• OPEN ACCESS

Inhibitor-Based Therapeutics for Treatment of Viral Hepatitis

  • Debajit Dey and
  • Manidipa Banerjee*,
 Author information
Journal of Clinical and Translational Hepatology 2016;4(3):248-257

DOI: 10.14218/JCTH.2016.00025

Abstract

Viral hepatitis remains a significant worldwide threat, in spite of the availability of several successful therapeutic and vaccination strategies. Complications associated with acute and chronic infections, such as liver failure, cirrhosis and hepatocellular carcinoma, are the cause of considerable morbidity and mortality. Given the significant burden on the healthcare system caused by viral hepatitis, it is essential that novel, more effective therapeutics be developed. The present review attempts to summarize the current treatments against viral hepatitis, and provides an outline for upcoming, promising new therapeutics. Development of novel therapeutics requires an understanding of the viral life cycles and viral effectors in molecular detail. As such, this review also discusses virally-encoded effectors, found to be essential for virus survival and replication in the host milieu, which may be utilized as potential candidates for development of alternative therapies in the future.

Keywords

Viral hepatitis, Viroporin, Inhibitor

Introduction

Hepatitis is a medical condition wherein the liver undergoes inflammation due to a plethora of reasons, including drug abuse, excessive alcohol abuse, disease conditions etc.1 When such inflammation, as manifested in symptoms such as jaundice, nausea, abdominal pain, malaise etc, is caused by viral infections, the condition is referred to as viral hepatitis.1 Five hepatotropic viruses – named hepatitis A, B, C, D and E viruses – target liver cells in humans and cause acute and chronic hepatitis. In addition, other viruses such as the adenovirus, cytomegalovirus (CMV) and Epstein-Barr virus (EBV), occasionally cause symptoms of hepatitis.2

While an acute infection in healthy, immunocompetent individuals is cleared spontaneously, complications like cirrhosis, hepatocellular carcinoma (HCC) and fulminant hepatic failure (FHF) may arise in immunocompromised individuals, due to associated secondary reasons such as existing infections, alcohol abuse, or genetic predisposition.1,3 HCC, the third leading cause of cancer-related deaths worldwide,4 is closely associated with hepatitis B virus (HBV) infections. Even though the therapeutic strategies devised till date are targeted towards chronic infections, treatment options become severely limited for advanced stage patients.1 In addition, current medications have significant side-effects, which poses an issue with disease management. Hence, there is an urgent requirement for safer and more potent drugs.

This review will focus on the therapeutics currently available for treating viral hepatitis of all forms. In addition, the potential of new therapeutics and targeted inhibitor-based therapies against viral membrane-penetrating peptides and viroporins, a group of virally encoded proteins involved in facilitating replication and other specific steps in the viral life cycle, are also discussed.

Life cycle of hepatotropic viruses

Life cycles of the known hepatotropic viruses – particularly A, B, C and E – have been studied in significant detail. Although the lack of appropriate cell culture systems, and the slow-growing nature of the virus, has hampered studies, considerable information is available on the entry, replication and exit mechanisms of these viruses, and on specific host-interacting partners for each virus. Hepatitis A virus (HAV) is thought to associate with a cell-surface protein receptor, HAVcr-1/TIM-1 (Hepatitis A virus cellular receptor 1/T-cell immunoglobulin and mucin domain 1), a member of the immunoglobulin superfamily, by which it gains entry into host cells.5 Post-entry, translation of the positive-sense RNA genome by the host ribosomal machinery, mediated via the presence of an internal ribosome entry site (IRES) encoded in the 5’-untranslated region (UTR) of viral RNA, results in the production of a large viral polyprotein. The polyprotein is cleaved into structural (VP1-2A, VP2, VP3, VP4) and non-structural proteins (2B, 2C, 3A, 3B, 3C, 3D) as well as intermediates (3CD, 2BC etc) by a virally-encoded protease 3Cpro. Following the cleavage event, the viral RNA replicase synthesizes a minus-sense strand corresponding to the viral RNA genome, which serves as a template for the production of multiple plus-sense strands. Meanwhile, structural proteins assemble to form virions, which package a majority of the plus-sense RNA generated during replication. Two specific cleavage events result in generation of mature infectious virions - the VP1-2A precursor cleavage and the VP4/VP2 junctional cleavage.6–11 Virions are subsequently released from the apical membrane of infected hepatocytes.11

Entry of hepatitis C virus (HCV) into target host cells is a multistep event, requiring several host components. The cellular receptors and surface molecules, which are thought to be involved in this process, include the C-type lectins liver/lymph node-specific ICAM-3-grabbing integrin (L-SIGN) and dendritic cell (DC)-specific intercellular adhesion molecule-3 (ICAM-3)-grabbing nonintegrin (DC-SIGN), glycosaminoglycans (GAGs), claudins 6 and 9 and CD-81.12–14 Binding to cellular receptor(s) is primarily mediated by the viral envelope proteins E1 and E2. Post-entry, the virus nucleocapsid disassembles releasing the plus-sense RNA genome in the cell cytosol, which acts as a template for 5’-IRES mediated translation. The single HCV polyprotein generated during translation, like the corresponding HAV polypeptide, also undergoes protease mediated co- and post-processing to yield the entire repertoire of viral structural and non-structural proteins.14 During HCV infection, membranous webs14–16 are formed, which are sites for RNA replication (NS5B mediated) and translational post-processing events. While the endoplasmic reticulum (ER) serves as the site for the formation of the viral nucleocapsid, envelope formation and final virus maturation occurs in the Golgi. Matured virions are exocytosed into the extracellular space.14

The life cycle of hepatitis E virus (HEV) is not well understood, primarily due to the non-availability of an appropriate cell culture system. Studies have shown the possible involvement of heat shock cognate 70 protein (HSC70) and heparan sulfate proteoglycans (HSPGs) in the initial attachment of HEV to its target cells, while the final entry is mediated by a yet unknown receptor.17,18 Several host factors, such as glucose-regulated protein 78 (Grp78) and heat shock protein 90 (HSP90), are also thought to be involved in viral entry.19,20 Capsid uncoating and RNA genome release in the cytosol is closely followed by translation of the viral open reading frame (ORF) 1, which encodes the entire non-structural cassette.21,22 Multiple copy synthesis of the positive-sense RNA genome is mediated by virus-encoded RNA-dependent RNA polymerase via a negative-sense RNA intermediate. The viral ORF2 encodes the capsid protein, which associates with the progeny viral genomes to form the nucleocapsids, followed by the intracellular transport and release of mature virions.21,22 It has been postulated that the viral ORF3 encodes a protein involved in intracellular trafficking process.21,22

