• OPEN ACCESS

Novel Approaches to Inhibition of HBsAg Expression from cccDNA and Chromosomal Integrants: A Review

  • Ahmed H. Abdelwahed* ,
  • Brent D. Heineman and
  • George Y. Wu
 Author information
Journal of Clinical and Translational Hepatology 2023;11(7):1485-1497

DOI: 10.14218/JCTH.2023.00067

Abstract

Hepatitis B virus (HBV) is a widely prevalent liver infection that can cause acute or chronic hepatitis. Although current treatment modalities are highly effective in the suppression of viral levels, they cannot eliminate the virus or achieve definitive cure. This is a consequence of the complex nature of HBV-host interactions. Major challenges to achieving sustained viral suppression include the presence of a high viral burden from the HBV DNA and hepatitis B surface antigen (HBsAg), the presence of reservoirs for HBV replication and antigen production, and the HBV-impaired innate and adaptive immune response of the host. Those therapeutic methods include cell entry inhibitors, HBsAg inhibitors, gene editing approaches, immune-targeting therapies and direct inhibitors of covalently closed circular DNA (cccDNA). Novel approaches that target these key mechanisms are now being studied in preclinical and clinical phases. In this review article, we provide a comprehensive review on mechanisms by which HBV escapes elimination from current treatments, and highlight new agents to achieve a definitive HBV cure.

Keywords

Hepatitis B, Viruses, Liver disease, Hepatocytes, Gene editing

Introduction

Hepatitis B virus (HBV) is a common cause of acute disease, and chronic hepatitis that can progress to cirrhosis and hepatocellular carcinoma.1 The World Health Organization (WHO) estimated that in 2019, 296 million people were hepatitis B surface antigen (HBsAg)-positive. Nearly one million people die from HBV annually.2 HBV is transmitted through blood and bodily fluids in perinatal and sexual exposure.3,4 In adults, the vast majority, 80–85%, of acute infections resolve spontaneously. In contrast, in neonates and infants, 80–85% of acute infections result in chronic hepatitis. Treatment with direct-acting nucleotide analogs (DAAs) are generally highly effective in suppression of viral levels, but withdrawal of treatment in the vast majority of cases results in a return to pretreatment levels. Despite suppression of viral replication to low and even undetectable levels, the virus is able to restore levels of replication in the absence of antiviral agents. The aims of this report are to review the mechanisms by which HBV escapes DAA-mediated elimination, and to highlight strategies by which these mechanisms can be exploited in the design of novel agents against HBV and result in sustained virological response.

General principles of HBV replication

The major HBV infectious genome consists of a partially double-stranded relaxed circular DNA form enclosed in a capsid within a viral envelope. Infection involves primarily hepatocytes and is mediated by the binding of the hepatitis B surface antigen (HBsAg) to heparin sulfate proteoglycans on the surface of hepatocytes (Fig. 1).5 The virus then interacts with sodium taurocholate co-transporting polypeptide (NTCP), a functional receptor for HBV, allowing for viral internalization.6 Upon entry into hepatocytes, the relaxed circular DNA (rcDNA) in the nucleocapsid travels to the nucleus where its replication is completed by the conversion to covalently closed circular DNA (cccDNA). The latter forms a template for the transcription of viral RNAs.7 HBV pregenomic RNA (pgRNA) is transcribed from the cccDNA and is encapsidated by the hepatitis B core protein (HBc) in the cytoplasm.8 Reverse transcription of the RNA pregenome occurs in the cytoplasmic nucleocapsid beginning with binding of DNA polymerase (reverse transcriptase) to the pgRNA stem loop forming nucleocapsids. Within nucleocapsids, about 90% of the pgRNA is reverse transcribed to partially double stranded rcDNA. In the remaining 10%, pgRNA is reverse transcribed to double-stranded linear DNA (dslDNA). The dslDNA nucleocapsids can either be enveloped and secreted as new virions, re-enter the nucleus to add to the cccDNA pool, or integrate into the host cell genome. The rcDNA nucleocapsids can be either enveloped along with polymerase and secreted as new virions ready to begin a new infection or re-enter the nucleus and add to the cccDNA pool.9,10 Because of its overlapping reading frames, linearization of HBV circular DNA results in disruption of genes with loss of replicative capability. However, genes not affected by the linearization can continue to be expressed as integrants. Because cccDNA and integrated HBV DNA forms are stable and protected within the nucleus,11 they represent key targets for novel therapy for chronic HBV infections. Although linearization terminates HBV replication by integrants, multiple integrants in the same cell could provide complementary gene products in trans for complete viral replication. This can complicate attempts to inactivate HBV replication.12

Mechanism of hepatitis B virus entry and replication.
Fig. 1  Mechanism of hepatitis B virus entry and replication.

Hepatitis B virus (HBV) enters the hepatocyte following hepatitis B surface antigen (HBsAg) binding to heparin sulfate proteoglycans (HSPGs) and interaction with the sodium taurocholate co-transporting polypeptide (NTCP). Within hepatocytes, relaxed circular DNA (rcDNA) enters the nucleus where it is converted to covalently closed circular DNA (cccDNA), the transcription template for pregenomic RNA (pgRNA). Following transcription, pgRNA is encapsulated by the hepatitis B core protein within the hepatocyte cytoplasm. Reverse transcription of pgRNA occurs in the nucleocapsid, forming rcDNA or double-stranded linear DNA (dslDNA). These reverse transcription products are either enveloped and secreted as new virions, or re-enter the nucleus.

Shortcomings of current HBV treatment

The complex nature of HBV-host interaction is the main challenge for new treatment modalities. The major barriers include the presence of a high levels of HBV DNA and HBsAg, the presence of reservoirs for HBV replication and antigen production (cccDNA and integrated chromosomal DNA), and the HBV-impaired innate and adaptive immune response of the host.13

HBV inhibits the host immune response through several complex mechanisms including HBV regulatory X (HBx)-dependent downregulation of innate immunity signaling proteins, inhibition of interferon (IFN) type 1 (IFN-1) response, induction of immunosuppressive cytokines, or interference with toll-like receptor (TLR) activity. These mechanisms of immune tolerance have been reported to play a role in HBV chronicity.14 However, the role of innate immunity in HBV natural immune clearance is controversial. It has been suggested that natural killer (NK) cells may play a role in early infection clearance through the activation of cytokines like IFN-gamma. Other innate immune cells like monocytes could play both pro- and anti-inflammatory roles.15 Martinet et al.16 revealed that HBsAg is a potential key factor in the dysfunction of the plasmacytoid dendritic cells by altering their interaction with NK cells disrupting the cytolytic activity of NK cells and enhancing immune tolerance.

On the other hand, the adaptive immunity represented in HBV-specific antibody producing B lymphocytes and functional T cells (T-cytotoxic and T-helper) are most important in determining the HBV course of infection. HBV neutralizing antibodies have a role in prevention and modulation of chronic HBV while HBV-specific polyclonal CD8 T cells can lyse infected hepatocytes. They also secrete cytokines that induce the noncytolytic HBV clearance and recruit the inflammatory immune cells. HBV-specific CD4 T cells regulate these processes. The increased antigen burden in chronic HBV infection can functionally exhaust the T cells, causing loss of cytotoxicity, tumor necrosis factor-alpha and IFN-gamma production, and ultimately T cell deletion. Furthermore, co-inhibitory molecules involved in programmed cell death protein 1 (PD-1) are highly expressed by exhausted intrahepatic HBV-specific T cells which further decrease the host immune reaction against HBV infection.17

Objectives of novel HBV therapeutic strategies

The ultimate aim for HBV treatment is to induce HBsAg loss, prevent new hepatocyte infection, regain host immune function, and entirely eliminate HBV DNA. Table 1 highlights various strategies that aim to reduce HBsAg expression.18–65 As the elimination of cccDNA and integrated HBV DNA remain challenging, the ideal goal of chronic HBV treatment is to achieve functional cure with sustained undetectable levels of HBsAg and HBV DNA after a finite duration of treatment. A more practical goal would be partial cure with detectable HBsAg but with minimal HBV DNA. Patients who achieve a partial cure have better outcomes than patients with untreated viremia but with inferior prognosis to those with functional cure.66

Table 1

Novel approaches to inhibit hepatitis B surface antigen expression

Category (Refs)Agent/MechanismDesignAdvantagesDisadvantages
Screening of novel inhibitors18,19VariousVariousAbility to identify potential targets of cccDNA suppressionExact mechanisms are unclear; require future study
Direct cccDNA inhibitor20GLP-26, HBV capsid assembly modulatorIn vitro and humanized miceSustained inhibition of viral loadMice were not reconstituted with a humanized immune system in the model
HBV regulatory X protein2124Nitazoxanide;21 Dicoumarol;22 Protein-carrier HBx vaccine;23 HBx monoclonal antibody24VariousPromising elimination of HBsAg and or cccDNAStudies are in preclinical phases
DNA methylation2629Transcriptional suppression of HBV cccDNAcccDNA extracted from liver biopsies;27 Lentiviral vector inducing methylation in vitro29Sustained inhibition of viral loadUnclear whether shRNA methylation of HBV cccDNA would occur in non-neoplastic human liver model29
Histone acetylation25,3033Curcumin;25 Silent mating type information regulation 2 homolog 3 (SIRT3) ;31 Histone acetyltransferase 1 (HAT1);32 Np95/ICBP90-like RING finger protein (NIRF)33HepG2 cells;25 HepG2 cells;31 Human liver-chimeric mouse model;32 HepG2 cells and mice33Ability to modulate the HBV cccDNA minichromosome through various mechanismsMechanisms of modulation require further study
Apolipoprotein B mRNA editing catalytic polypeptide-like (APOBEC) protein family regulators3440APOBEC cytidine deaminasesIn vitro and in vivo studiesMultiple mechanisms by which inhibition can occurMechanisms require further study
Nivolumab in patients with chronic hepatitis B41Nivolumab programmed death receptor (PD-1) inhibitorClinical studyHBsAg decline in most patients and sustained HBsAg loss in one patient; well toleratedSmall pilot sample warranting further investigation
Toll-like receptor agonists42,43Vesatolimoid;42 Selgantolimod43Clinical studyClinical studies showing good safety and tolerance of agentsNo significant decline in HBsAg levels
Gene editingZinc finger agents44,45In vitroDemonstrated accurate localization of DNA target sites45Not all sequences are available for binding; Potential for off-target cutting
Gene editingTranscription activator-like effectors nucleases (TALENs)46,47In vitroHigh precision and can specifically target any DNA sequencePotential difficulty in vivo since a large number of amino acids are required to bind to a single nucleotide
Gene editingClustered regularly interspaced short palindromic repeats (CRISPR)/CRISPR-associated (Cas)4860In vitroDoes not require engineering of a site-specific nucleaseHBV genomes are highly heterogeneous, though this mechanism has been used to excise full-length HBV genomes from a stable HBV cell line
HBsAg inhibitors6163Nucleic acid polymers (NAPs);61 S-antigen traffic inhibiting oligonucleotide polymers (STOPS)62,63Randomized controlled trialsResults demonstrate HBsAg seroconversion and functional cure among patients61Concern for potential toxicity with STOPS62
Small molecule cccDNA inhibitor64Myrcludex B, inhibitor of HBV and HDV receptor sodium taurocholate co-transporting polypeptidePhase Ib/IIa clinical trialClinical trial, treatment was well toleratedHBsAg levels remained unchanged after 24 weeks of treatment
Small molecule cccDNA inhibitor65Bepirovirsen, antisense oligonucleotide that targets HBV mRNAsPhase IIb clinical trialClinical trial, treatment could show promise when incorporated with other combination therapiesHBV DNA loss only occurred in 9–10% of patients who received treatment for 24 weeks

