoo SD, Lai CL. A randomized placebo-controlled, dose-finding study of oral LB80380 in HBeAg-positive patients with chronic hepatitis B. Antivir Ther. 2006;11:977-983.  [PubMed]  [DOI]  [Cited in This Article: ]
37.  Yuen MF, Han KH, Um SH, Yoon SK, Kim HR, Kim J, Kim CR, Lai CL. Antiviral activity and safety of LB80380 in hepatitis B e antigen-positive chronic hepatitis B patients with lamivudine-resistant disease. Hepatology. 2010;51:767-776.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 34]  [Cited by in F6Publishing: 38]  [Article Influence: 2.7]  [Reference Citation Analysis (0)]
38.  Lai CL, Ahn SH, Lee KS, Um SH, Cho M, Yoon SK, Lee JW, Park NH, Kweon YO, Sohn JH. Phase IIb multicentred randomised trial of besifovir (LB80380) versus entecavir in Asian patients with chronic hepatitis B. Gut. 2014;63:996-1004.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 54]  [Cited by in F6Publishing: 58]  [Article Influence: 5.8]  [Reference Citation Analysis (0)]
39.  Lee WA, He GX, Eisenberg E, Cihlar T, Swaminathan S, Mulato A, Cundy KC. Selective intracellular activation of a novel prodrug of the human immunodeficiency virus reverse transcriptase inhibitor tenofovir leads to preferential distribution and accumulation in lymphatic tissue. Antimicrob Agents Chemother. 2005;49:1898-1906.  [PubMed]  [DOI]  [Cited in This Article: ]
40.  Birkus G, Kutty N, He GX, Mulato A, Lee W, McDermott M, Cihlar T. Activation of 9-[(R)-2-[[(S)-[[(S)-1-(Isopropoxycarbonyl)ethyl]amino] phenoxyphosphinyl]-methoxy]propyl]adenine (GS-7340) and other tenofovir phosphonoamidate prodrugs by human proteases. Mol Pharmacol. 2008;74:92-100.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 79]  [Cited by in F6Publishing: 81]  [Article Influence: 5.1]  [Reference Citation Analysis (0)]
41.  García-Lerma JG, Aung W, Cong ME, Zheng Q, Youngpairoj AS, Mitchell J, Holder A, Martin A, Kuklenyik S, Luo W. Natural substrate concentrations can modulate the prophylactic efficacy of nucleotide HIV reverse transcriptase inhibitors. J Virol. 2011;85:6610-6617.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 61]  [Cited by in F6Publishing: 61]  [Article Influence: 4.7]  [Reference Citation Analysis (0)]
42.  Painter GR, Almond MR, Trost LC, Lampert BM, Neyts J, De Clercq E, Korba BE, Aldern KA, Beadle JR, Hostetler KY. Evaluation of hexadecyloxypropyl-9-R-[2-(Phosphonomethoxy)propyl]- adenine, CMX157, as a potential treatment for human immunodeficiency virus type 1 and hepatitis B virus infections. Antimicrob Agents Chemother. 2007;51:3505-3509.  [PubMed]  [DOI]  [Cited in This Article: ]
43.  Gripon P, Rumin S, Urban S, Le Seyec J, Glaise D, Cannie I, Guyomard C, Lucas J, Trepo C, Guguen-Guillouzo C. Infection of a human hepatoma cell line by hepatitis B virus. Proc Natl Acad Sci USA. 2002;99:15655-15660.  [PubMed]  [DOI]  [Cited in This Article: ]
44.  Barrera A, Guerra B, Notvall L, Lanford RE. Mapping of the hepatitis B virus pre-S1 domain involved in receptor recognition. J Virol. 2005;79:9786-9798.  [PubMed]  [DOI]  [Cited in This Article: ]
45.  Glebe D, Urban S, Knoop EV, Cag N, Krass P, Grün S, Bulavaite A, Sasnauskas K, Gerlich WH. Mapping of the hepatitis B virus attachment site by use of infection-inhibiting preS1 lipopeptides and tupaia hepatocytes. Gastroenterology. 2005;129:234-245.  [PubMed]  [DOI]  [Cited in This Article: ]
46.  Schulze A, Schieck A, Ni Y, Mier W, Urban S. Fine mapping of pre-S sequence requirements for hepatitis B virus large envelope protein-mediated receptor interaction. J Virol. 2010;84:1989-2000.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 162]  [Cited by in F6Publishing: 173]  [Article Influence: 11.5]  [Reference Citation Analysis (0)]