HBV, unlike other hepatotropic viruses, has a distinct mechanism for propagation within its host. Cellular entry of incoming particles via endocytosis is initiated through interactions with HSPGs,23,24 followed by that with the sodium taurocholate cotransporting polypeptide (NTCP).24,25 After gaining access to the cytosol, the virus travels to the nuclear pore complex, where the DNA genome is released into the nucleus. The partially double-stranded DNA genome is subsequently converted into a covalently closed circular DNA (cccDNA) form which associates with histones and other nuclear proteins. Within the nucleus, transcription events lead to the production of mRNA, encoding for the reverse transcriptase and the nucleocapsid (pregenomic). In addition, transcripts encoding the surface antigens (subgenomic) are also produced. Both pre- and subgenomic transcrpits are transferred to the cytosol. Surface protein synthesis, to generate core and subviral particles, occurs in the ER. Translation products (viral polymerase and core protein) of the pregenomic RNA associate with protein kinases (PKs),26,27 HSPs27,28 and pregenomic RNA to yield mature core particles, which are either released from infected cells, or are recirculated to the nucleus to maintain the cccDNA level.25,28 Interestingly, while subviral particles are released via the secretory pathway involving the Golgi and ER, mature core particles are released through multivesicular particles.25

Although not much is known regarding the cellular entry process of hepatitis D virus (HDV), studies have suggested similarities with the HBV entry pathway involving interaction of HSPGs and NTCPs with the viral large hepatitis B surface antigen (L-HBsAg; pre-S1 domain).24,29 Post-entry, transfer of the RNA genome to the nucleus is facilitated by the virally-encoded hepatitis D antigen (HDAg).30 Within the nucleus, host RNA polymerases initiate genome replication, which proceeds via a rolling circle mechanism.30 Subsequent transcriptional and translational events lead to the production of the large delta antigen which undergoes prenylation prior to association with progeny viral genomes. Subsequent assembly and viral release requires assistance from HBV.30

Current treatment scenario

HAV

Currently there are no specific treatment options available for HAV. Infected individuals usually receive symptomatic treatment, with liver transplantation being the only viable option in cases of FHF.11 Vaccination, with inactivated HAV virions, is fairly effective for conferring protection against all prevalent strains of the virus found in the human population.31,32 Vaccination with attenuated virions has also been employed, but has met with limited success.33 In spite of active vaccination efforts, however, the number of clinical cases of HAV reported is quite high, being ∼1.5 million worldwide.11

HCV

Due to the mostly asymptomatic nature of acute HCV infection, treatment options are limited. However, studies have reported that interferon-based treatment at an early stage might be helpful.34 Individuals exhibiting HCV RNA and antibodies at detectable levels in their blood after more than 6 months post-infection are considered to be cases of chronic HCV infection. A combination therapy of ribavirin and Peg-IFN-α (pegylated interferon α), for a duration of either six months or one year, was the accepted treatment regimen for chronic cases of HCV infection up until 2011.35 However, achievement of a sustained virological response (SVR), which is the key to cure, was observed to vary between genotypes and populations.36 Western European patients infected with HCV genotype 1 reportedly achieve a higher SVR (50%) than the North American patient population (40%). The SVR achieved with this treatment in case of patients infected with genotypes 2, 3, 5 and 6 was observed to be higher in general compared to genotype 1-infected patients (SVR of up to 80%). In particular, genotype 2 was the found to be most susceptible (SVR > 80%).36 Direct-acting antivirals (DAAs) such as telaprevir and boceprevir (first-wave, first generation), which target the HCV NS3-4A serine protease, were introduced in 2011 for treating infections caused by HCV genotype 1.37 Phase III clinical trials on treatment-naïve patients found that administration of telaprevir and boceprevir in conjunction with ribavirin and Peg-IFN-α (triple-combination) was effective in achieving a SVR in the range between 65% and 75%.37 However, this treatment regimen comes with its own set of disadvantages, including significant side-effects and not being cost-effective in the case of patients with advanced fibrosis.37 Since 2011, the following DAAs have been approved which can be used either independently or in combination with Peg-IFN-α and/or ribavirin. These DAAs are (a) sofosbuvir (nucleotide analogue), (b) simeprevir (NS3-4A protease inhibitor), (c) daclatasvir (NS3-4A protease inhibitor), (d) paritaprevir (NS3-4A protease inhibitor), (e) ombitasvir (NS5A inhibitor), (f) asunaprevir (NS3-4A protease inhibitor), (g) grazoprevir (NS3-4A protease inhibitor) and (h) elbasvir (NS5A replication complex inhibitor).35 The rate of SVR achieved depends on the combination used, the genotype, the presence of resistant strains and the disease severity. Finally, attempts to create an effective vaccine against HCV have not been successful till date due to the extreme sequence variation of the virus genome.

HEV

Acute HEV infections in immunocompetent individuals usually culminate in spontaneous clearance of the virus after a certain time period. However, in severe cases leading to decreased liver functioning, administration of ribavirin was found to help in prompt clearance of the virus and inhibition of further liver damage.38 It remains unclear if ribavirin intake can arrest the progress towards complete liver failure (fulminant hepatitis). Till date, liver transplantation from appropriate donors remains the only choice for such patients.39 For treatment of transplantation patients suffering from chronic infection with HEV, the following course of action is usually undertaken post-transplantation. The first step involves the reduction of immunosuppression, which has been shown to have a 25% clearance rate.40 If this is unsuccessful, the next step usually involves monotherapy with ribavirin for an initial period of 90 days.39,41,42 Administration of Peg-IFN-α is also an option; however, due to its many side-effects and complications in solid organ transplantation recipient patients, it is not preferred.39 HEV infection in pregnant women is associated with significant complications like stillbirths, abortions, premature distribution and maternal or fetal death.43 Interestingly, significant differences exist in the severity of complications caused by HEV infection in diverse endemic populations. For instance, while the infection typically has severe outcomes in pregnant women from northern India, it follows a fairly benign course in populations from Egypt and western countries.44,45 Studies suggest that poor prenatal care and nutrition may constitute major contributory factors for such severe outcomes in selected populations.46 Efforts are ongoing to develop an effective vaccine against HEV, and currently a promising candidate (trade name: Hecolin) is in Phase IV of clinical trials (clinicaltrials.gov database).

HBV

As with other forms of hepatitis, acute infections with HBV are treated symptomatically47,48 and no specific treatment regimen exists. However, patients who develop acute liver failure resulting from severe HBV infection are administered oral anti-HBV medication.49 The major emphasis so far is on treatment of chronic infection of HBV. The currently approved drugs are interferons, Peg-IFN-2a for adults and IFN-α-2b for children, and nucleotide/nucleoside analogues or NAs (nucleoside analogues: lamivudine, entecavir, emtricitabine, telbivudine; nucleotide analogues: adefovir, tenofovir).50,51 Due to their high barrier to development of resistance, tenofovir and entecavir are used as the first-line drugs for treatment.50,51 Other analogues like lamivudine, adefovir and telbivudine are only prescribed when the more potent drugs are either not appropriate or not available.50,51 This is due to fact that these drugs either have a low barrier to resistance (telbivudine), are less efficacious (adefovir) or have been shown to develop resistance over long-term usage (lamivudine).50 Interferons are utilized as they enhance the host antviral immune response.50,51 The therapeutic strategy usually involves two specific routes applicable to chronic hepatitis B (CHB): (i) a short treatment (1 year) with Peg-IFNs or NAs and (ii) treatment with NAs on a long-term basis.