Although highly effective, nucleos(t)ide analogs only block reverse transcription and do not directly act on the cccDNA.67 Additionally, the drugs do not prevent the formation of new cccDNA from incoming virions.67–70A phase III clinical trial comparing entecavir and lamivudine confirmed that short-term nucleos(t)ide analog therapy cannot eliminate hepatic cccDNA.71 Most individuals on nucleos(t)ide analogs must continue treatment indefinitely as the drugs only rarely result in long term HBsAg seroconversion.72

Novel cccDNA targets for sustained virological response against HBV

Studies are currently in progress to directly or indirectly inhibit cccDNA transcription/replication which will decrease the antigen burden of HBV. These strategies include inhibiting structures or targets involved in the formation of cccDNA, epigenetic modification including methylation and histone deacetylation that silence cccDNA transcription and HBV replication, improving host immune defense, and employing gene editing techniques to target and cleave cccDNA.

Direct cccDNA inhibitors

Some studies have identified potential targets that inhibit cccDNA directly. For example, Cai et al.18 utilized a cell-based screening strategy to measure cccDNA levels through expression of hepatitis B e antigen (HBeAg). Two disubstituted sulfonamide compounds called CCC-0975 and CCC-0346 reduced levels of cccDNA and rcDNA without directly affecting viral DNA replication. The results suggested that the disubstituted sulfonamide may interfere with the conversion of rcDNA to cccDNA through the inhibition of rcDNA deproteinization, a potentially intermediate step in cccDNA production, though the exact mechanism is unclear. If this result is confirmed, it may offer a new strategy for development of anti-cccDNA agents.

A screen of Chinese herbal remedies for HBV cccDNA inhibitors reported that hydrolyzable tannins, punicalagin, punicalin, and geraniin significantly reduced the production of HBeAg and cccDNA in a dose-dependent manner. It was proposed that the hydrolyzable tannins reduce cccDNA by blocking its formation and promoting its decay.19 However, the compounds failed to alter viral DNA replication. The evidence suggests that the observed effects of the herbal remedies are directed toward stability or degradation rather than production of cccDNA. These agents may have complementary effects when used in combination with inhibitors of cccDNA replication.

Recently, Amblard et al.20 identified GLP-26, a novel glyoxamide derivative, as a potential HBV inhibitor both in vitro and in humanized mice. GLP-26 is an HBV capsid assembly modulator that disrupts the HBV nucleocapsid and likely impacts the stability of cccDNA. GLP-26 inhibited HBeAg secretion and cccDNA amplification effectively. In the humanized mouse model of infection, it reduced HBsAg and HBeAg during and after treatment synergistically with entecavir. A strength of the study was inhibition of the viral load sustained for more than 12 weeks. GLP-26 had good oral bioavailability and did not show signs of mitochondrial toxicity as well when used at concentration less than 50 µm. A weakness of the study was that mice were not reconstituted with a humanized immune system in the model. As a special mouse model was employed, the results in a may not translate to similar results in humans.

There are problems in the evaluation of efficacy of anti-cccDNA agents. This is due in large part to a lack of standardized PCR-based methods that allow accurate cccDNA quantification in HBV-infected samples. For example, Southern blot hybridization is too insensitive, and real-time PCR for this purpose has not been standardized. Another problem is the presence of coexisting replicative intermediates which are identical in sequence to progenitor cccDNA. Furthermore, because cccDNA is located within hepatocytes, accurate quantification currently requires study of liver tissue which is an obvious clinical disadvantage.73 More research is needed to accurately validate and assess the efficacy of reliable and standardized quantification methods.

Targeting HBV regulatory X protein

Another potential target is the HBx protein which promotes the transcription of the viral genome.74 HBx assembles a damage-specific DNA-binding protein 1 (DDB1)-containing E3 ubiquitin ligase complex that targets the structural maintenance of chromosomes 5/6 (SMC-5/6), a complex that blocks viral transcription.74–77 HBx also up-regulates the degradation of apolipoprotein B mRNA editing catalytic polypeptide-like (APOBEC)3B which increases cccDNA.78 Shen et al.80 found that cccDNA specifically associated with an enhancer region on chromosome 19p13.11 that promotes activation of cccDNA transcription mediated by both HBx and a DNA-binding factor called Yin-Yang 1. It has been previously shown that mutations that prevent this interaction inhibit hepadnavirus infection.[21,77,80–72]

HBx-based therapeutics trials are only in preclinical phases. Sekiba et al.22 found that nitazoxanide (NTZ), an antiprotozoan agent, suppressed HBV transcription. Specifically, it decreased cccDNA levels and subsequent viral products through inhibition of the interaction between HBx and DDB1. The initial data suggested that NTZ was a promising HBV therapeutic agent and a potential tool to achieve genomic cure. The effects of NTZ against HBV in this study, while statistically significant, were modest. As NTZ is already used in clinical practice, approval for an HBV indication would likely be less costly and time consuming compared a totally new agent if further studies showed significant decrease in cccDNA.

Cheng et al.23 found that dicoumarol, an inhibitor for NADP(H): quinone oxidoreductase 1, destabilized HBx and blocked cccDNA transcription. Additionally, it decreased HBsAg, HBc protein, and HBV DNA levels in a humanized liver mouse model. The study demonstrated a prolonged sustained inhibitory effect on HBV cccDNA in vitro and in vivo. Although the study revealed that dicoumarol silenced cccDNA transcription, other mechanisms cannot be excluded as it independently acts as microtubule stabilizing agent and induces reactive oxygen species. However, it also carries a risk of uncontrolled bleeding. Treating chronic hepatitis B (CHB) patients who are already at high risk for bleeding complications with dicoumarol would be challenging. If research on dicumarol analogs reveals agents that retain cccDNA inhibitory effects without affecting coagulation, this class of agents might find better anti-HBV utility.

A protein-carrier HBx vaccine showed a significant elimination of HBsAg and HBV DNA by inducing a systemic CD4+ and CD8+ T cell response in HBV carrier mice.24 The study demonstrated that HBx-induced adaptive immunity eliminated HBV expressing cells. However, no signs of protective antiHBs were observed indicating that treatment with the HBx vaccine alone is not sufficient to restore the humoral immunity. Another problem is that human immune response to HBx vaccine differs from that of rodents, which are not natural hosts for HBV.

Another study demonstrated HBx monoclonal antibody as a potential potent therapeutic agent for HBV. A cell-penetrating antibody targeting HBx was developed by conjugation of HBx antibody and an HIV tat protein. The tat monoclonal antibody suppressed viral replication and protein production in cell and mice models that mimicked chronic HBV infection.83 Although significant decreases in HBsAg and HBV DNA levels were observed in mice after a single infusion, virological rebound in serum occurred in most mice between days 7 and 9. More studies are needed to improve cell delivery, efficacy and sustainability to promote HBx monoclonal antibodies as a therapeutic agent.

Epigenetic modification to reduce cccDNA expression

DNA methylation

Epigenetic modifications through DNA methylation and histone deacetylation affect the function of cccDNA.25,84,85 CpG methylation regulates the transcription of cccDNA.86 Increased cccDNA methylation is associated with low serum HBV DNA-titer, suggesting suppressed viral activity.26 Additionally, the methylation of cccDNA in human tissue of CHB patients has been associated with reduced cccDNA mRNA synthesis and viral expression.27–29 For example, a study on HBV cccDNA extracted from liver biopsies of HBsAg-positive patients found that HBeAg-negative patients had significantly higher positive ratios of cccDNA methylation than those of HBeAg-positive patients.28 The study showed that methylation of cccDNA is associated with impaired replication of HBV which could result in silencing and ultimately depletion of HBV cccDNA. The study demonstrated that cccDNA may be relevant to HBeAg seroconversion. However, it failed to identify a mechanism of epigenetic modulation.

Another study by Park et al.30 developed a third generation lentiviral vector through cloning of a short hairpin RNA (referred to as shRNA) sequence against the HBx gene into an HIV-based plasmid. It induced methylation and transcriptional suppression of HBV cccDNA in a hepatoma cell line. The study showed that lentiviral vector-mediated of shRNA may be a potential novel tool for suppression and potentially elimination of cccDNA through epigenetic modulation. However, the study did not demonstrate suppression activity in nonneoplastic liver cells and nor demonstrate prolonged suppression of cccDNA. Safety of lentiviral integration would be further required for clinical studies.

Histone acetylation

Acetylated histones bound to cccDNA have been shown to regulate the propagation of HBV.84 Hyperacetylation of H3 and H4 histones increased transcription of cccDNA and HBV replication.25 Since HBx can bind to the cccDNA mini-chromosome to promote acetylation and replication of the virus, researchers have identified various targets that help down-regulate histone acetylation.

