You are on page 1of 13

Received: 27 November 2017 | Accepted: 27 November 2017

DOI: 10.1111/liv.13656

REVIEW ARTICLE

Towards HBV curative therapies

Raymond F. Schinazi1 | Maryam Ehteshami1 | Leda Bassit1 | Tarik Asselah2

1
Center for AIDS Research, Emory University
School of Medicine, Atlanta, GA, USA Abstract
2
Department of Hepatology, Centre de Tremendous progress has been made over the last 2 decades to discover and develop
Recherche sur l’Inflammation, Viral Hepatitis
approaches to control hepatitis B virus (HBV) infections and to prevent the develop-
INSERM UMR 1149, AP-HP Hôpital
Beaujon, Université Paris Diderot, Clichy, ment of hepatocellular carcinoma using various interferons and small molecules as
France
antiviral agents. However, none of these agents have significant impact on eliminating
Correspondence HBV from infected cells. Currently the emphasis is on silencing or eliminating cccDNA,
Raymond F. Schinazi, PhD, DSc, Center for
which could lead to a cure for HBV. Various approaches are being developed including
AIDS Research, Emory University School of
Medicine, Atlanta, GA, USA. the development of capsid effectors, CRISPR/Cas9, TALENS, siRNA, entry and secre-
Email: rschina@emory.edu
tion inhibitors, as well as immunological approaches. It is very likely that a combination
of these modalities will need to be employed to successfully eliminate HBV or prevent
Funding information
This work was supported by NIH grant 1R01-­ virus rebound on discontinuation of therapy. In the next 5 years clinical data will
AI-­132833 and in part by funding from the emerge which will provide insight on the safety and feasibility of these approaches and
NIH funded Emory Center for AIDS Research
grant P30-­AI-­050409 to RFS. if they can be applied to eradicate HBV infections globally. In this review, we summa-
rize current treatments and we highlight and examine recent therapeutic strategies
Handling Editor: Zobair Younossi
that are currently being evaluated at the preclinical and clinical stage.

KEYWORDS
antiviral agents, cccDNA, HBV cure, immunotherapy

1 | INTRODUCTION 2 | HBV REPLICATION CYCLE AND


THERAPEUTIC TARGETS
Chronic HBV infection affects around 240 million people worldwide
(Figure 1A) and long-­term risks such as cirrhosis and hepatocellular HBV belongs to the hepadnaviridae family of viruses. Similar to
carcinoma (HCC) account for approximately 600 000 deaths annu- other related species such as duck hepatitis virus and woodchuck
1
ally. HCC is one of the most frequent cancers in Africa and Asia hepatitis virus, the relatively small genome of HBV consists of a
and fibrosis is the most important prognostic predictor of survival.2 3.2 kb partially double-­stranded DNA, referred to as relaxed cir-
Despite the availability of several FDA-­approved drugs (Figure 1B), cular DNA (rcDNA). HBV infects hepatocytes through a recently
continuous treatment is necessary for virus replication control. identified human sodium taurocholate cotransporting polypep-
Therefore, current HBV research and development programmes aim tide (hNTCP) receptor.3 Upon fusion with the host membrane, the
to achieve a curative strategy that either eliminates or permanently rcDNA-­containing nucleocapsid is released into the cytoplasm and
silences HBV infection. This review will discuss approved therapies travels to the nucleus (Figure 2). Once inside, rcDNA is converted
and will review novel therapeutic agents currently in development. to a highly stable episomal DNA known as covalently closed circular

Abbreviations: AAV, adeno-associated virus; AE, adverse event; ALT, alanine aminotransferase; AST, aspartate aminotransferase; cccDNA, covalently closed circular DNA; CHB, chronic hepatitis
B; eGFRCG, estimated glomerular filtration rate, Cockcroft-Gault method; ETV, entecavir; HAP, heteroarylpyrimidines; HBeAg, Hepatitis B e antigen; nucleocapsid. precore protein; HBeAg-ve,
antigen-negative; HBeAg+ve, HBe antigen-positive; HBsAg, Hepatitis B surface antigen; HBV, hepatitis B virus; HBx, viral protein X; HCC, hepatocellular carcinoma; hNTCP, human sodium
taurocholate cotransporting polypeptide; IFN-α, interferon alpha; LAM, lamivudine; LdT, telbivudine; mRNA, messenger RNA; NA, nucleoside analogue; NI, nucleoside inhibitors; NNI, non-­
nucleoside inhibitors; ORF, open reading frames; PEG-IFN, pegylated interferon; pgRNA, pregenomic RNA; PP, phenylpropenimides; QD, once daily; rcDNA, relaxed circular DNA; SBA, sulfam-
oylbenzamide; siRNA, small interfering RNA; TAF, tenofovir alafenamide; TDF, tenofovir disoproxil fumarate; TFV, tenofovir; TLR, toll-like receptor.