Patients undergoing liver transplantation due to HBV-related HCC or decompensated liver cirrhosis undergo a pre-transplantation treatment regime with a potent NA having a high resistance barrier in order to reduce the viral DNA levels to a minimum before transplantation.58,59 Post-treatment, a combination of adefovir and/or lamivudine with hepatitis B immunoglobulin (HBIg) has been found to be effective (graft rejection rate of less than 10%).59,60 Given the treatment scenario and the complications associated, prevention of HBV infections is a priority. An effective vaccine against HBV exists and is administered 3–4 times over a 6 month period.58

HDV

Till date, there are no known effective therapeutic strategies available for treatment of either the acute or chronic forms of HDV infection.61 Studies have shown that long-term treatment with Peg-IFNα helps in clearance of the virus in about 25% of the cases.61 The efficacy is similar when interferon is administered in combination with adefovir.61 Thus, in the absence of an effective antiviral therapy, liver transplantation is the only option for patients suffering from liver damage due to chronic infection. There are no commercially available vaccines against HDV; however, the vaccine available for HBV can be used to prevent co-infection of HBV and HDV.61

Viral inhibitors and their mechanism of action

Based on the current knowledge of the molecular details of viral life cycles and virus-host interactions, there can be several targets for designing inhibitors. The following classes of inhibitors are discussed below: (i) viral polymerase inhibitors, (ii) viral protease inhibitors, (iii) CyPA inhibitors and (iv) host lipid modulators.

Several new targets have been identified in HBV against which inhibitors have been designed, and many are presently at different stages of development or in clinical trials. These include potent polymerase inhibitors, entry inhibitors, cccDNA inhibitors, nucleocapsid assembly inhibitors, inhibitors of viral gene expression and HBsAg release inhibitors. Equally effective but safer derivatives of tenofovir and entecavir (tenofovir alafenamide and besofovir) have shown promising results in randomized phase III trials with respect to bone and renal safety profiles.74–77 Besofovir is another drug which is being tested as an alternative to entecavir.78,79 Myrcludex-B is an engineered lipopeptide that effectively blocks NTCP receptors, the entry points for incoming HBV virions.80 Mouse model studies with Myrcludex-B have shown a reduction in both cccDNA levels and virus spread, and phase II clinical trials are currently ongoing.80 Three types of site-specific DNA binding proteins are being engineered to target cccDNA: zinc finger nucleases (ZFNs), transcription activator-like effector nucleases (TALENs) and clustered regulatory interspaced short palindromic repeats (CRISPR)-Cas endonucleases.81In vitro studies in cells have shown that all three systems can deactivate cccDNA by either site-specific cleavage,82 by introducing frameshifts in viral ORFs83 or by specific disruption (CRISPR-Cas).84,85 Disubstituted sulphonamides (CCC-0975 and CCC-0346) have also been identified; these have been shown to prevent the formation of cccDNA from relaxed circular DNA in vitro.86 Compounds that prevent the encapsidation of viral pregenomic RNA, such as sulfamoylbenzamide and phenylpropenamide derivatives, also constitute attractive options for therapy.87,88 These are effective against both NA-resistant and wild-type HBV.89 Inhibition of viral gene expression using RNA interference (RNAi) is a very attractive option since low levels of viral antigens will allow the immune system to recover. ARC-520, one such drug under development, has shown promising results in vitro.90,91 A nucleic acid polymer, REP 2055, has been shown to be able to inhibit the release of HBsAg from infected hepatocytes in in vivo studies.92 Finally, recent studies have also shown that the HBV RnaseH, which is critical for virus replication, can also be an important drug target.93,94 As in the case of HBV, novel drug targets have been identified in other hepatotropic viruses as well (Table 1).

Table 1.

List of potential inhibitors against viral proteins currently under investigation

VirusInhibitorTargetReference
HCVHydroxyanthraquinoneNS3 helicaseFuruta et al, 2015100
HBVHydroxlate tropolonesRibonuclease HHu et al, 201393
Lu et al, 201494
HAVNiranthinNot knownLiu et al, 2014102
AZD1480JAK2Jiang et al, 2015103
SirtinolSirtuinKanda et al, 2015104
HDVLonafarnibViral prenylationKoh et al, 2015105

Viroporins as promising inhibitor targets

The virus life cycle begins when it infects a suitable host in order to make multiple copies of itself by utilizing the host machinery. The process from infection to generation of progeny virions is executed as a series of very well coordinated steps and involves attachment, entry, uncoating, replication-transcription-translation of genetic material, assembly and egress. The virus genome encodes a limited number of proteins, all of which can be categorized primarily into structural and non-structural proteins. While structural proteins generate the virus capsid, non-structural proteins are produced after infection and are usually not a part of the capsid.74,95 Recently, a set of viral proteins have been identified which play essential roles in the virus life cycle, primarily through their ability to interact with and penetrate cellular membranes. This class of multifunctional proteins, designated viroporins, form pores or channels in cellular membranes, which directly or indirectly facilitate the intracellular survival of the virus.96 It has been suggested that the ability of viroporins to alter ionic concentration and remodel host cell membranes, enhances viral uncoating, replication and egress.96 Usually viroporins are small peptides (up to 100 amino acids) and contain one or more transmembrane domain(s). Formation of pores or channels in cellular membranes is facilitated by homo-oligomerization between viroporins. Oligomerization levels can range from dimeric association (e.g. in HAV 2B)97 to large heptameric assemblies (e.g. in HCV p7).96 Till date, several viroporins have been identified and functionally characterized for both enveloped and non-enveloped viruses (Fig. 1, Table 2). Some of the well-known examples are influenza A virus M2, HIV-1 Vpu, picornavirus 2B and HCV p7.96

General schematic showing various steps in the life cycle of enveloped and non-enveloped viruses and the involvement of viroporins or viral membrane penetrating peptides.
Fig. 1.  General schematic showing various steps in the life cycle of enveloped and non-enveloped viruses and the involvement of viroporins or viral membrane penetrating peptides.

In the case of enveloped viruses, steps 1, 2, 3, 4 and 5 represent attachment, entry, uncoating and replication, assembly and egress, respectively. For non-enveloped viruses, steps 1, 2, 3, 4, 5 and 6 represent attachment, entry, release from the intracellular vesicle, uncoating and replication, assembly and egress processes.

Table 2.