In a HepG2.2.15 cell line transfected with HBV, Wei et al.31 determined that curcumin, a hypomethylating agent, caused a dose-dependent reduction in HBsAg and HBeAg expression and significant reduction in HBV DNA replication through decreased cccDNA-bound histone acetylation. However, cytotoxicity occurred with concentrations of more than 20 μmol/L. Ren et al.32 demonstrated the effect of silent mating type information regulation 2 homolog 3 (SIRT3), a NAD-1 dependent histone deacetylase, on HBV replication. Ectopic SIRT3 overexpression inhibited HBV replication and transcription by acting cooperatively with methyl transferase to restrict HBV cccDNA. In contrast, gene silencing of SIRT3 increased HBV activity in primary human hepatocytes and HBV-infected HepG2-NA1/taurocholate co-transporting polypeptide cells. However, the reduction in cccDNA in HBV-infected cells did not reach a statically significant difference. Animal studies are needed to more fully evaluate efficacy and toxicity.

Yang et al.33 demonstrated that histone acetyltransferase 1 (HAT1), an important factor in chromatin assembly, regulated the cccDNA mini-chromosome. The authors first demonstrated that HAT1 expression in a human liver-chimeric mouse model increased in HBV-infected humanized mice. They also developed cell lines that could detect and quantify HBV cccDNA and HBV DNA. They found that depletion of HAT1 significantly decreased HBV DNA, HBeAg, and HBsAg in HBV-infected primary human hepatocytes, dHepaRG, and HepG2-NTCP cells. Additionally, qPCR and Southern blot analysis demonstrated that HAT1 was crucial for cccDNA accumulation. They also found that infected nontumorous liver had a higher expression of HAT1 indicating a relationship between the molecule and HBV cccDNA. A strength of the study was demonstration that HAT1 promoted HBV replication and cccDNA accumulation. The authors also demonstrated that HBV upregulated HAT1 to enhance its replication in a positive feedback manner. Weaknesses of the study included a relatively small sample size, 43 liver tissue samples, of which only 39 were positive for HBV DNA and only 24 positive for cccDNA. While the depletion of HAT1, HBV cccDNA and HBV DNA is of interest, the clinical impact of the finding remains unclear.

Qian et al.34 found that Np95/ICBP90-like RING finger protein (commonly known as NIRF), an E3 ubiquitin ligase, reduced the acetylation of HBV cccDNA-bound H3 histones, and inhibited the replication and secretion of HBV through proteasome degradation of HBc proteins. That led to a decreased burden of HBsAg and HBeAg. A strength of the study was the inhibition of replication and viral antigens in vivo and in vitro. A lack of sustained significant inhibition of HBeAg, HBsAg, HBV DNA, and HBV cccDNA after 72 h in culture cells injected with NIRF compared to the control cells in vitro tempers optimism.

Targeting the immune system

APOBEC

APOBECs make up a family of endogenous cytidine deaminases that initiate the destruction of cccDNA in the nucleus.35–37 They have been shown to inhibit HBV replication through deaminase-dependent and independent mechanisms.36,38,39 APOBEC3G can bind to the HBc and gain access to DNA during reverse transcription, editing the core associated DNA but not the pgDNA.40 Furthermore, Lucifora et al.36 demonstrated that activation of lymphotoxin (LT) β receptor (LTβR) suppresses HBV replication and leads to nuclear cccDNA degradation through upregulation of APOBEC3A and APOBEC3B cytidine deaminase in vitro. The study demonstrated a persistent antiviral effect with no rebound in HBV replication. A problem with constitutive expression of LTβR is its association with hepatocellular carcinoma and liver inflammation which make its clinical use as an antiviral challenging.

Various tumor necrosis factor superfamily members are physiological ligands for this receptor and can activate inflammatory, anti-inflammatory survival pathways or induce apoptosis.36–41

Nivolumab

As explained earlier, patients with chronic HBV infection have impaired immune response to HBV. This is partially due to chronic exposure of HBsAg. Several approaches to stimulate or decrease inhibition of HBV-specific immune responses have been studied. Gane et al.42 administrated nivolumab, a programmed death receptor inhibitor specifically targeting PD-1, with or without GS-4774, a yeast-based therapeutic T cell vaccine, to chronic HBeAg-negative HBV patients. The study showed that three of 22 patients who received high dose nivolumab had significant decreases in HBsAg levels. However, only one patient had undetectable levels of HBsAg at week 20 which was sustained for 12 months. A strength of this study was evidence that immune checkpoint inhibition can improve the immune response in chronic HBV infection. However, one patient developed an acute flare of alanine aminotransferase. Another weakness was the exclusion of HBeAg-positive patients, a bias toward inclusion of low levels of HBV infection. More studies are needed to assess the safety and efficacy of immune checkpoint inhibition on a larger scale.

TLR agonists

There have been several studies on induction of innate immune response in CHB patients. Vesatolimoid (GS-9620), an oral agonist of TLR7, was studied in 162 HBV patients. Although safe and well tolerated, the study failed to show any significant decline in the levels of HBsAg.43

Gane et al.87 evaluated the efficacy of selgantolimod, a TLR8 agonist, in viremic CHB patients. Doses of selgantolimod for two or four weeks did not show a significant decline from baseline in HBsAg or HBV DNA. However, study showed good safety and tolerability profile. More research is required to more fully evaluate efficacy and toxicity of these agents.

Gene editing approaches

It is established that double-stranded breaks (DSBs) stimulate cellular endogenous repair machinery.88 DNA is typically repaired through two major pathways, nonhomologous end joining (NHEJ) or homology directed breaks. NHEJ results in direct re-ligation of the two ends of the DSB and does not require a DNA template. This may introduce or remove a few nucleotides causing frameshift mutations that could produce truncated proteins or degradation of the mRNA. Homology directed breaks is more complex and functions to repair DSB in a DNA-template-dependent manner. Both pathways are error-prone and can be used to control the DNA repair machinery to engineer a wide variety of genomic alterations.89,90 Various gene therapeutic approaches have been studied to treat HBV infection through introducing site-specific DSBs in HBV cccDNA. Four major mechanisms have been described: zinc finger nuclease (ZFN), transcription activator-like effector nucleases (TALENs), clustered regularly interspaced short palindromic repeats (CRISPR)/CRISPR-associated (Cas) systems have targeted HBV cccDNA, and meganucleases.91

Zinc finger agents

Zinc fingers (ZFs) are multifunctional mammalian proteins that naturally serve as transcription factors. The DNA-binding domain (DBD) of ZF attaches to a certain base triplet. The DBD is connected to the cleavage domain of the FoKL restriction endonuclease. DBD proteins provide the targeting domains while FoKL restriction endonuclease results in cleavage, double-stranded DNA breaks and potential gene knockout through the DNA repair process (Fig. 2).89,91–93 ZF agent site selection is limited by the fact that not all sequences are available for binding by ZF proteins. Another challenge is off-target cutting by a pair of ZFNs, one that binds the forward strand and another that binds the reverse strand, can double the number of recognized base pairs.44

Gene editing mechanisms of zinc finger nucleases and transcription activator-like effector nucleases.
Fig. 2  Gene editing mechanisms of zinc finger nucleases and transcription activator-like effector nucleases.

A. Zinc finger nucleases (ZNFs) recognize and bind to target nucleotide triplets. Transcription activator-like effector nucleases (TALENs) operate in a similar mechanism but can directly cleave specific DNA sequences. The nucleases create a double-stranded break (DSB) around the splice target. Nonhomologous end joining (NHEJ) repair causes gene knockout, deletion, correction, or addition. cccDNA, covalently closed circular DNA.

Epigenetic gene silencing is performed by targeting locus-specific, epigenetic changes in effector or repressor domains to inhibit HBV gene expression. cccDNA epigenome editors are currently being developed. In a Hep3B HCC cell line, Singh et al.45 downregulated integrated HBx using an artificial transcription factor (ATF). They created a ZF domain that specifically targeted an 18-bp DNA target in the enhancer region of HBx. In cell lines that expressed HBx, ATF resulted in significant growth arrest. However, the variation of HBV DNA sequence in different patients presents a challenging barrier, as ATF would need to be tailored to individual HBV sequences. Animal studies are needed to more fully evaluate efficacy and toxicity.

Weber et al.94 created three ZFNs to target cccDNA in human embryonic kidney 293 T cells and HepAD238 cells with an encoded adeno-associated viral vector. Through open frame reading, the ZFNs targeted the polymerase/x (1), polymerase/core (2) and polymerase regions (3) to inflict mutations in the DNA. ZFN pair three showed a significant antiviral activity and decreased HBV DNA production.

Without the use of nucleases, Zimmerman et al.95 used ZF proteins to target the cccDNA in duck hepatitis B virus (DHBV). They avoided off-binding by selecting sites based on BLAST searches of the DNA target sites against chicken genome. They screened binding efficiency through a gel shift assay (i.e. EMSA) and DHBV enhancers which inhibited the core, small, and large surface protein production were introduced into a DHBV culture system. They concluded that expression of designed ZF proteins in DHBV culture system resulted in decreased viral RNA and protein expression. Strengths of the study included localization of DNA target sites accurately, and demonstration of a lack of off-binding using Western blotting. A disadvantage is that the authors did not demonstrate the effect of ZF proteins in vivo. Studies in humanized models may be helpful in more fully evaluating efficacy and toxicity.

TALENs

TALENs are DNA-binding proteins that are produced by xanthomona bacteria.46 Similar to ZFNs, TALEs can be fused to the catalytic domain of FoKL nuclease to produce TALENs. TALENs can specifically target any DNA sequence and are easy to produce (Fig. 2). However, a large number, 34, of amino acids required to bind to a single nucleotide making them potentially difficult to be delivered in vivo.47

Bloom et al.96 demonstrated the first targeted nuclease-mediated disruption of HBV cccDNA using TALENs in cell culture and in mice. They generated four TALENS which targeted specific sites within the S/pol (S-TALEN), C/pol (C-TALEN), and pol ORFs (P-TALEN), of the HBV genome. In Huh-7 cells, there was a significant decrease in the hepatitis B core antigen (HBcAg) in transfected cells. S-TALENs showed increasingly inhibitory effects on HBcAg in a hypothermic environment. The targeted cccDNA was isolated from HepG2.2.15 cells to assess for the TALEN-mediated targeted mutagenesis. PCR-based analysis and primer amplification methods showed no significant contamination with cellular DNA genome or HBV rcDNA. Assays using T7E1 and CEL1 showed that the S-TALEN disrupted 31% of the target molecules that correlated with a decrease in HBcAg secretion in HEPG2.2.15 cells. C-TALEN and P-TALEN showed no significant effect in disruption of HBV DNA.