102 | © 2018 John Wiley & Sons A/S. wileyonlinelibrary.com/journal/liv Liver International. 2018;38(Suppl. 1):102–114.
Published by John Wiley & Sons Ltd
14783231, 2018, S1, Downloaded from https://onlinelibrary.wiley.com/doi/10.1111/liv.13656 by Cochrane Israel, Wiley Online Library on [02/12/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
SCHINAZI et al. | 103

DNA (cccDNA) via the host DNA repair machinery molecule.4,5 cc-
cDNA is the transcriptional template for subsequent virus gene ex- Key points
pression and generation of pregenomic RNA (pgRNA).6 HBV DNA • Chronic hepatitis B affects an estimated 240 million peo-
is also found integrated into the host chromosome. Integration of ple worldwide.
viral DNA does not appear to play a direct role in virus replication. • Current treatment includes the use of nucleoside ana-
Instead, it is thought that HBV DNA integration may render the logue inhibitors and interferons which can control virus
cellular environment more permissive to virus replication through replication but are not curative.
modulating gene expression. It also likely plays an important role in • Several novel therapeutic strategies are in development.
hepatocellular carcinogenesis (reviewed in Ref. 7). These include immunotherapy, gene-editing technology
Gene organization of cccDNA is uniquely intricate whereby sev- and small molecule inhibitors that target the viral capsid
eral overlapping open reading frames (ORFs) code for 7 viral pro- and cccDNA.
teins: pgRNA serves as the template for viral polymerase-­mediated • These novel therapies aim to provide a functional cure for
reverse transcription and subsequent synthesis of rcDNA. Another chronic hepatitis B.
gene product of HBV is the core protein, also known as hepatitis B
core antigen (HBcAg), which forms the viral nucleocapsid. Precore
protein (hepatitis B e antigen, HBeAg), is a proteolytically processed
viral protein that is often secreted from the infected cells and can 3 | APPROVED THERAPIES FOR
8
serve as a marker for disease stage. Viral surface proteins, S, M and TREATMENT OF HBV INFECTION
L (named based on their small, medium and large sizes respectively)
are coded from the S gene whereby S is translated from S mRNA, The goal of CHB therapy is to improve survival by preventing risk
M is translated from preS2 + S mRNA and L is translated from of cirrhosis and end-­stage liver disease, and to improve quality of
preS1 + PreS2 + S mRNA. All surface proteins of different length life.11,12 Several therapies have been approved for CHB infection
are collectively referred to as HBsAg. Finally, cccDNA also codes (Figure 1B). Historically, under long-­term lamivudine (LAM) treatment,
for viral protein X (HBx), a nonstructural protein that likely acts as a HBV DNA suppression leads to reduction in cirrhosis decompensa-
transcriptional transactivator and plays a role in regulating viral gene tion and HCC (Figure 1C).13,14 Thus, achieving HBV DNA clearance
expression. allows for reduction in necro-­inflammation and reduction in the risk
Once all viral proteins are synthesized and the rcDNA-­ of fibrosis progression. There are currently 2 principal therapeutic
containing nucleocapsid is formed, it can travel through the cel- strategies approved for both HBe antigen-­positive (HBeAg+ve) and
lular secretion pathway and be released as an enveloped and antigen-­negative (HBeAg-­ve) patients (Table 1). This includes a finite
infectious virion. Alternatively, the nucleocapsid can cycle back treatment course of interferon alpha (IFN-­α)/pegylated interferon
to the nucleus intracellularly, whereby the recently synthesized (PEG-­IFN) or, a long-­term maintenance treatment with nucleoside
rcDNA serves to replenish the cccDNA pool. In this way, cccDNA analogues (NAs).
can be maintained even in the absence of observable viremia.9 A 1-­year treatment with PEG-­IFN offers the potential for immune
Furthermore, cccDNA can remain dormant for a long time and control of HBV infection, with higher rates of HBeAg seroconversion
only become transcriptionally activated years or decades after and the possibility of viral suppression after stopping treatment, with
the initial infection. Although several therapeutic strategies have loss of hepatitis B surface antigen (HBsAg) in a significant propor-
been developed against HBV replication, targeting or eliminat- tion of patients who maintain undetectable HBV DNA (Figure 3).15-19
ing cccDNA, as an inert, yet highly stable mini-­chromosome has However, PEG-­IFN is administered by subcutaneous injection and is
proven challenging. In this way, cccDNA elimination, or at the very associated with poor tolerability and a risk of depression. Furthermore,
least, transcriptional control, lies at the heart of the quest for a PEG-­IFN is contraindicated in subjects with decompensated cirrhosis,
cure for chronic hepatitis B. in persons with autoimmune disease and during pregnancy.
In theory, any step of the virus replication cycle can be a target for NAs suppress HBV replication via direct antiviral activity. Usually,
10
antiviral therapeutics (Figure 2). The most successful target so far tolerability is good and compliance to treatment appears adequate.
has been the reverse transcription activity of pol. Interferon (IFN) treat- Entecavir (ETV) and tenofovir disoproximal fumarate (TDF) are potent
ment has also shown modest success in inhibiting virus replication. HBV inhibitors with a high barrier to resistance and should be used
Other areas of therapeutic research include targeting of the core/cap- as first-­line monotherapies. The chemical structures of various HBV
sid protein, mRNA transcription and cccDNA stability and formation. reverse transcriptase inhibitors are depicted in Figure 1B.20,21 ETV
In addition to gene expression targeting technologies such as CRISPR/ is a nucleoside analogue that disrupts DNA elongation as a delayed
Cas9 and small interfering RNA (siRNA), several immune-­modulatory chain-­terminator, whereas tenofovir (TFV) is a nucleotide (adenosine)
strategies that aim to enhance both the innate and adaptive response analogue that causes immediate chain termination when incorporated
to CHB infection are also under investigation. The following sections into HBV DNA during reverse transcription. ETV is unusual in that it is
will give a more detailed account of these novel strategies in develop- a D-­enantiomer of guanosine, and this in part accounts for its tighter
ment and their mechanism of action. binding in the active site of the enzyme as part of the mechanism of
14783231, 2018, S1, Downloaded from https://onlinelibrary.wiley.com/doi/10.1111/liv.13656 by Cochrane Israel, Wiley Online Library on [02/12/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
104 | SCHINAZI et al.