List of reported viral membrane penetrating peptides and their role in the virus life cycle

NameLength (residue length)VirusStage of life cycle
Vpu77–86HIV-1Egress106
Vpr96HIV-1Replication116
M297Influenza AReplication and egress108
BM2115Influenza BReplication95
E76–109CoronavirusAssembly and egress109
3a274CoronavirusEgress110
Kcv94PBCV-1Replication111
VP423HAVEntry101
VP4125SV40Egress112
2B251HAVReplication97
2B97–99CoxsackievirusReplication and egress113
2B97PoliovirusReplication114
6K58–61AlphavirusEgress115
p763HCVAssembly107
μ129ReovirusEntry117
Pep4646IBDVEntry118

The central role of viroporins in facilitating several steps in the life cycle of viruses suggests that this class of viral proteins are promising candidates for development of inhibitor-based therapeutics. In fact, small molecule inhibitors have been successfully designed and tested against certain viroporins with encouraging outcomes; drugs such as amantadine and rimantadine have been shown to be effective in inhibiting the ion channel activity of M2.98,99 Interestingly, several influenza strains, with specific point mutations (Leu26Phe, Ser31Asn, Val27Ala, Ala30Thr, Leu28Phe Ser31Asn and Gly34Glu) which cause resistance to M2 inhibitors have been identified recently.79 These mutations most likely affect the drug binding sites of the M2 channel by altering binding pocket hydrophobicity or the overall conformation of the channel. The most prevalent point mutation is Ser31Asn, which has been identified in the H1N1, H7N9 and H1N5 strains of the virus. Attempts are currently being made to synthesize inhibitors against the resistant strains using derivatives of M2 inhibitors. Although novel effective derivatives have been obtained against several of the mutations, those against point mutations at Asn31 have proven to be difficult to generate.98

Hexamethylene amilorides (HMA), and its derivatives (e.g. BIT225), have been found to be effective in blocking Vpu channel activity and is thought to bind in the channel lumen. The BIT225 HMA derivative is currently undergoing clinical trials.98 This inhibitor is specific to HIV-1 and till date no resistant strains have been reported.

Coronavirus-encoded E peptides are also effectively blocked by HMA, as has been observed in electrophysiology studies using HEK-293T cells.98 Amantadine is also capable of inhibiting activity, but the effect requires significantly higher concentrations.98

Adamantanes, alkyl imino-sugars and HMA were identified as HCV p7 blockers based on protein based in vitro assays.98,100 However, studies reported inconsistency in efficacy data, which was preliminarily due to the genotypic/subtypic differences between the p7 channels, even though compounds like rimatadine and N-nonyl deoxynojirimycin (NN-DNJ) exhibited wide-spectrum activity amongst genotypes.98 Availability of high resolution structural data and in silico docking studies helped in unravelling the mechanism of action of these inhibitors.80 NN-DNJ was found to interfere with p7 oligomerization via interaction with a specific phenylalanine residue at the 25th position. Interestingly, the inhibitory activity of adamantane was found to be due to binding of the inhibitor at a peripheral site (Leu20) that is exposed on the membrane, separate from the NN-DNJ binding site.98 The disparate binding sites for inhibitors provides the possibility of using a combination therapy approach to treat resistant strains. Current drug development efforts typically use a combination of rational and high-throughput approaches. This has resulted in identification of compounds against resistant p7 phenotypes with higher potency at minimal concentrations.98

Current interest and the pool of available data leads to the conclusion that viroporin-based therapeutic approaches will subsequently play an important role in treating clinically relevant human pathogens. The approaches towards designing inhibitors for treatment of HCV and its resistant phenotypes can also be extended to the other hepatotropic viruses. In fact, recent work from our laboratory has characterized the membrane activity of capsid component VP4 and non-structural protein 2B from HAV.97,101 Utilizing a combination of biophysical studies, MD simulation and electron microscopy, it was shown that VP4 forms discreet pores of 5∼9 nm in artificial membranes. Our data thus indicates that VP4 is a membrane penetrating peptide which may allow HAV to escape from endosomes and gain access to the cellular cytosol. The potentially critical role played by VP4 in HAV entry makes it an important target for design of inhibitors.

We further investigated the non-structural 2B protein of HAV, which was found to contain a stretch of 60 residues in its C-terminus, and which has viroporin-like activity.97 A combination of crosslinking studies, biophysical assays and simulation was employed to determine that 2B probably forms dimers and generates ∼3 nm pores in membranes. The resulting alteration of membrane permeability is thought to be essential for HAV replication. It is probable that inhibition of the membrane activity of 2B would negatively impact the viral replication process. Thus, a treatment regimen against HAV, which includes a combination therapy targeting VP4 and 2B, could be a very promising and effective approach.

Conclusion and future perspectives

Given the essential roles played by viroporins and viral membrane penetrating peptides in enhancing several aspects of the virus life cycle, targeted inhibitor therapy holds promise for development of virus-specific drugs. Establishing the commonalities between the structures and functions of these membrane active components might make it easier to design small molecule inhibitors. Finally, given the promise of p7-based therapy against HCV, it is essential to pinpoint such potential candidates in other hepatotropic viruses, in order to expand therapy options against chronic and acute viral hepatitis.

Abbreviations

CMV: 

Cytomegalovirus

EBV: 

Epstein-Barr virus

HCC: 

Hepatocellular carcinoma

FHF: 

Fulminant hepatic failure

HBV: 

Hepatitis B virus

HAV: 

Hepatitis A virus

HCV: 

Hepatitis C virus

HEV: 

Hepatitis E virus

HDV: 

Hepatitis D virus

HAVcr-1/TIM-1: 

Hepatitis A virus cellular receptor 1/T-cell immunoglobulin and mucin domain 1

IRES: 

Internal ribosome entry site

5’-UTR: 

5’-Untranslated region

L-SIGN: 

Liver/lymph node-specific intercellular adhesion molecule-3-grabbing integrin

DC-SIGN: 

Dendritic cell-specific intercellular adhesion molecule-3

GAG: 

glycosaminoglycan

HSC70: 

Heat shock cognate 70 protein

HSPG: 

Heparan sulfate proteoglycans

Grp78: 

Glucose-regulated protein 78

HSP90: 

Heat shock protein 90

ORF1: 

Open reading frame 1

NTCP: 

sodium taurocholate cotransporting polypeptide

cccDNA: 

covalently closed circular DNA

PK: 

Protein kinase

NA: 

Nucleos(t)ide analog

IFN: 

Interferon

L-HBsAg: 

Large hepatitis B surface antigen

HDAg: 

Hepatitis D antigen

PegIFN-α: 

Pegylated IFN-α

SVR: 

Sustained virological response

HBeAg: 

Hepatitis B e antigen

HBIg: 

Hepatitis B immunoglobulin

CHB: 

Chronic hepatitis B

DAAs: 

Direct-acting antivirals

ZFNs: 

Zinc finger nucleases

TALEN’s: 

Transcription activator-like effector nucleases

CRISPR: 

Clustered regulatory interspaced short palindromic repeats

RNAi: 

RNA interference

HMA: 

Hexamethylene amilorides

NN-DNJ: 

N-nonyl deoxynojirimycin

Declarations

Acknowledgement

This work was supported by a grant from the Department of Biotechnology, India (BT/PR7439/MED/29/676/2012). D.D. was supported by a research fellowship from the Council of Scientific and Industrial Research (CSIR), India.

Conflict of interest

None

Authors’ contributions

Wrote the manuscript (DD, MB).