A hydrodynamic injection method was employed to assess the effect of S- and C-TALENs on HBV replication measuring HBsAg concentration and viral particles equivalents in mice. More than 90% of HBcAg was knocked down by TALEN S and 70% of viral particles equivalents were decreased by either S-TALEN or C-TALEN The immune-histological assessment and transaminases levels demonstrated minimal toxicity. T7E1 assays demonstrated mutations of 57–87% of amplified HBV DNA in treated mice. Study strengths included demonstrating a significant decrease in HBcAg with a minimal toxicity on liver cells. However, the study failed to demonstrate any significant effect of TALENS on transcription of HBV. mRNA concentrations were similar in both treated mice and controls.96 Confirmation of these results without using hydrodynamic methods for introduction of the agents may show the potential under conditions closer to clinical applications.

Chen et al.97 designed TALENs to target highly conserved regions among the different genotypes of HBV. L1/R1 TALEN recognized the region around the RNase H (one of the four domains of viral polymerase), while L2/R2 and L3/R3 recognized DNA sequences in the core protein regions. Huh-7 cells were transfected with HBV DNA and plasmid encoding TALENS. Their expression was detected mainly in the nucleus using immunoblotting. CCK8 assays showed no difference in cell viability and cell growth between the control and cells targeted with TALENs. L1/RI and L2/R2 TALENs were able to significantly decrease the HBsAg, HBeAb, and pgRNA production, but only L2/R2 drastically decreased the levels of HBcAg. cccDNA was decreased by 10–20% and 30–40% in L1/R1 and L2/R2, respectively, in TALENs-expressing cells. The authors confirmed their results in a hydrodynamic mouse model with L2/R2 infected cells. Significant decreases in HBsAg, HBeAb, HBV DNA, and pgRNA levels were found. Moreover, they demonstrated a synergistic inhibitory effect by TALENs and INF-alpha on the HBV transcription. A strength of this study was the demonstration of significant decreases in both viral RNA production and transcription across HBV genome with different genotypes (B, C, or D). A weakness of the study is that TALEN L3/R3 failed to show an effect against transcription or replication. The variation in effects between different TALENs suggest that efficacy depended on the target sequence of HBV DNA.

CRISPR/Cas-9

The CRSIPR/Cas-9 system utilizes a short RNA sequence that drives the formation of a DSB at a target site (Fig. 3). It only requires the synthesis of new RNA rather than the engineering of a site-specific nuclease. Various factors influence the efficacy of the CRISPR/Cas-9 system including guide RNA design, off-target cutting, Cas-9 activity, and the method of delivery.98 One challenge of targeting HBV cccDNA using CRISPR/Cas-9 is that HBV genomes are highly heterogeneous.

Clustered regularly interspaced short palindromic repeats/CRISPR-associated gene editing mechanism.
Fig. 3  Clustered regularly interspaced short palindromic repeats/CRISPR-associated gene editing mechanism.

Clustered regularly interspaced short palindromic repeats (CRISPR) often utilizes a CRISPR-associated (Cas)-9 nuclease that locates a single guide RNA (sgRNA) and makes a double-strand break, cleaving DNA targets. These breaks trigger DNA repair which can knockout genes, as demonstrated by experiments involving CRISPR/Cas-9 and covalently closed circular DNA (cccDNA).

CRISPR/Cas-9 has been shown to inhibit HBV infections by introducing mutations into the viral cccDNA. Combinations of HBV-targeting nucleases cleaved DNA strands repaired by NHEJ, an error-prone repair mechanism.48 Seeger and Sohn found that the CRISPR/Cas 9 system was extremely efficient at editing of HBV DNA in HepG2 cells. Using next generation sequencing (NGS), the authors showed a spectrum of mutations in cccDNA following Cas-9 cleavage and repair following NHEJ. Cas-9 cleaved over 90% of HBV DNA, and was 10,000 to 15,000 times more efficient than APOBEC-mediated cytosine deamination following the treatment of infected cells with IFNα. The authors designed single guide RNAs (sgRNAs) called HBx2 and HBx4 that targeted HBx and the overlapping polymerase gene. They also found that approximately 7% of cleaved cccDNA genomes were repaired in a way that might not terminate the function of HBx. The need to use multiple sgRNAs targeting different loci on the HBV genome to inactive cccDNA was recognized.49

Kennedy et al.50 used the CRISPR/Cas system to target HBV cccDNA, resulting in decreases in total HBV, cccDNA, and the secretion of HBV antigens including HBsAg. Experimental systems have included de novo infection of hepatoma cell lines with HBV produced by HepAD38 cells,50–58 transfection of plasmids that express HBV into hepatoma cell lines and in vivo co-transfection of HBV DNA and CRISPR/Cas expression plasmids in mouse livers.51,54,56,58,59 Additionally, CRISPR/Cas-9 technology has been used to excise full-length integrated HBV genomes from a stable HBV cell line. Post excision, cccDNA, supernatant HBV DNA, or HBsAg was undetectable for 10 months.60 However, many of these studies did not consider the high heterogeneity of the HBV genome when identifying and selecting guide RNA (gRNA) targets.49,50,54,59

Li et al.99 pyrosequenced the whole HBV genome in 17 patients with advanced liver disease and 30 chronic carriers and found a variety of viral quasispecies, demonstrating heterogeneity. Liu et al.55 designed eight gRNAs to target 26 conserved regions of different HBV genotypes. The gRNA/Cas-9 systems were co-transfected into HepG2 cells with HBV, and the levels of HBV RNA were detected by northern blotting. Four days after transfection, it was found that all systems significantly suppressed HBV replication compared to controls. CCK8 assays demonstrated no cytotoxicity. Two representative gRNA/Cas-9 systems were amplified and sequenced and found not to have detectable off-target effects. Additionally, the application of multiple gRNA/Cas-9 systems significantly inhibited the replication of HBV all the genotypes used, suggesting that using several gRNAs to simultaneously target conserved regions of HBV may increase the ability to inactivate HBV replication and prevent viral escape from treatment. Moreover, the authors demonstrated inhibition of HBV in vivo by injecting mice with the HBV-specific gRNA/Cas-9 systems and HBV. Assays and sequencing showed that these systems could introduce approximately 11% mutagenesis in vivo. A weakness of this study is that only the human codon-optimized SpCas9 plasmid was used which only covered a portion of the HBV genotypes and subgenotypes even though the gRNA/Cas-9 systems targeted conserved regions.

To show direct cccDNA targeting, Martinez et al.61 used HBV-specific gRNAs and CRISPR/CAS-9 to determine the effect on cccDNA after gene editing in HBV-infected HepG2-NTCP cells. The gRNA/CAS-9 was delivered using ribonucleoproteins. In several single and dual combinations, persistent reduction in RNA, HBsAg and HBeAg levels were observed most significantly in dual combination with targeting Sp5 and Sp7. The study demonstrated the formation of new small transcriptionally active HBV variants. The study pointed out the presence of episomal HBV DNA variants after the cleavage by CRISPR/CAS-9.

The use of nuclease-based therapy in HBV therapy faces several obstacles including safety, efficacy, and specificity. There is potential toxicity associated with gene targeting as nucleases may be considered as foreign bodies by immune system.89 In addition, DSBs could result in aberrant chromosomal rearrangements with unexpected adverse effects. Targeted and efficient delivery remains a major challenge as gene editing components are relatively large in size.90 Various delivery methods are being explored including the use of viruses such adenoviruses or adeno-associated viruses and nonviral methods such as lipid-based nano-formulations.91 Another challenge is the high sequence heterogeneity within HBV genotypes which make it difficult to find effective gRNAs to target conserved HBV sequences across different genotypes.89–91

Novel inhibitors of HBsAg

Figure 4 shows various novel mechanisms of agents for inhibition of HBsAg synthesis and expression. Targeting integrated HBV DNA could help eliminate an important source of HBsAg expression which is partially responsible for immune tolerance and T cell exhaustion.17

Mechanisms of novel methods to interfere with hepatitis B surface antigen synthesis (HBsAg).
Fig. 4  Mechanisms of novel methods to interfere with hepatitis B surface antigen synthesis (HBsAg).

(A) Nucleic acid polymers (NAPs), which interfere with the production of subviral particles that carry HBsAg; (B) S-antigen traffic inhibiting oligonucleotide polymers (STOPS), which inhibit expression of HBsAg; (C) Myrcludex B, a small molecule cccDNA inhibitor that targets the sodium taurocholate co-transporting polypeptide (NTCP); and, (D) Bepirovirsen, an antisense oligonucleotide that targets Hepatitis B virus (HBV) mRNA.

Nucleic acid polymers

Nucleic acid polymers (NAPs) are oligonucleotide-based, broad-spectrum antiviral agents. They act on the apolipoprotein interaction involved in the assembly and release of HBV subviral particles which are made of HBsAg. The elimination of HBsAg can improve the efficacy of the immune therapy and potentially achieve functional cure.62

Bazinet et al.63 studied two NAPs, REP 2165 and REP 2139 combined with pegylated (Peg) IFN and tenofovir disoproxil fumarate in HBeAg-negative CHB patients. Patients were randomly assigned to combination therapy (with REP 2165 or 2139) or only TNF and Peg-IFN. The study demonstrated a significant decrease in HBsAg levels in the experimental group. Furthermore, 60% of patients who received the combination therapy had HBsAg seroconversion and 39% achieved functional cure. Levels of HBsAg did not differ significantly between patients who received REP 2165 or REP 2139. There was a greater elevation in the levels of transaminases in the experimental group, and was associated with symptoms. The levels correlated with the initial increase in HBsAg, and normalized during therapy. The study did not demonstrate the mechanism of NAPs in suppressing HBsAg. Evidence of hepatotoxicity as reflected by aminotransferase elevations is a concern. It is possible that other congeners that have similar anti-HBV effects without hepatotoxicity may be developed.