47.  Petersen J, Dandri M, Mier W, Lütgehetmann M, Volz T, von Weizsäcker F, Haberkorn U, Fischer L, Pollok JM, Erbes B. Prevention of hepatitis B virus infection in vivo by entry inhibitors derived from the large envelope protein. Nat Biotechnol. 2008;26:335-341.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 309]  [Cited by in F6Publishing: 325]  [Article Influence: 20.3]  [Reference Citation Analysis (0)]
48.  Volz T, Allweiss L, Ben MBarek M, Warlich M, Lohse AW, Pollok JM, Alexandrov A, Urban S, Petersen J, Lütgehetmann M. The entry inhibitor Myrcludex-B efficiently blocks intrahepatic virus spreading in humanized mice previously infected with hepatitis B virus. J Hepatol. 2013;58:861-867.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 250]  [Cited by in F6Publishing: 249]  [Article Influence: 22.6]  [Reference Citation Analysis (0)]
49.  Haefeli WE, Blank A, Mikus G, Mier W, Alexandrov A, Urban S. Successful first administration of Myrcludex B, a first-in-class Hepatitis B and D Virus entry inhibitor, in humans. Hepatology. 2012;56 Supplement:369.  [PubMed]  [DOI]  [Cited in This Article: ]
50.  Engelke M, Mills K, Seitz S, Simon P, Gripon P, Schnölzer M, Urban S. Characterization of a hepatitis B and hepatitis delta virus receptor binding site. Hepatology. 2006;43:750-760.  [PubMed]  [DOI]  [Cited in This Article: ]
51.  Lütgehetmann M, Mancke LV, Volz T, Helbig M, Allweiss L, Bornscheuer T, Pollok JM, Lohse AW, Petersen J, Urban S. Humanized chimeric uPA mouse model for the study of hepatitis B and D virus interactions and preclinical drug evaluation. Hepatology. 2012;55:685-694.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 165]  [Cited by in F6Publishing: 171]  [Article Influence: 14.3]  [Reference Citation Analysis (0)]
52.  Weber O, Schlemmer KH, Hartmann E, Hagelschuer I, Paessens A, Graef E, Deres K, Goldmann S, Niewoehner U, Stoltefuss J. Inhibition of human hepatitis B virus (HBV) by a novel non-nucleosidic compound in a transgenic mouse model. Antiviral Res. 2002;54:69-78.  [PubMed]  [DOI]  [Cited in This Article: ]
53.  Deres K, Schröder CH, Paessens A, Goldmann S, Hacker HJ, Weber O, Krämer T, Niewöhner U, Pleiss U, Stoltefuss J. Inhibition of hepatitis B virus replication by drug-induced depletion of nucleocapsids. Science. 2003;299:893-896.  [PubMed]  [DOI]  [Cited in This Article: ]
54.  Wu GY, Zheng XJ, Yin CC, Jiang D, Zhu L, Liu Y, Wei L, Wang Y, Chen HS. Inhibition of hepatitis B virus replication by Bay 41-4109 and its association with nucleocapsid disassembly. J Chemother. 2008;20:458-467.  [PubMed]  [DOI]  [Cited in This Article: ]
55.  Brezillon N, Brunelle MN, Massinet H, Giang E, Lamant C, DaSilva L, Berissi S, Belghiti J, Hannoun L, Puerstinger G. Antiviral activity of Bay 41-4109 on hepatitis B virus in humanized Alb-uPA/SCID mice. PLoS One. 2011;6:e25096.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 68]  [Cited by in F6Publishing: 71]  [Article Influence: 5.5]  [Reference Citation Analysis (0)]
56.  Stray SJ, Bourne CR, Punna S, Lewis WG, Finn MG, Zlotnick A. A heteroaryldihydropyrimidine activates and can misdirect hepatitis B virus capsid assembly. Proc Natl Acad Sci USA. 2005;102:8138-8143.  [PubMed]  [DOI]  [Cited in This Article: ]
57.  Stray SJ, Zlotnick A. BAY 41-4109 has multiple effects on Hepatitis B virus capsid assembly. J Mol Recognit. 2006;19:542-548.  [PubMed]  [DOI]  [Cited in This Article: ]
58.  Wang XY, Wei ZM, Wu GY, Wang JH, Zhang YJ, Li J, Zhang HH, Xie XW, Wang X, Wang ZH. In vitro inhibition of HBV replication by a novel compound, GLS4, and its efficacy against adefovir-dipivoxil-resistant HBV mutations. Antivir Ther. 2012;17:793-803.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 96]  [Cited by in F6Publishing: 83]  [Article Influence: 6.9]  [Reference Citation Analysis (0)]
59.  King RW, Ladner SK, Miller TJ, Zaifert K, Perni RB, Conway SC, Otto MJ. Inhibition of human hepatitis B virus replication by AT-61, a phenylpropenamide derivative, alone and in combination with (-)beta-L-2’,3’-dideoxy-3’-thiacytidine. Antimicrob Agents Chemother. 1998;42:3179-3186.  [PubMed]  [DOI]  [Cited in This Article: ]