References

  1. Wood NJ. Viral hepatitis: progress and promise. Nat Rev Gastroenterol Hepatol 2011;8:239 View Article PubMed/NCBI
  2. Adams DH, Hubscher SG. Systemic viral infections and collateral damage in the liver. Am J Pathol 2006;168:1057-1059 View Article PubMed/NCBI
  3. Wasley A, Grytdal S, Gallagher K. Centers for Disease Control and Prevention (CDC). Surveillance for acute viral hepatitis—United States, 2006. MMWR Surveill Summ 2008;57:1-24 View Article PubMed/NCBI
  4. Raza A, Sood GK. Hepatocellular carcinoma review: current treatment, and evidence-based medicine. World J Gastroenterol 2014;20:4115-4127 View Article PubMed/NCBI
  5. Sui L, Zhang W, Chen Y, Zheng Y, Wan T, Zhang W. Human membrane protein Tim-3 facilitates hepatitis A virus entry into target cells. Int J Mol Med 2006;17:1093-1099 View Article PubMed/NCBI
  6. Anderson DA, Ross BC. Morphogenesis of hepatitis A virus: isolation and characterization of subviral particles. J Virol 1990;64:5284-5289 View Article PubMed/NCBI
  7. Beard MR, Cohen L, Lemon SM, Martin A. Characterization of recombinant hepatitis A virus genomes containing exogenous sequences at the 2A/2B junction. J Virol 2001;75:1414-1426 View Article PubMed/NCBI
  8. Cohen L, Bénichou D, Martin A. Analysis of deletion mutants indicates that the 2A polypeptide of hepatitis A virus participates in virion morphogenesis. J Virol 2002;76:7495-7505 View Article PubMed/NCBI
  9. Martin A, Bénichou D, Chao SF, Cohen LM, Lemon SM. Maturation of the hepatitis A virus capsid protein VP1 is not dependent on processing by the 3Cpro proteinase. J Virol 1999;73:6220-6227 View Article PubMed/NCBI
  10. Martin A, Escriou N, Chao SF, Girard M, Lemon SM, Wychowski C. Identification and site-directed mutagenesis of the primary (2A/2B) cleavage site of the hepatitis A virus polyprotein: functional impact on the infectivity of HAV RNA transcripts. Virology 1995;213:213-222 View Article PubMed/NCBI
  11. Martin A, Lemon SM. Hepatitis A virus: from discovery to vaccines. Hepatology 2006;43:S164-S172 View Article PubMed/NCBI
  12. Helle F, Dubuisson J. Hepatitis C virus entry into host cells. Cell Mol Life Sci 2008;65:100-112 View Article PubMed/NCBI
  13. Barth H, Liang TJ, Baumert TF. Hepatitis C virus entry: molecular biology and clinical implications. Hepatology 2006;44:527-535 View Article PubMed/NCBI
  14. Ashfaq UA, Javed T, Rehman S, Nawaz Z, Riazuddin S. An overview of HCV molecular biology, replication and immune responses. Virol J 2011;8:161 View Article PubMed/NCBI
  15. El-Hage N, Luo G. Replication of hepatitis C virus RNA occurs in a membrane-bound replication complex containing nonstructural viral proteins and RNA. J Gen Virol 2003;84:2761-2769 View Article PubMed/NCBI
  16. Gosert R, Egger D, Lohmann V, Bartenschlager R, Blum HE, Bienz K. Identification of the hepatitis C virus RNA replication complex in Huh-7 cells harboring subgenomic replicons. J Virol 2003;77:5487-5492 View Article PubMed/NCBI
  17. Kalia M, Chandra V, Rahman SA, Sehgal D, Jameel S. Heparan sulfate proteoglycans are required for cellular binding of the hepatitis E virus ORF2 capsid protein and for viral infection. J Virol 2009;83:12714-12724 View Article PubMed/NCBI
  18. Zhou Y, Emerson SU. Heat shock cognate protein 70 may mediate the entry of hepatitis E virus into host cells. J Clin Virol 2006;36:S155 View Article PubMed/NCBI
  19. Zheng ZZ, Miao J, Zhao M, Tang M, Yeo AE, Yu H. Role of heat-shock protein 90 in hepatitis E virus capsid trafficking. J Gen Virol 2010;91:1728-1736 View Article PubMed/NCBI
  20. Yu H, Li S, Yang C, Wei M, Song C, Zheng Z. Homology model and potential virus-capsid binding site of a putative HEV receptor Grp78. J Mol Model 2011;17:987-995 View Article PubMed/NCBI
  21. Cao D, Meng XJ. Molecular biology and replication of hepatitis E virus. Emerg Microbes Infect 2012;1:e17 View Article PubMed/NCBI
  22. Ahmad I, Holla RP, Jameel S. Molecular virology of hepatitis E virus. Virus Res 2011;161:47-58 View Article PubMed/NCBI
  23. Schulze A, Gripon P, Urban S. Hepatitis B virus infection initiates with a large surface protein-dependent binding to heparan sulfate proteoglycans. Hepatology 2007;46:1759-1768 View Article PubMed/NCBI
  24. Glebe D, Bremer CM. The molecular virology of hepatitis B virus. Semin Liver Dis 2013;33:103-112 View Article PubMed/NCBI
  25. Yan H, Zhong G, Xu G, He W, Jing Z, Gao Z. Sodium taurocholate cotransporting polypeptide is a functional receptor for human hepatitis B and D virus. Elife 2012;1:e00049 View Article PubMed/NCBI
  26. Kann M, Gerlich WH. Effect of core protein phosphorylation by protein kinase C on encapsidation of RNA within core particles of hepatitis B virus. J Virol 1994;68:7993-8000 View Article PubMed/NCBI
  27. Wittkop L, Schwarz A, Cassany A, Grün-Bernhard S, Delaleau M, Rabe B. Inhibition of protein kinase C phosphorylation of hepatitis B virus capsids inhibits virion formation and causes intracellular capsid accumulation. Cell Microbiol 2010;12:962-975 View Article PubMed/NCBI
  28. Beck J, Nassal M. Hepatitis B virus replication. World J Gastroenterol 2007;13:48-64 View Article PubMed/NCBI
  29. Lamas Longarela O, Schmidt TT, Schöneweis K, Romeo R, Wedemeyer H, Urban S. Proteoglycans act as cellular hepatitis delta virus attachment receptors. PLoS One 2013;8:e58340 View Article PubMed/NCBI
  30. Abbas Z, Afzal R. Life cycle and pathogenesis of hepatitis D virus: A review. World J Hepatol 2013;5:666-675 View Article PubMed/NCBI
  31. Werzberger A, Mensch B, Kuter B, Brown L, Lewis J, Sitrin R. A controlled trial of a formalin-inactivated hepatitis A vaccine in healthy children. N Engl J Med 1992;327:453-457 View Article PubMed/NCBI
  32. Innis BL, Snitbhan R, Kunasol P, Laorakpongse T, Poopatanakool W, Kozik CA. Protection against hepatitis A by an inactivated vaccine. JAMA 1994;271:1328-1334 View Article PubMed/NCBI