S-antigen traffic inhibiting oligonucleotide polymers

S-antigen traffic inhibiting oligonucleotide polymers (STOPS) are a class of oligonucleotides that are similar to NAPs. They can potentially inhibit HBsAg synthesis through the sequestration of cellular proteins that are required for proper expression and folding of HBsAg. A challenge of using STOPS in clinical practice is potential toxicity.64 Currently, the safety and efficacy of the first STOP, ALG-010133, are being studied in preclinical phases.65

Viral entry inhibitors

Myrcludex B

Myrcludex B is a linear, chemically synthesized, myristoylated peptide. It acts as a specific entry inhibitor of the HBV and hepatitis D virus (HDV) receptor NTCP and ultimately blocks entry of HBV and HDV into hepatocytes (Fig. 4). In addition, because bile acids themselves have been shown to promote HBV transcription and gene expression, myrcludex B was shown to inhibit HBV replication on a transcriptional level at a post entry step by interfering with the farnesoid X receptor alpha. In hepatoma cell lines with stable NTCP expression, myrcludex B significantly reduced the levels of HBsAg in cell cultures. The results were consistent in mouse studies in which HBsAg, HBeAg, and HBV DNA were all significantly decreased by myrcludex B adminstration.100 A limiting factor was that the effect was only seen after 5 days of treatment which indicates that only sustained treatment may impair replication. The authors did not comment on the effect of the myrcludex on the physiological function as a bile acid transporter and its importance in bile acid homeostasis in liver cells which might be challenge for clinical use of myrcludex B. The study is important because it shows the role of myrcludex B in inhibiting HBV replication at a post entry level.

A clinical trial compared daily myrcludex B with entecavir administration in patients with CHB. One cohort received myrcludex B daily for 24 weeks followed by Peg-IFNα-2a for 48 weeks, another cohort received myrcludex B daily in combination with Peg-IFNα-2a weekly for 24 weeks followed by 24 weeks of Peg-IFNα-2a alone, and the last cohort received only Peg-IFNα-2a weekly. The primary endpoint was HBsAg response at week 12 of therapy. Secondary endpoints included the responses of HBsAg at 24 weeks and those of HDV RNA, HBV DNA, and ALT to therapy at 24 and 48 weeks, as well as at the end of a 24-week treatment-free follow up period. The inhibitor caused a significant decrease in HDV in chronic hepatitis D patients after 24 weeks of treatment. However, HBsAg levels remained unchanged. In a cohort receiving daily myrcludex B in combination with weekly Peg-IFNα for 24 weeks, HBD DNA decreased significantly at the end of the study. A strength of this study was that the monotherapy myrcludex B was well tolerated and its combination with Peg-IFNα did not increase frequency or severity of adverse events.101 A weakness of this clinical trial was the small sample size. Although the study reported that the agents were well tolerated, if IFN is required for optimal effects of myrcludex B, patient acceptance of IFN side effects could be a practical clinical issue.

In mouse reconstituted human hepatocytes that were infected with HBV, Volz et al.102 showed that daily administration of myrcludex B inhibited viral spreading from the initially infected human cells demonstrated by decreased HBcAg levels after 6 weeks of infection. Furthermore, intrahepatic cccDNA loads remained stable compared to the values found in mice sacrificed 3 weeks post-infection. A study strength is the demonstration of an inhibitory effect of myrcludex B on the intrahepatic cccDNA pool. It was suggested that myrcludex B might hinder conversion of rcDNA to cccDNA, the mechanism by which this occurs was not clarified.

Small molecule cccDNA inhibitors

Bepirovirsen

Yeun et al.103 assessed the safety and efficacy of bepirovirsen (GSK3228836), an antisense oligonucleotide. Bepirovirsen targets HBV mRNA and may have an immune stimulatory activity mediated by TLR8. The study included 475 patients, half receiving nucleotide analogue therapy. Sustained HBsAg and HBV DNA loss occurred in 9–10% of patients who received bepirovirsen for 24 weeks. Adverse effects occurred more in the experimental group than the placebo group, and included pyrexia, fever, and elevated liver aminotransferases. The loss of HBsAg that was observed predominantly in HBeAg-negative patients who potentially have integrated HBV sequences in their genome. This suggested but did not prove that bepirovirsen targeted integrated HBV DNA. There was no apparent response in HBsAg-positive patients who were not on nucleotide analoguetreatment. That observation might be related to the increased baseline levels of HBsAg and its role in prediction the response to HBV therapies. The study showed a relative low efficacy of bepirovirsen. However, there is a potential for enhancement with other combination therapies and more selective baseline characteristics of the patients. Currently, the durability of response is under investigation.

Discussion

Complete eradication of cccDNA and ultimately complete cure of HBV infection remains elusive with many challenges ahead. One of the main challenges is the need for efficient targeted delivery that could decrease degradation of the agent by untimely elimination, and avoid side effects and toxicity to nonhepatic cells at the same time. Polymeric nanoparticles and lipid nanoparticles have been shown to improve inhibition of viral replication and clearance of cccDNA. The understanding of the tissue and intracellular environments has enabled the utilization of receptor specific ligands in the design of the nanocarrier systems.104 Several examples of targeted delivery to hepatocytes have been described105 Receptor mediated-delivery with high affinity ligands may have advantages of specificity if the receptors are cell-specific. Any nonspecific uptake by scavenger receptors such as those present on Kupffer cells may decrease the efficiency of delievery.105,106

The most commonly used viral vectors are retroviruses, adenoviruses, and adeno-associated viruses. Although many studies approached increasing the transfection efficiency of viral vectors, there are many concerns regarding their cytotoxicity to hepatocytes and potential inflammatory response.105 Hydrodynamic-based delivery has been used in small animals but the rapid injection large fluid volumes is likely to increase the risk serious adverse effects making that technique unlikely to become a clinical application.107 Despite numerous developments, novel targeted drugs delivery methods remain in early phases.108

To treat HBV-infected patients with agents that affect cccDNA, the efficacy of such novel anti-HBV agents can only be monitored by accurate quantitation of cccDNA levels. However, such measurements currently present a major technical challenge.73 Obstacles include the current need for liver tissue, the lack of standardized PCR-methods, and the presence of coexisting identical replicative intermediates to cccDNA. More studies are needed to develop more sensitive techniques to overcome the small size liver samples, low levels of viral DNA during therapy and the need to investigate the number of infected hepatocytes harboring cccDNA in the liver.109,110 Therefore, there is an urgent need for a reliable and convenient standardized cccDNA assay. Tu et al.111 developed a new assay called cccDNA inversion quantitative PCR which showed high precision and sensitivity. However, that study did not demonstrate that cccDNA inversion quantitative PCR could be used to assay clinical isolates from patient samples.

Another challenge of gene editing approaches is the potential for off-target effects resulting in toxicity. There are multiple mechanisms under investigation to reduce off-target effects. For example, for CRISPR/Cas-9, improvement, and engineering Cas-9 variants as suitable sgRNAs, finding new versions of gene editors that do not induce DSBs, and improving methods to deliver Cas-9/sgRNA into target cells have been developed.112 Similar strategies can be developed for novel HBsAg treatments through gene editing techniques.

Lastly, as new agents are developed, it is necessary to consider the logistical, global health challenge of distributing new medications to areas with a higher viral burden. The prevalence of the virus is highest in the resource-limited regions of China, South America, Southeast Asia, and sub-Saharan Africa. Distribution of new agents can be limited by geography, public health infrastructure and surveillance, and government cooperation.113 As new novel agents are investigated in clinical trials, it is also important to consider the global representation of HBV. Ideally, new agents should be studied in clinical trials in areas most burdened by chronic HBV, as these populations would benefit most from treatment.

Conclusions

A rapid surge in the development of novel agents has sparked hope that it might be possible to finally cure HBV. However, fulfillment of that hope appears to remain in the distant future as significant challenges remain due to the biological characteristics of cccDNA and integrated HBV DNA. Targeted delivery, sustained action, without off-target effects remain the most challenging objectives. Creative solutions are needed to make PCR-based assay methods affordable, minimally invasive, and readily available for quantification of cccDNA accurately in HBV infections. Novel agents targeting multiple steps of the viral life cycle may be necessary to achieve effective inhibition of HBsAg and cccDNA. While promising, even the most advanced agents will require more investigation to make HBV cure a reality.

Abbreviations

AFT: 

artificial transcription factor

APOBEC: 

apolipoprotein B mRNA editing catalytic polypeptide-like

Cas: 

CRISPR-associated

cccDNA: 

covalently closed circular DNA

CHB: 

chronic hepatitis B

CRISPR: 

clustered regularly interspaced short palindromic repeats

C-TALEN: 

C/pol TALEN

DAA: 

direct acting antivirals

DBD: 

DNA-binding domain

DDB1: 

damage-specific DNA-binding protein 1

DHBV: 

duck hepatitis B virus

DSB: 

double-stranded break

dslDNA: 

double-stranded linear DNA

HAT1: 

histone acetyltransferase 1

HBcAg: 

hepatitis B core antigen

HBeAg: 

hepatitis B e antigen

HBsAg: 

hepatitis B surface antigen

HBV: 

hepatitis B virus

HBx: 

HBV regulatory X

HDV: 

hepatitis D virus

IFN-1: 

interferon type 1

LTβR: 

lymphotoxin β receptor

NAP: 

Nucleic acid polymer

NHEJ: 

nonhomologous end joining

NIRF: 

Np95/ICBP90-like RING finger protein

NK: 

natural killer

NTCP: 

sodium taurocholate co-transporting polypeptide

NTZ: 

nitazoxanide

PD-1: 

programmed cell death protein

Peg: 

pegylated

pgRNA: 

pregenomic RNA

rcDNA: 

relaxed circular DNA

sgRNA: 

single guide RNA

shRNA: 

short hairpin RNA

SIRT3: 

silent mating type information regulation 2 homolog 3

SMC-5/6: 

structural maintenance of chromosomes 5/6

S-TALEN: 

S/pol TALEN

STOPS: 

S-antigen traffic inhibiting oligonucleotides polymers

TALEN: 

transcription activator-like effector nuclease

TLR: 

Toll-like receptor

ZFs: 

zinc fingers

ZFNs: 

zinc finger nucleases

Declarations

Acknowledgement

This work was made possible by the Herman Lopata Chair in Hepatitis Research.

Funding

None to declare.