60.  Delaney WE, Edwards R, Colledge D, Shaw T, Furman P, Painter G, Locarnini S. 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. 2002;46:3057-3060.  [PubMed]  [DOI]  [Cited in This Article: ]
61.  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. 2007;76:168-177.  [PubMed]  [DOI]  [Cited in This Article: ]
62.  Katen SP, Chirapu SR, Finn MG, Zlotnick A. Trapping of hepatitis B virus capsid assembly intermediates by phenylpropenamide assembly accelerators. ACS Chem Biol. 2010;5:1125-1136.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 125]  [Cited by in F6Publishing: 120]  [Article Influence: 8.6]  [Reference Citation Analysis (0)]
63.  Anderson KP, Fox MC, Brown-Driver V, Martin MJ, Azad RF. Inhibition of human cytomegalovirus immediate-early gene expression by an antisense oligonucleotide complementary to immediate-early RNA. Antimicrob Agents Chemother. 1996;40:2004-2011.  [PubMed]  [DOI]  [Cited in This Article: ]
64.  Vaillant A, Juteau JM, Lu H, Liu S, Lackman-Smith C, Ptak R, Jiang S. Phosphorothioate oligonucleotides inhibit human immunodeficiency virus type 1 fusion by blocking gp41 core formation. Antimicrob Agents Chemother. 2006;50:1393-1401.  [PubMed]  [DOI]  [Cited in This Article: ]
65.  Guzman EM, Cheshenko N, Shende V, Keller MJ, Goyette N, Juteau JM, Boivin G, Vaillant A, Herold BC. Amphipathic DNA polymers are candidate vaginal microbicides and block herpes simplex virus binding, entry and viral gene expression. Antivir Ther. 2007;12:1147-1156.  [PubMed]  [DOI]  [Cited in This Article: ]
66.  Bernstein DI, Goyette N, Cardin R, Kern ER, Boivin G, Ireland J, Juteau JM, Vaillant A. Amphipathic DNA polymers exhibit antiherpetic activity in vitro and in vivo. Antimicrob Agents Chemother. 2008;52:2727-2733.  [PubMed]  [DOI]  [Cited in This Article: ]
67.  Cardin RD, Bravo FJ, Sewell AP, Cummins J, Flamand L, Juteau JM, Bernstein DI, Vaillant A. Amphipathic DNA polymers exhibit antiviral activity against systemic murine Cytomegalovirus infection. Virol J. 2009;6:214.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 26]  [Cited by in F6Publishing: 28]  [Article Influence: 1.9]  [Reference Citation Analysis (0)]
68.  Lee AM, Rojek JM, Gundersen A, Ströher U, Juteau JM, Vaillant A, Kunz S. Inhibition of cellular entry of lymphocytic choriomeningitis virus by amphipathic DNA polymers. Virology. 2008;372:107-117.  [PubMed]  [DOI]  [Cited in This Article: ]
69.  Matsumura T, Hu Z, Kato T, Dreux M, Zhang YY, Imamura M, Hiraga N, Juteau JM, Cosset FL, Chayama K. Amphipathic DNA polymers inhibit hepatitis C virus infection by blocking viral entry. Gastroenterology. 2009;137:673-681.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 64]  [Cited by in F6Publishing: 70]  [Article Influence: 4.7]  [Reference Citation Analysis (0)]
70.  Mahtab MA, Bazinet M, Vaillant A. REP 9 AC: a potent HBsAg release inhibitor that elicits durable immunological control of chronic HBV infection. Hepatology. 2011;54:478A.  [PubMed]  [DOI]  [Cited in This Article: ]
71.  Mahtab MA, Bazinet M, Patient R, Roingeard P, Vaillant A. Nucleic acid polymers REP 9 AC/REP 9 AC’ elicit sustained immunologic control of chronic HBV infection. Glob Antiviral J. 2011;7 Suppl 1:64A.  [PubMed]  [DOI]  [Cited in This Article: ]
72.  Anderson VR, Curran MP. Nitazoxanide: a review of its use in the treatment of gastrointestinal infections. Drugs. 2007;67:1947-1967.  [PubMed]  [DOI]  [Cited in This Article: ]
73.  Korba BE, Montero AB, Farrar K, Gaye K, Mukerjee S, Ayers MS, Rossignol JF. Nitazoxanide, tizoxanide and other thiazolides are potent inhibitors of hepatitis B virus and hepatitis C virus replication. Antiviral Res. 2008;77:56-63.  [PubMed]  [DOI]  [Cited in This Article: ]