  33. Midthun K, Ellerbeck E, Gershman K, Calandra G, Krah D, McCaughtry M. Safety and immunogenicity of a live attenuated hepatitis A virus vaccine in seronegative volunteers. J Infect Dis 1991;163:735-739 View Article PubMed/NCBI
  34. Gerlach JT, Diepolder HM, Zachoval R, Gruener NH, Jung MC, Ulsenheimer A. Acute hepatitis C: high rate of both spontaneous and treatment-induced viral clearance. Gastroenterology 2003;125:80-88 View Article PubMed/NCBI
  35. EASL Clinical Practice Guidelines: management of hepatitis C virus infection. J Hepatol 2011;55:245-264 View Article PubMed/NCBI
  36. Antaki N, Craxi A, Kamal S, Moucari R, Van der Merwe S, Haffar S. The neglected hepatitis C virus genotypes 4, 5 and 6: an international consensus report. Liver Int 2010;30:342-355 View Article PubMed/NCBI
  37. EASL recommendations on treatment of hepatitis C 2015.. J Hepatol 2015;63:199-236 View Article PubMed/NCBI
  38. Gerolami R, Borentain P, Raissouni F, Motte A, Solas C, Colson P. Treatment of severe acute hepatitis E by ribavirin. J Clin Virol 2011;52:60-62 View Article PubMed/NCBI
  39. Wedemeyer H, Pischke S, Manns MP. Pathogenesis and treatment of hepatitis e virus infection. Gastroenterology 2012;142:1388-1397 View Article PubMed/NCBI
  40. Kamar N, Abravanel F, Selves J, Garrouste C, Esposito L, Lavayssière L. Influence of immunosuppressive therapy on the natural history of genotype 3 hepatitis-E virus infection after organ transplantation. Transplantation 2010;89:353-360 View Article PubMed/NCBI
  41. Mallet V, Nicand E, Sultanik P, Chakvetadze C, Tessé S, Thervet E. Brief communication: case reports of ribavirin treatment for chronic hepatitis E. Ann Intern Med 2010;153:85-89 View Article PubMed/NCBI
  42. Kamar N, Rostaing L, Abravanel F, Garrouste C, Lhomme S, Esposito L. Ribavirin therapy inhibits viral replication on patients with chronic hepatitis e virus infection. Gastroenterology 2010;139:1612-1618 View Article PubMed/NCBI
  43. El Sayed Zaki M, El Razek MM, El Razek HM. Maternal-fetal hepatitis E transmission: is it underestimated?. J Clin Transl Hepatol 2014;2:117-123 View Article PubMed/NCBI
  44. Khuroo MS, Kamili S, Jameel S. Vertical transmission of hepatitis E virus. Lancet 1995;345:1025-1026 View Article PubMed/NCBI
  45. Patra S, Kumar A, Trivedi SS, Puri M, Sarin SK. Maternal and fetal outcomes in pregnant women with acute hepatitis E virus infection. Ann Intern Med 2007;147:28-33 View Article PubMed/NCBI
  46. Siebenlist U, Franzoso G, Brown K. Structure, regulation and function of NF-kappa B. Annu Rev Cell Biol 1994;10:405-455 View Article PubMed/NCBI
  47. Liang TJ. Hepatitis B: the virus and disease. Hepatology 2009;49:S13-S21 View Article PubMed/NCBI
  48. Dény P, Zoulim F. Hepatitis B virus: from diagnosis to treatment. Pathol Biol (Paris) 2010;58:245-253 View Article PubMed/NCBI
  49. Shiffman ML. Management of acute hepatitis B. Clin Liver Dis 2010;14:75-91 View Article PubMed/NCBI
  50. EASL clinical practice guidelines: Management of chronic hepatitis B virus infection. J Hepatol 2012;57:167-185 View Article PubMed/NCBI
  51. Terrault NA, Bzowej NH, Chang KM, Hwang JP, Jonas MM, Murad MH. AASLD guidelines for treatment of chronic hepatitis B. Hepatology 2016;63:261-283 View Article PubMed/NCBI
  52. Lau GK, Piratvisuth T, Luo KX, Marcellin P, Thongsawat S, Cooksley G. Peginterferon Alfa-2a, lamivudine, and the combination for HBeAg-positive chronic hepatitis B. N Engl J Med 2005;352:2682-2695 View Article PubMed/NCBI
  53. Marcellin P, Avila C, Wursthorn K, Chuang WL, Lau GK, Peng CY. Telbivudine (Ldt) Plus Peg-Interferon (Pegifn) in Hbeag-positive chronic hepatitis B - very potent antiviral efficacy but risk of peripheral neuropathy (Pn). J Hepatol 2010;52:S6-S7 View Article PubMed/NCBI
  54. Reijnders JG, Perquin MJ, Zhang N, Hansen BE, Janssen HL. Nucleos(t)ide analogues only induce temporary hepatitis B e antigen seroconversion in most patients with chronic hepatitis B. Gastroenterology 2010;139:491-498 View Article PubMed/NCBI
  55. Chang TT, Lai CL, Kew Yoon S, Lee SS, Coelho HS, Carrilho FJ. Entecavir treatment for up to 5 years in patients with hepatitis B e antigen-positive chronic hepatitis B. Hepatology 2010;51:422-430 View Article PubMed/NCBI
  56. Marcellin P, Buti M, Gane EJ, Krastev Z, Flisiak R, Germanidis G. Five years of treatment with tenofovir DF (TDF) for chronic hepatitis B (CHB) infection is associated with sustained viral suppression and significant regression of histological fibrosis and cirrhosis. Hepatology 2011;54:1011A-1012A View Article PubMed/NCBI
  57. Heathcote EJ, Marcellin P, Buti M, Gane E, De Man RA, Krastev Z. Three-year efficacy and safety of tenofovir disoproxil fumarate treatment for chronic hepatitis B. Gastroenterology 2011;140:132-143 View Article PubMed/NCBI
  58. Geier MR, Geier DA, Zahalsky AC. A review of hepatitis B vaccination. Expert Opin Drug Saf 2003;2:113-122 View Article PubMed/NCBI
  59. Schiff E, Lai CL, Hadziyannis S, Neuhaus P, Terrault N, Colombo M. Adefovir dipivoxil for wait-listed and post-liver transplantation patients with lamivudine-resistant hepatitis B: final long-term results. Liver Transpl 2007;13:349-360 View Article PubMed/NCBI
  60. Papatheodoridis GV, Cholongitas E, Archimandritis AJ, Burroughs AK. Current management of hepatitis B virus infection before and after liver transplantation. Liver Int 2009;29:1294-1305 View Article PubMed/NCBI
  61. Heidrich B, Manns MP, Wedemeyer H. Treatment options for hepatitis delta virus infection. Curr Infect Dis Rep 2013;15:31-38 View Article PubMed/NCBI
  62. Debing Y, Neyts J, Delang L. The future of antivirals: broad-spectrum inhibitors. Curr Opin Infect Dis 2015;28:596-602 View Article PubMed/NCBI
  63. Jordheim LP, Durantel D, Zoulim F, Dumontet C. Advances in the development of nucleoside and nucleotide analogues for cancer and viral diseases. Nat Rev Drug Discov 2013;12:447-464 View Article PubMed/NCBI