Conflict of interest

GYW has been an editor-in-chief of Journal of Clinical and Translational Hepatology since 2013. The other authors have no conflict of interests related to this publication.

Authors’ contributions

Proposed concept for review and revised manuscript with critical revisions (GYW), drafted the manuscript (AHA and BDH).

References

  1. Michielsen P, Ho E. Viral hepatitis B and hepatocellular carcinoma. Acta Gastroenterol Belg 2011;74(1):4-8 View Article PubMed/NCBI
  2. Hepatitis B. Available from: https://www.who.int/news-room/fact-sheets/detail/hepatitis-b View Article PubMed/NCBI
  3. Tsai KN, Kuo CF, Ou JJ. Mechanisms of Hepatitis B Virus Persistence. Trends Microbiol 2018;26(1):33-42 View Article PubMed/NCBI
  4. Yuen MF, Chen DS, Dusheiko GM, Janssen HLA, Lau DTY, Locarnini SA, et al. Hepatitis B virus infection. Nat Rev Dis Primers 2018;4:18035 View Article PubMed/NCBI
  5. Sureau C, Salisse J. A conformational heparan sulfate binding site essential to infectivity overlaps with the conserved hepatitis B virus a-determinant. Hepatology 2013;57(3):985-994 View Article PubMed/NCBI
  6. Yan H, Zhong G, Xu G, He W, Jing Z, Gao Z, et al. Sodium taurocholate cotransporting polypeptide is a functional receptor for human hepatitis B and D virus. Elife 2012;1:e00049 View Article PubMed/NCBI
  7. Guo H, Jiang D, Zhou T, Cuconati A, Block TM, Guo JT. Characterization of the intracellular deproteinized relaxed circular DNA of hepatitis B virus: an intermediate of covalently closed circular DNA formation. J Virol 2007;81(22):12472-12484 View Article PubMed/NCBI
  8. Chong CK, Cheng CYS, Tsoi SYJ, Huang FY, Liu F, Seto WK, et al. Role of hepatitis B core protein in HBV transcription and recruitment of histone acetyltransferases to cccDNA minichromosome. Antiviral Res 2017;144:1-7 View Article PubMed/NCBI
  9. Nassal M. Hepatitis B viruses: reverse transcription a different way. Virus Res 2008;134(1-2):235-249 View Article PubMed/NCBI
  10. Hu J, Cheng J, Tang L, Hu Z, Luo Y, Li Y, et al. Virological Basis for the Cure of Chronic Hepatitis B. ACS Infect Dis 2019;5(5):659-674 View Article PubMed/NCBI
  11. Zhang D, Zhang K, Protzer U, Zeng C. HBV Integration Induces Complex Interactions between Host and Viral Genomic Functions at the Insertion Site. J Clin Transl Hepatol 2021;9(3):399-408 View Article PubMed/NCBI
  12. Levrero M, Pollicino T, Petersen J, Belloni L, Raimondo G, Dandri M. Control of cccDNA function in hepatitis B virus infection. J Hepatol 2009;51(3):581-592 View Article PubMed/NCBI
  13. Wong GLH, Gane E, Lok ASF. How to achieve functional cure of HBV: Stopping NUCs, adding interferon or new drug development?. J Hepatol 2022;76(6):1249-1262 View Article PubMed/NCBI
  14. Lebossé F, Testoni B, Fresquet J, Facchetti F, Galmozzi E, Fournier M, et al. Intrahepatic innate immune response pathways are downregulated in untreated chronic hepatitis B. J Hepatol 2017;66(5):897-909 View Article PubMed/NCBI
  15. Maini MK, Gehring AJ. The role of innate immunity in the immunopathology and treatment of HBV infection. J Hepatol 2016;64(1 Suppl):S60-S70 View Article PubMed/NCBI
  16. Martinet J, Dufeu-Duchesne T, Bruder Costa J, Larrat S, Marlu A, Leroy V, et al. Altered functions of plasmacytoid dendritic cells and reduced cytolytic activity of natural killer cells in patients with chronic HBV infection. Gastroenterology 2012;143(6):1586-1596.e8 View Article PubMed/NCBI
  17. Bertoletti A, Ferrari C. Adaptive immunity in HBV infection. J Hepatol 2016;64(1 Suppl):S71-S83 View Article PubMed/NCBI
  18. Cai D, Mills C, Yu W, Yan R, Aldrich CE, Saputelli JR, et al. Identification of disubstituted sulfonamide compounds as specific inhibitors of hepatitis B virus covalently closed circular DNA formation. Antimicrob Agents Chemother 2012;56(8):4277-4288 View Article PubMed/NCBI
  19. Liu C, Cai D, Zhang L, Tang W, Yan R, Guo H, et al. Identification of hydrolyzable tannins (punicalagin, punicalin and geraniin) as novel inhibitors of hepatitis B virus covalently closed circular DNA. Antiviral Res 2016;134:97-107 View Article PubMed/NCBI
  20. Amblard F, Boucle S, Bassit L, Cox B, Sari O, Tao S, et al. Erratum for Amblard et al., “Novel Hepatitis B Virus Capsid Assembly Modulator Induces Potent Antiviral Responses In Vitro and in Humanized Mice”. Antimicrob Agents Chemother 2020;64(9):e01351-20 View Article PubMed/NCBI
  21. Li T, Robert EI, van Breugel PC, Strubin M, Zheng N. A promiscuous alpha-helical motif anchors viral hijackers and substrate receptors to the CUL4-DDB1 ubiquitin ligase machinery. Nat Struct Mol Biol 2010;17(1):105-111 View Article PubMed/NCBI
  22. Sekiba K, Otsuka M, Ohno M, Yamagami M, Kishikawa T, Suzuki T, et al. Inhibition of HBV Transcription From cccDNA With Nitazoxanide by Targeting the HBx-DDB1 Interaction. Cell Mol Gastroenterol Hepatol 2019;7(2):297-312 View Article PubMed/NCBI
  23. Cheng ST, Hu JL, Ren JH, Yu HB, Zhong S, Wai Wong VK, et al. Dicoumarol, an NQO1 inhibitor, blocks cccDNA transcription by promoting degradation of HBx. J Hepatol 2021;74(3):522-534 View Article PubMed/NCBI
  24. Horng JH, Lin WH, Wu CR, Lin YY, Wu LL, Chen DS, et al. HBV X protein-based therapeutic vaccine accelerates viral antigen clearance by mobilizing monocyte infiltration into the liver in HBV carrier mice. J Biomed Sci 2020;27(1):70 View Article PubMed/NCBI
  25. Pollicino T, Belloni L, Raffa G, Pediconi N, Squadrito G, Raimondo G, et al. Hepatitis B virus replication is regulated by the acetylation status of hepatitis B virus cccDNA-bound H3 and H4 histones. Gastroenterology 2006;130(3):823-837 View Article PubMed/NCBI
  26. Kim JW, Lee SH, Park YS, Hwang JH, Jeong SH, Kim N, et al. Replicative activity of hepatitis B virus is negatively associated with methylation of covalently closed circular DNA in advanced hepatitis B virus infection. Intervirology 2011;54(6):316-325 View Article PubMed/NCBI
  27. Gan W, Gao N, Gu L, Mo Z, Pang X, Lei Z, et al. Reduction in Intrahepatic cccDNA and Integration of HBV in Chronic Hepatitis B Patients with a Functional Cure. J Clin Transl Hepatol 2023;11(2):314-322 View Article PubMed/NCBI
  28. Guo Y, Li Y, Mu S, Zhang J, Yan Z. Evidence that methylation of hepatitis B virus covalently closed circular DNA in liver tissues of patients with chronic hepatitis B modulates HBV replication. J Med Virol 2009;81(7):1177-1183 View Article PubMed/NCBI
  29. Lee JY, Kim NA, Sanford A, Sullivan KE. Histone acetylation and chromatin conformation are regulated separately at the TNF-alpha promoter in monocytes and macrophages. J Leukoc Biol 2003;73(6):862-871 View Article PubMed/NCBI
  30. Park HK, Min BY, Kim NY, Jang ES, Shin CM, Park YS, et al. Short hairpin RNA induces methylation of hepatitis B virus covalently closed circular DNA in human hepatoma cells. Biochem Biophys Res Commun 2013;436(2):152-155 View Article PubMed/NCBI
  31. Wei ZQ, Zhang YH, Ke CZ, Chen HX, Ren P, He YL, et al. Curcumin inhibits hepatitis B virus infection by down-regulating cccDNA-bound histone acetylation. World J Gastroenterol 2017;23(34):6252-6260 View Article PubMed/NCBI
  32. Ren JH, Hu JL, Cheng ST, Yu HB, Wong VKW, Law BYK, et al. SIRT3 restricts hepatitis B virus transcription and replication through epigenetic regulation of covalently closed circular DNA involving suppressor of variegation 3-9 homolog 1 and SET domain containing 1A histone methyltransferases. Hepatology 2018;68(4):1260-1276 View Article PubMed/NCBI
  33. Yang G, Feng J, Liu Y, Zhao M, Yuan Y, Yuan H, et al. HAT1 signaling confers to assembly and epigenetic regulation of HBV cccDNA minichromosome. Theranostics 2019;9(24):7345-7358 View Article PubMed/NCBI
  34. Qian G, Hu B, Zhou D, Xuan Y, Bai L, Duan C. NIRF, a Novel Ubiquitin Ligase, Inhibits Hepatitis B Virus Replication Through Effect on HBV Core Protein and H3 Histones. DNA Cell Biol 2015;34(5):327-332 View Article PubMed/NCBI
  35. Janahi EM, McGarvey MJ. The inhibition of hepatitis B virus by APOBEC cytidine deaminases. J Viral Hepat 2013;20(12):821-828 View Article PubMed/NCBI
  36. Lucifora J, Xia Y, Reisinger F, Zhang K, Stadler D, Cheng X, et al. Specific and nonhepatotoxic degradation of nuclear hepatitis B virus cccDNA. Science 2014;343(6176):1221-1228 View Article PubMed/NCBI
  37. Turelli P, Mangeat B, Jost S, Vianin S, Trono D. Inhibition of hepatitis B virus replication by APOBEC3G. Science 2004;303(5665):1829 View Article PubMed/NCBI