74.  Rossignol JF, Keeffe EB. Thiazolides: a new class of drugs for the treatment of chronic hepatitis B and C. Future Microbiol. 2008;3:539-545.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 37]  [Cited by in F6Publishing: 41]  [Article Influence: 2.6]  [Reference Citation Analysis (0)]
75.  Keeffe EB, Rossignol JF. Treatment of chronic viral hepatitis with nitazoxanide and second generation thiazolides. World J Gastroenterol. 2009;15:1805-1808.  [PubMed]  [DOI]  [Cited in This Article: ]
76.  Fire A, Xu S, Montgomery MK, Kostas SA, Driver SE, Mello CC. Potent and specific genetic interference by double-stranded RNA in Caenorhabditis elegans. Nature. 1998;391:806-811.  [PubMed]  [DOI]  [Cited in This Article: ]
77.  Wilson R, Purcell D, Netter HJ, Revill PA. Does RNA interference provide new hope for control of chronic hepatitis B infection? Antivir Ther. 2009;14:879-889.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 9]  [Cited by in F6Publishing: 10]  [Article Influence: 0.7]  [Reference Citation Analysis (0)]
78.  Klein C, Bock CT, Wedemeyer H, Wüstefeld T, Locarnini S, Dienes HP, Kubicka S, Manns MP, Trautwein C. Inhibition of hepatitis B virus replication in vivo by nucleoside analogues and siRNA. Gastroenterology. 2003;125:9-18.  [PubMed]  [DOI]  [Cited in This Article: ]
79.  McCaffrey AP, Nakai H, Pandey K, Huang Z, Salazar FH, Xu H, Wieland SF, Marion PL, Kay MA. Inhibition of hepatitis B virus in mice by RNA interference. Nat Biotechnol. 2003;21:639-644.  [PubMed]  [DOI]  [Cited in This Article: ]
80.  Morrissey DV, Lockridge JA, Shaw L, Blanchard K, Jensen K, Breen W, Hartsough K, Machemer L, Radka S, Jadhav V. Potent and persistent in vivo anti-HBV activity of chemically modified siRNAs. Nat Biotechnol. 2005;23:1002-1007.  [PubMed]  [DOI]  [Cited in This Article: ]
81.  Lanford RE, Wooddell CI, Chavez D, Oropeza C, Chu Q, Hamilton HL, McLachlan A, Given B, Anzalone CR, Lewis DL. ARC-520 RNAi therapeutic reduces hepatitis B virus DNA, S antigen and e antigen in a chimpanzee with a very high viral titer. Hepatology. 2013;58:1305.  [PubMed]  [DOI]  [Cited in This Article: ]
82.  Lee HW, Lee JI, Um SH, Ahn SH, Chang HY, Park YK, Hong SP, Moon YM, Han KH. Combination therapy of thymosin alpha-1 and lamivudine for HBeAg positive chronic hepatitis B: A prospective randomized, comparative pilot study. J Gastroenterol Hepatol. 2008;23:729-735.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 14]  [Cited by in F6Publishing: 16]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
83.  Yang YF, Zhao W, Zhong YD, Yang YJ, Shen L, Zhang N, Huang P. Comparison of the efficacy of thymosin alpha-1 and interferon alpha in the treatment of chronic hepatitis B: a meta-analysis. Antiviral Res. 2008;77:136-141.  [PubMed]  [DOI]  [Cited in This Article: ]
84.  Isogawa M, Robek MD, Furuichi Y, Chisari FV. Toll-like receptor signaling inhibits hepatitis B virus replication in vivo. J Virol. 2005;79:7269-7272.  [PubMed]  [DOI]  [Cited in This Article: ]
85.  Lanford RE, Guerra B, Chavez D, Giavedoni L, Hodara VL, Brasky KM, Fosdick A, Frey CR, Zheng J, Wolfgang G. GS-9620, an oral agonist of Toll-like receptor-7, induces prolonged suppression of hepatitis B virus in chronically infected chimpanzees. Gastroenterology. 2013;144:1508-117, 1508-117.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 283]  [Cited by in F6Publishing: 304]  [Article Influence: 27.6]  [Reference Citation Analysis (0)]
86.  Lopatin U, Wolfgang G, Tumas D, Frey CR, Ohmstede C, Hesselgesser J, Kearney B, Moorehead L, Subramanian GM, McHutchison JG. Safety, pharmacokinetics and pharmacodynamics of GS-9620, an oral Toll-like receptor 7 agonist. Antivir Ther. 2013;18:409-418.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 46]  [Cited by in F6Publishing: 45]  [Article Influence: 4.1]  [Reference Citation Analysis (0)]
87.  Spellman M, Martin JT. Treatment of chronic hepatitis b infection with DV-601, a therapeutic vaccine. J Hepatol. 2011;54:s302.  [PubMed]  [DOI]  [Cited in This Article: ]