  64. Sluis-Cremer N, Tachedjian G. Mechanisms of inhibition of HIV replication by non-nucleoside reverse transcriptase inhibitors. Virus Res 2008;134:147-156 View Article PubMed/NCBI
  65. Gerber L, Welzel TM, Zeuzem S. New therapeutic strategies in HCV: polymerase inhibitors. Liver Int 2013;33:85-92 View Article PubMed/NCBI
  66. Lv Z, Chu Y, Wang Y. HIV protease inhibitors: a review of molecular selectivity and toxicity. HIV AIDS (Auckl) 2015;7:95-104 View Article PubMed/NCBI
  67. Patick AK, Binford SL, Brothers MA, Jackson RL, Ford CE, Diem MD. In vitro antiviral activity of AG7088, a potent inhibitor of human rhinovirus 3C protease. Antimicrob Agents Chemother 1999;43:2444-2450 View Article PubMed/NCBI
  68. Göthel SF, Marahiel MA. Peptidyl-prolyl cis-trans isomerases, a superfamily of ubiquitous folding catalysts. Cell Mol Life Sci 1999;55:423-436 View Article PubMed/NCBI
  69. Kaul A, Stauffer S, Berger C, Pertel T, Schmitt J, Kallis S. Essential role of cyclophilin A for hepatitis C virus replication and virus production and possible link to polyprotein cleavage kinetics. PLoS Pathog 2009;5:e1000546 View Article PubMed/NCBI
  70. Coelmont L, Hanoulle X, Chatterji U, Berger C, Snoeck J, Bobardt M. DEB025 (Alisporivir) inhibits hepatitis C virus replication by preventing a cyclophilin A induced cis-trans isomerisation in domain II of NS5A. PLoS One 2010;5:e13687 View Article PubMed/NCBI
  71. Gamble TR, Vajdos FF, Yoo S, Worthylake DK, Houseweart M, Sundquist WI. Crystal structure of human cyclophilin A bound to the amino-terminal domain of HIV-1 capsid. Cell 1996;87:1285-1294 View Article PubMed/NCBI
  72. Mazzon M, Mercer J. Lipid interactions during virus entry and infection. Cell Microbiol 2014;16:1493-1502 View Article PubMed/NCBI
  73. Lucas W. Viral capsids and envelopes: Structure and function. eLS 2010 View Article PubMed/NCBI
  74. Markowitz M, Zolopa A, Squires K, Ruane P, Coakley D, Kearney B. Phase I/II study of the pharmacokinetics, safety and antiretroviral activity of tenofovir alafenamide, a new prodrug of the HIV reverse transcriptase inhibitor tenofovir, in HIV-infected adults. J Antimicrob Chemother 2014;69:1362-1369 View Article PubMed/NCBI
  75. Murakami E, Wang T, Park Y, Hao J, Lepist EI, Babusis D. Implications of efficient hepatic delivery by tenofovir alafenamide (GS-7340) for hepatitis B virus therapy. Antimicrob Agents Chemother 2015;59:3563-3569 View Article PubMed/NCBI
  76. Chan HLY, Fung S, Seto WK, Chuang WL, Chen CY, Kim HJ. A phase 3 study of tenofovir alafenamide compared with tenofovir disoproxil fumarate in patients with HBeAg-positive, chronic hepatitis B: week 48 efficacy and safety results. J Hepatol 2016;64:S161 View Article PubMed/NCBI
  77. Buti M, Gane E, Seto WK, Chan HLY, Chuang WL, Stepanova T. A phase 3 study of tenofovir alafenamide compared with tenofovir disoproxil fumarate in patients with HBeAg-negative, chronic hepatitis B: week 48 efficacy and safety results. J Hepatol 2016;64:S135 View Article PubMed/NCBI
  78. Lai CL, Ahn SH, Lee KS, Um SH, Cho M, Yoon SK. Phase IIb multicentred randomised trial of besifovir (LB80380) versus entecavir in Asian patients with chronic hepatitis B. Gut 2014;63:996-1004 View Article PubMed/NCBI
  79. Yuen MF, Ahn SH, Lee KS, Um SH, Cho M, Yoon SK. Two-year treatment outcome of chronic hepatitis B infection treated with besifovir vs. entecavir: results from a multicentre study. J Hepatol 2015;62:526-532 View Article PubMed/NCBI
  80. Volz T, Allweiss L, Ben MBarek M, Warlich M, Lohse AW, Pollok JM. 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 View Article PubMed/NCBI
  81. Maepa MB, Roelofse I, Ely A, Arbuthnot P. Progress and prospects of anti-HBV gene therapy development. Int J Mol Sci 2015;16:17589-17610 View Article PubMed/NCBI
  82. Cradick TJ, Keck K, Bradshaw S, Jamieson AC, McCaffrey AP. Zinc-finger nucleases as a novel therapeutic strategy for targeting hepatitis B virus DNAs. Mol Ther 2010;18:947-954 View Article PubMed/NCBI
  83. Chen J, Zhang W, Lin J, Wang F, Wu M, Chen C. An efficient antiviral strategy for targeting hepatitis B virus genome using transcription activator-like effector nucleases. Mol Ther 2014;22:303-311 View Article PubMed/NCBI
  84. Makarova KS, Haft DH, Barrangou R, Brouns SJ, Charpentier E, Horvath P. Evolution and classification of the CRISPR-Cas systems. Nat Rev Microbiol 2011;9:467-477 View Article PubMed/NCBI
  85. Lin SR, Yang HC, Kuo YT, Liu CJ, Yang TY, Sung KC. The CRISPR/Cas9 system facilitates clearance of the intrahepatic HBV templates in vivo. Mol Ther Nucleic Acids 2014;3:e186 View Article PubMed/NCBI
  86. Cai D, Mills C, Yu W, Yan R, Aldrich CE, Saputelli JR. Identification of disubstituted sulfonamide compounds as specific inhibitors of hepatitis B virus covalently closed circular DNA formation. Antimicrob Agents Chemother 2012;56:4277-4288 View Article PubMed/NCBI
  87. 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 View Article PubMed/NCBI
  88. Wang P, Naduthambi D, Mosley RT, Niu C, Furman PA, Otto MJ. Phenylpropenamide derivatives: anti-hepatitis B virus activity of the Z isomer, SAR and the search for novel analogs. Bioorg Med Chem Lett 2011;21:4642-4647 View Article PubMed/NCBI
  89. Campagna MR, Liu F, Mao R, Mills C, Cai D, Guo F. Sulfamoylbenzamide derivatives inhibit the assembly of hepatitis B virus nucleocapsids. J Virol 2013;87:6931-6942 View Article PubMed/NCBI
  90. Wooddell CI, Rozema DB, Hossbach M, John M, Hamilton HL, Chu Q. Hepatocyte-targeted RNAi therapeutics for the treatment of chronic hepatitis B virus infection. Mol Ther 2013;21:973-985 View Article PubMed/NCBI
  91. Gish RG, Yuen MF, Chan HL, Given BD, Lai CL, Locarnini SA. Synthetic RNAi triggers and their use in chronic hepatitis B therapies with curative intent. Antiviral Res 2015;121:97-108 View Article PubMed/NCBI