  38. Baumert TF, Rösler C, Malim MH, von Weizsäcker F. Hepatitis B virus DNA is subject to extensive editing by the human deaminase APOBEC3C. Hepatology 2007;46(3):682-689 View Article PubMed/NCBI
  39. Nguyen DH, Gummuluru S, Hu J. Deamination-independent inhibition of hepatitis B virus reverse transcription by APOBEC3G. J Virol 2007;81(9):4465-4472 View Article PubMed/NCBI
  40. Chen Y, Hu J, Cai X, Huang Y, Zhou X, Tu Z, et al. APOBEC3B edits HBV DNA and inhibits HBV replication during reverse transcription. Antiviral Res 2018;149:16-25 View Article PubMed/NCBI
  41. Wolf MJ, Seleznik GM, Zeller N, Heikenwalder M. The unexpected role of lymphotoxin beta receptor signaling in carcinogenesis: from lymphoid tissue formation to liver and prostate cancer development. Oncogene 2010;29(36):5006-5018 View Article PubMed/NCBI
  42. Gane E, Verdon DJ, Brooks AE, Gaggar A, Nguyen AH, Subramanian GM, et al. Anti-PD-1 blockade with nivolumab with and without therapeutic vaccination for virally suppressed chronic hepatitis B: A pilot study. J Hepatol 2019;71(5):900-907 View Article PubMed/NCBI
  43. Janssen HLA, Brunetto MR, Kim YJ, Ferrari C, Massetto B, Nguyen AH, et al. Safety, efficacy and pharmacodynamics of vesatolimod (GS-9620) in virally suppressed patients with chronic hepatitis B. J Hepatol 2018;68(3):431-440 View Article PubMed/NCBI
  44. Lino CA, Harper JC, Carney JP, Timlin JA. Delivering CRISPR: a review of the challenges and approaches. Drug Deliv 2018;25(1):1234-1257 View Article PubMed/NCBI
  45. Singh P, Kairuz D, Arbuthnot P, Bloom K. Silencing hepatitis B virus covalently closed circular DNA: The potential of an epigenetic therapy approach. World J Gastroenterol 2021;27(23):3182-3207 View Article PubMed/NCBI
  46. Mak AN, Bradley P, Bogdanove AJ, Stoddard BL. TAL effectors: function, structure, engineering and applications. Curr Opin Struct Biol 2013;23(1):93-99 View Article PubMed/NCBI
  47. Cermak T, Doyle EL, Christian M, Wang L, Zhang Y, Schmidt C, et al. Efficient design and assembly of custom TALEN and other TAL effector-based constructs for DNA targeting. Nucleic Acids Res 2011;39(12):e82 View Article PubMed/NCBI
  48. Moyo B, Bloom K, Scott T, Ely A, Arbuthnot P. Advances with using CRISPR/Cas-mediated gene editing to treat infections with hepatitis B virus and hepatitis C virus. Virus Res 2018;244:311-320 View Article PubMed/NCBI
  49. Seeger C, Sohn JA. Complete Spectrum of CRISPR/Cas9-induced Mutations on HBV cccDNA. Mol Ther 2016;24(7):1258-1266 View Article PubMed/NCBI
  50. Kennedy EM, Kornepati AV, Cullen BR. Targeting hepatitis B virus cccDNA using CRISPR/Cas9. Antiviral Res 2015;123:188-192 View Article PubMed/NCBI
  51. Dong C, Qu L, Wang H, Wei L, Dong Y, Xiong S. Targeting hepatitis B virus cccDNA by CRISPR/Cas9 nuclease efficiently inhibits viral replication. Antiviral Res 2015;118:110-117 View Article PubMed/NCBI
  52. Karimova M, Beschorner N, Dammermann W, Chemnitz J, Indenbirken D, Bockmann JH, et al. CRISPR/Cas9 nickase-mediated disruption of hepatitis B virus open reading frame S and X. Sci Rep 2015;5:13734 View Article PubMed/NCBI
  53. Kennedy EM, Bassit LC, Mueller H, Kornepati AVR, Bogerd HP, Nie T, et al. Suppression of hepatitis B virus DNA accumulation in chronically infected cells using a bacterial CRISPR/Cas RNA-guided DNA endonuclease. Virology 2015;476:196-205 View Article PubMed/NCBI
  54. Lin SR, Yang HC, Kuo YT, Liu CJ, Yang TY, Sung KC, et al. The CRISPR/Cas9 System Facilitates Clearance of the Intrahepatic HBV Templates In Vivo. Mol Ther Nucleic Acids 2014;3(8):e186 View Article PubMed/NCBI
  55. Liu X, Hao R, Chen S, Guo D, Chen Y. Inhibition of hepatitis B virus by the CRISPR/Cas9 system via targeting the conserved regions of the viral genome. J Gen Virol 2015;96(8):2252-2261 View Article PubMed/NCBI
  56. Ramanan V, Shlomai A, Cox DB, Schwartz RE, Michailidis E, Bhatta A, et al. CRISPR/Cas9 cleavage of viral DNA efficiently suppresses hepatitis B virus. Sci Rep 2015;5:10833 View Article PubMed/NCBI
  57. Seeger C, Sohn JA. Targeting Hepatitis B Virus With CRISPR/Cas9. Mol Ther Nucleic Acids 2014;3(12):e216 View Article PubMed/NCBI
  58. Wang J, Xu ZW, Liu S, Zhang RY, Ding SL, Xie XM, et al. Dual gRNAs guided CRISPR/Cas9 system inhibits hepatitis B virus replication. World J Gastroenterol 2015;21(32):9554-9565 View Article PubMed/NCBI
  59. Zhen S, Hua L, Liu YH, Gao LC, Fu J, Wan DY, et al. Harnessing the clustered regularly interspaced short palindromic repeat (CRISPR)/CRISPR-associated Cas9 system to disrupt the hepatitis B virus. Gene Ther 2015;22(5):404-412 View Article PubMed/NCBI
  60. Li H, Sheng C, Wang S, Yang L, Liang Y, Huang Y, et al. Removal of Integrated Hepatitis B Virus DNA Using CRISPR-Cas9. Front Cell Infect Microbiol 2017;7:91 View Article PubMed/NCBI
  61. Martinez MG, Combe E, Inchauspe A, Mangeot PE, Delberghe E, Chapus F, et al. CRISPR-Cas9 Targeting of Hepatitis B Virus Covalently Closed Circular DNA Generates Transcriptionally Active Episomal Variants. mBio 2022;13(2):e0288821 View Article PubMed/NCBI
  62. Roehl I, Seiffert S, Brikh C, Quinet J, Jamard C, Dorfler N, et al. Nucleic Acid Polymers with Accelerated Plasma and Tissue Clearance for Chronic Hepatitis B Therapy. Mol Ther Nucleic Acids 2017;8:1-12 View Article PubMed/NCBI
  63. Bazinet M, Pântea V, Placinta G, Moscalu I, Cebotarescu V, Cojuhari L, et al. Safety and Efficacy of 48 Weeks REP 2139 or REP 2165, Tenofovir Disoproxil, and Pegylated Interferon Alfa-2a in Patients With Chronic HBV Infection Naïve to Nucleos(t)ide Therapy. Gastroenterology 2020;158(8):2180-2194 View Article PubMed/NCBI
  64. Kao CC, Nie Y, Ren S, De Costa NTTS, Pandey RK, Hong J, et al. Mechanism of action of hepatitis B virus S antigen transport-inhibiting oligonucleotide polymer, STOPS, molecules. Mol Ther Nucleic Acids 2022;27:335-348 View Article PubMed/NCBI
  65. Gane E, Yuen M, Jucov A, Kultgen S, Kim HJ, Tseng CH, et al. Safety, tolerability and pharmacokinetics (PK) of single and multiple doses of ALG-010133, an S-antigen Transport Inhibiting Oligonucleotide Polymer (STOP) for the treatment of chronic hepatitis B. J Hepatol 2021;75:S741 View Article PubMed/NCBI
  66. Alexopoulou A, Vasilieva L, Karayiannis P. New Approaches to the Treatment of Chronic Hepatitis B. J Clin Med 2020;9(10):3187 View Article PubMed/NCBI
  67. Yang HC, Kao JH. Persistence of hepatitis B virus covalently closed circular DNA in hepatocytes: molecular mechanisms and clinical significance. Emerg Microbes Infect 2014;3(9):e64 View Article PubMed/NCBI
  68. Köck J, Schlicht HJ. Analysis of the earliest steps of hepadnavirus replication: genome repair after infectious entry into hepatocytes does not depend on viral polymerase activity. J Virol 1993;67(8):4867-4874 View Article PubMed/NCBI
  69. Delmas J, Schorr O, Jamard C, Gibbs C, Trépo C, Hantz O, et al. Inhibitory effect of adefovir on viral DNA synthesis and covalently closed circular DNA formation in duck hepatitis B virus-infected hepatocytes in vivo and in vitro. Antimicrob Agents Chemother 2002;46(2):425-433 View Article PubMed/NCBI
  70. Köck J, Baumert TF, Delaney WE, Blum HE, von Weizsäcker F. Inhibitory effect of adefovir and lamivudine on the initiation of hepatitis B virus infection in primary tupaia hepatocytes. Hepatology 2003;38(6):1410-1418 View Article PubMed/NCBI
  71. Bowden S, Locarnini S, Chang TT, Chao YC, Han KH, Gish RG, et al. Covalently closed-circular hepatitis B virus DNA reduction with entecavir or lamivudine. World J Gastroenterol 2015;21(15):4644-4651 View Article PubMed/NCBI
  72. Tang LSY, Covert E, Wilson E, Kottilil S. Chronic Hepatitis B Infection: A Review. JAMA 2018;319(17):1802-1813 View Article PubMed/NCBI
  73. Allweiss L, Testoni B, Yu M, Lucifora J, Ko C, Qu B, et al. Quantification of the hepatitis B virus cccDNA: evidence-based guidelines for monitoring the key obstacle of HBV cure. Gut 2023;72(5):972-983 View Article PubMed/NCBI
  74. Keasler VV, Hodgson AJ, Madden CR, Slagle BL. Enhancement of hepatitis B virus replication by the regulatory X protein in vitro and in vivo. J Virol 2007;81(6):2656-2662 View Article PubMed/NCBI
  75. Decorsière A, Mueller H, van Breugel PC, Abdul F, Gerossier L, Beran RK, et al. Hepatitis B virus X protein identifies the Smc5/6 complex as a host restriction factor. Nature 2016;531(7594):386-389 View Article PubMed/NCBI
  76. Murphy CM, Xu Y, Li F, Nio K, Reszka-Blanco N, Li X, et al. Hepatitis B Virus X Protein Promotes Degradation of SMC5/6 to Enhance HBV Replication. Cell Rep 2016;16(11):2846-2854 View Article PubMed/NCBI
  77. Livingston CM, Ramakrishnan D, Strubin M, Fletcher SP, Beran RK. Identifying and Characterizing Interplay between Hepatitis B Virus X Protein and Smc5/6. Viruses 2017;9(4):69 View Article PubMed/NCBI
  78. Gao Y, Feng J, Yang G, Zhang S, Liu Y, Bu Y, et al. Hepatitis B virus X protein-elevated MSL2 modulates hepatitis B virus covalently closed circular DNA by inducing degradation of APOBEC3B to enhance hepatocarcinogenesis. Hepatology 2017;66(5):1413-1429 View Article PubMed/NCBI
  79. Shen C, Feng X, Mao T, Yang D, Zou J, Zao X, et al. Yin-Yang 1 and HBx protein activate HBV transcription by mediating the spatial interaction of cccDNA minichromosome with cellular chromosome 19p13.11. Emerg Microbes Infect 2020;9(1):2455-2464 View Article PubMed/NCBI
  80. Sitterlin D, Bergametti F, Tiollais P, Tennant BC, Transy C. Correct binding of viral X protein to UVDDB-p127 cellular protein is critical for efficient infection by hepatitis B viruses. Oncogene 2000;19(38):4427-4431 View Article PubMed/NCBI
  81. Sitterlin D, Bergametti F, Transy C. UVDDB p127-binding modulates activities and intracellular distribution of hepatitis B virus X protein. Oncogene 2000;19(38):4417-4426 View Article PubMed/NCBI
  82. Sitterlin D, Lee TH, Prigent S, Tiollais P, Butel JS, Transy C. Interaction of the UV-damaged DNA-binding protein with hepatitis B virus X protein is conserved among mammalian hepadnaviruses and restricted to transactivation-proficient X-insertion mutants. J Virol 1997;71(8):6194-6199 View Article PubMed/NCBI
  83. Zhang JF, Xiong HL, Cao JL, Wang SJ, Guo XR, Lin BY, et al. A cell-penetrating whole molecule antibody targeting intracellular HBx suppresses hepatitis B virus via TRIM21-dependent pathway. Theranostics 2018;8(2):549-562 View Article PubMed/NCBI
  84. Belloni L, Pollicino T, De Nicola F, Guerrieri F, Raffa G, Fanciulli M, et al. Nuclear HBx binds the HBV minichromosome and modifies the epigenetic regulation of cccDNA function. Proc Natl Acad Sci U S A 2009;106(47):19975-19979 View Article PubMed/NCBI
  85. Vivekanandan P, Thomas D, Torbenson M. Methylation regulates hepatitis B viral protein expression. J Infect Dis 2009;199(9):1286-1291 View Article PubMed/NCBI
  86. Zhang Y, Mao R, Yan R, Cai D, Zhang Y, Zhu H, et al. Transcription of hepatitis B virus covalently closed circular DNA is regulated by CpG methylation during chronic infection. PLoS One 2014;9(10):e110442 View Article PubMed/NCBI
  87. Gane EJ, Kim HJ, Visvanathan K, Kim YJ, Nguyen AH, Wallin JJ, et al. Safety, Pharmacokinetics, and Pharmacodynamics of the Oral TLR8 Agonist Selgantolimod in Chronic Hepatitis B. Hepatology 2021;74(4):1737-1749 View Article PubMed/NCBI
  88. Porteus MH. Towards a new era in medicine: therapeutic genome editing. Genome Biol 2015;16:286 View Article PubMed/NCBI
  89. Ruiz de Galarreta M, Lujambio A. Therapeutic editing of hepatocyte genome in vivo. J Hepatol 2017;67(4):818-828 View Article PubMed/NCBI
  90. Takata M, Sasaki MS, Sonoda E, Morrison C, Hashimoto M, Utsumi H, et al. Homologous recombination and non-homologous end-joining pathways of DNA double-strand break repair have overlapping roles in the maintenance of chromosomal integrity in vertebrate cells. EMBO J 1998;17(18):5497-5508 View Article PubMed/NCBI
  91. Bloom K, Maepa MB, Ely A, Arbuthnot P. Gene Therapy for Chronic HBV-Can We Eliminate cccDNA?. Genes (Basel) 2018;9(4):207 View Article PubMed/NCBI
  92. Maeder ML, Gersbach CA. Genome-editing Technologies for Gene and Cell Therapy. Mol Ther 2016;24(3):430-446 View Article PubMed/NCBI
  93. Urnov FD, Rebar EJ, Holmes MC, Zhang HS, Gregory PD. Genome editing with engineered zinc finger nucleases. Nat Rev Genet 2010;11(9):636-646 View Article PubMed/NCBI
  94. Weber ND, Stone D, Sedlak RH, De Silva Feelixge HS, Roychoudhury P, Schiffer JT, et al. AAV-mediated delivery of zinc finger nucleases targeting hepatitis B virus inhibits active replication. PLoS One 2014;9(5):e97579 View Article PubMed/NCBI
  95. Zimmerman KA, Fischer KP, Joyce MA, Tyrrell DL. Zinc finger proteins designed to specifically target duck hepatitis B virus covalently closed circular DNA inhibit viral transcription in tissue culture. J Virol 2008;82(16):8013-8021 View Article PubMed/NCBI
  96. Bloom K, Ely A, Mussolino C, Cathomen T, Arbuthnot P. Inactivation of hepatitis B virus replication in cultured cells and in vivo with engineered transcription activator-like effector nucleases. Mol Ther 2013;21(10):1889-1897 View Article PubMed/NCBI
  97. Chen J, Zhang W, Lin J, Wang F, Wu M, Chen C, et al. An efficient antiviral strategy for targeting hepatitis B virus genome using transcription activator-like effector nucleases. Mol Ther 2014;22(2):303-311 View Article PubMed/NCBI
  98. Croagh CM, Desmond PV, Bell SJ. Genotypes and viral variants in chronic hepatitis B: A review of epidemiology and clinical relevance. World J Hepatol 2015;7(3):289-303 View Article PubMed/NCBI
  99. Li F, Zhang D, Li Y, Jiang D, Luo S, Du N, et al. Whole genome characterization of hepatitis B virus quasispecies with massively parallel pyrosequencing. Clin Microbiol Infect 2015;21(3):280-287 View Article PubMed/NCBI
  100. Zhao K, Liu S, Chen Y, Yao Y, Zhou M, Yuan Y, et al. Upregulation of HBV transcription by sodium taurocholate cotransporting polypeptide at the postentry step is inhibited by the entry inhibitor Myrcludex B. Emerg Microbes Infect 2018;7(1):186 View Article PubMed/NCBI
  101. Bogomolov P, Alexandrov A, Voronkova N, Macievich M, Kokina K, Petrachenkova M, et al. Treatment of chronic hepatitis D with the entry inhibitor myrcludex B: First results of a phase Ib/IIa study. J Hepatol 2016;65(3):490-498 View Article PubMed/NCBI
  102. Volz T, Allweiss L, Ben MBarek M, Warlich M, Lohse AW, Pollok JM, et al. The entry inhibitor Myrcludex-B efficiently blocks intrahepatic virus spreading in humanized mice previously infected with hepatitis B virus. J Hepatol 2013;58(5):861-867 View Article PubMed/NCBI
  103. Yuen MF, Lim SG, Plesniak R, Tsuji K, Janssen HLA, Pojoga C, et al. Efficacy and Safety of Bepirovirsen in Chronic Hepatitis B Infection. N Engl J Med 2022;387(21):1957-1968 View Article PubMed/NCBI
  104. Miao J, Gao P, Li Q, He K, Zhang L, Wang J, et al. Advances in Nanoparticle Drug Delivery Systems for Anti-Hepatitis B Virus Therapy: A Narrative Review. Int J Mol Sci 2021;22(20):11227 View Article PubMed/NCBI
  105. Kang JH, Toita R, Murata M. Liver cell-targeted delivery of therapeutic molecules. Crit Rev Biotechnol 2016;36(1):132-143 View Article PubMed/NCBI
  106. Glebe D, Urban S. Viral and cellular determinants involved in hepadnaviral entry. World J Gastroenterol 2007;13(1):22-38 View Article PubMed/NCBI
  107. Bonamassa B, Hai L, Liu D. Hydrodynamic gene delivery and its applications in pharmaceutical research. Pharm Res 2011;28(4):694-701 View Article PubMed/NCBI
  108. Singh L, Indermun S, Govender M, Kumar P, du Toit LC, Choonara YE, et al. Drug Delivery Strategies for Antivirals against Hepatitis B Virus. Viruses 2018;10(5):267 View Article PubMed/NCBI
  109. Kapoor R, Kottilil S. Strategies to eliminate HBV infection. Future Virol 2014;9(6):565-585 View Article PubMed/NCBI
  110. Lebossé F, Inchauspé A, Locatelli M, Miaglia C, Diederichs A, Fresquet J, et al. Quantification and epigenetic evaluation of the residual pool of hepatitis B covalently closed circular DNA in long-term nucleoside analogue-treated patients. Sci Rep 2020;10(1):21097 View Article PubMed/NCBI
  111. Tu T, Zehnder B, Qu B, Ni Y, Main N, Allweiss L, et al. A novel method to precisely quantify hepatitis B virus covalently closed circular (ccc)DNA formation and maintenance. Antiviral Res 2020;181:104865 View Article PubMed/NCBI
  112. Naeem M, Majeed S, Hoque MZ, Ahmad I. Latest Developed Strategies to Minimize the Off-Target Effects in CRISPR-Cas-Mediated Genome Editing. Cells 2020;9(7):1608 View Article PubMed/NCBI
  113. GBD 2019 Hepatitis B Collaborators. Global, regional, and national burden of hepatitis B, 1990-2019: a systematic analysis for the Global Burden of Disease Study 2019. Lancet Gastroenterol Hepatol 2022;7(9):796-829 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