  92. Noordeen F, Scougall CA, Grosse A, Qiao Q, Ajilian BB, Reaiche-Miller G. Therapeutic antiviral effect of the nucleic acid polymer REP 2055 against persistent duck hepatitis B virus infection. PLoS One 2015;10:e0140909 View Article PubMed/NCBI
  93. Hu Y, Cheng X, Cao F, Huang A, Tavis JE. Beta-Thujaplicinol inhibits hepatitis B virus replication by blocking the viral ribonuclease H activity. Antiviral Res 2013;99:221-229 View Article PubMed/NCBI
  94. Lu G, Lomonosova E, Cheng X, Moran EA, Meyers MJ, Le Grice SF. Hydroxylated tropolones inhibit hepatitis B virus replication by blocking viral ribonuclease H activity. Antimicrob Agents Chemother 2015;59:1070-1079 View Article PubMed/NCBI
  95. Franssen H, Leunissen J, Goldbach R, Lomonossoff G, Zimmern D. Homologous sequences in non-structural proteins from cowpea mosaic virus and picornaviruses. EMBO J 1984;3:855-861 View Article PubMed/NCBI
  96. Gonzalez ME, Carrasco L. Viroporins. FEBS Lett 2003;552:28-34 View Article PubMed/NCBI
  97. Shukla A, Dey D, Banerjee K, Nain A, Banerjee M. The C-terminal region of the non-structural protein 2B from Hepatitis A Virus demonstrates lipid-specific viroporin-like activity. Sci Rep 2015;5:15884 View Article PubMed/NCBI
  98. Scott C, Griffin S. Viroporins: structure, function and potential as antiviral targets. J Gen Virol 2015;96:2000-2027 View Article PubMed/NCBI
  99. OuYang B, Chou JJ. The minimalist architectures of viroporins and their therapeutic implications. Biochim Biophys Acta 2014;1838:1058-1067 View Article PubMed/NCBI
  100. Furuta A, Tsubuki M, Endoh M, Miyamoto T, Tanaka J, Salam KA. Identification of Hydroxyanthraquinones as novel inhibitors of hepatitis C virus NS3 helicase. Int J Mol Sci 2015;16:18439-18453 View Article PubMed/NCBI
  101. Shukla A, Padhi AK, Gomes J, Banerjee M. The VP4 peptide of hepatitis A virus ruptures membranes through formation of discrete pores. J Virol 2014;88:12409-12421 View Article PubMed/NCBI
  102. Liu S, Wei W, Shi K, Cao X, Zhou M, Liu Z. In vitro and in vivo anti-hepatitis B virus activities of the lignan niranthin isolated from Phyllanthus niruri L. J Ethnopharmacol 2014;155:1061-1067 View Article PubMed/NCBI
  103. Jiang X, Kanda T, Nakamoto S, Saito K, Nakamura M, Wu S. The JAK2 inhibitor AZD1480 inhibits hepatitis A virus replication in Huh7 cells. Biochem Biophys Res Commun 2015;458:908-912 View Article PubMed/NCBI
  104. Kanda T, Sasaki R, Nakamoto S, Haga Y, Nakamura M, Shirasawa H. The sirtuin inhibitor sirtinol inhibits hepatitis A virus (HAV) replication by inhibiting HAV internal ribosomal entry site activity. Biochem Biophys Res Commun 2015;466:567-571 View Article PubMed/NCBI
  105. Koh C, Canini L, Dahari H, Zhao X, Uprichard SL, Haynes-Williams V. Oral prenylation inhibition with lonafarnib in chronic hepatitis D infection: a proof-of-concept randomised, double-blind, placebo-controlled phase 2A trial. Lancet Infect Dis 2015;15:1167-1174 View Article PubMed/NCBI
  106. Lu JX, Sharpe S, Ghirlando R, Yau WM, Tycko R. Oligomerization state and supramolecular structure of the HIV-1 Vpu protein transmembrane segment in phospholipid bilayers. Protein Sci 2010;19:1877-1896 View Article PubMed/NCBI
  107. Atoom AM, Taylor NG, Russell RS. The elusive function of the hepatitis C virus p7 protein. Virology 2014;462–463:377-387 View Article PubMed/NCBI
  108. Pinto LH, Lamb RA. The M2 proton channels of influenza A and B viruses. J Biol Chem 2006;281:8997-9000 View Article PubMed/NCBI
  109. Ruch TR, Machamer CE. The coronavirus E protein: assembly and beyond. Viruses 2012;4:363-382 View Article PubMed/NCBI
  110. Lu W, Zheng BJ, Xu K, Schwarz W, Du L, Wong CK. Severe acute respiratory syndrome-associated coronavirus 3a protein forms an ion channel and modulates virus release. Proc Natl Acad Sci U S A 2006;103:12540-12545 View Article PubMed/NCBI
  111. Mehmel M, Rothermel M, Meckel T, Van Etten JL, Moroni A, Thiel G. Possible function for virus encoded K+ channel Kcv in the replication of chlorella virus PBCV-1. FEBS Lett 2003;552:7-11 View Article PubMed/NCBI
  112. Daniels R, Sadowicz D, Hebert DN. A very late viral protein triggers the lytic release of SV40. PLoS Pathog 2007;3:e98 View Article PubMed/NCBI
  113. van Kuppeveld FJ, Hoenderop JG, Smeets RL, Willems PH, Dijkman HB, Galama JM. Coxsackievirus protein 2B modifies endoplasmic reticulum membrane and plasma membrane permeability and facilitates virus release. EMBO J 1997;16:3519-3532 View Article PubMed/NCBI
  114. Martínez-Gil L, Bañó-Polo M, Redondo N, Sánchez-Martínez S, Nieva JL, Carrasco L. Membrane integration of poliovirus 2B viroporin. J Virol 2011;85:11315-11324 View Article PubMed/NCBI
  115. Melton JV, Ewart GD, Weir RC, Board PG, Lee E, Gage PW. Alphavirus 6K proteins form ion channels. J Biol Chem 2002;277:46923-46931 View Article PubMed/NCBI
  116. Piller SC, Ewart GD, Jans DA, Gage PW, Cox GB. The amino-terminal region of Vpr from human immunodeficiency virus type 1 forms ion channels and kills neurons. J Virol 1999;73:4230-4238 View Article PubMed/NCBI
  117. Kim JW, Lyi SM, Parrish CR, Parker JS. A proapoptotic peptide derived from reovirus outer capsid protein {micro}1 has membrane-destabilizing activity. J Virol 2011;85:1507-1516 View Article PubMed/NCBI
  118. Galloux M, Libersou S, Morellet N, Bouaziz S, Da Costa B, Ouldali M. Infectious bursal disease virus, a non-enveloped virus, possesses a capsid-associated peptide that deforms and perforates biological membranes. J Biol Chem 2007;282:20774-20784 View Article PubMed/NCBI
  • Journal of Clinical and Translational Hepatology
  • pISSN 2225-0719
  • eISSN 2310-8819
  • Copyright © 2022 JCTH. All Rights Reserved.
  • Published by Xia & He Publishing Inc.
  • Address: 14090 Southwest Freeway, Suite 300, Sugar Land, Texas 77478, USA
  • Email: service@xiahepublishing.com