interference. Normal nucleotides can be incorporated following ETV indirect (or delayed) chain terminator, in contrast to the other agents
at the 3′ hydroxyl; nevertheless, chain termination occurs shortly that are used to treat HBV. TFV, in contrast, lacks a 2-­deoxyribose
thereafter because of structural disruptions to the enzyme, so it is an (cyclic) moiety including the 3′ hydroxyl group, and therefore once

(A)

≥ 8% = High
2-7% = Intermediate
<2% = Low

(B)

ADV TDF

ETV

TAF
ADV ETV TDF Approved Jan, 2017

1998 2002 2005 2006 2008 2016

LAM Peg-IFN LdT

TAF
LdT
LAM

(C)

Placebo (n = 215) P = 0.047


Patient with Diagnosis (%)

Lamivudine (n = 436)
Excluding 5 cases in yr1: HR = 0.47; P = 0.052
F I G U R E 1 Global distribution of HBV
10% and current therapies. (A) Worldwide
distribution of chronic HBV infection.
Placebo (B) Approved therapies for chronic HBV
infection: ETV (entecavir), PEG-­IFN
(pegylated-­interferon), TDF (tenofovir),
Lamivudine 5% LAM (lamivudine), LdT (telbivudine), TAF
(tenofovir alafenamide). (C) Long-­term
lamivudine therapy leads to reduction in
Time to Diagnosis (months) cirrhosis decompensation and HCC13,14
14783231, 2018, S1, Downloaded from https://onlinelibrary.wiley.com/doi/10.1111/liv.13656 by Cochrane Israel, Wiley Online Library on [02/12/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
SCHINAZI et al. | 105

incorporated, it terminates polymerization. For TFV, the innovation is serve to remove the phosphate group to form a nucleoside derivative,
the use of the acyclic phosphonate group, which achieves 2 functions: and (b) in contrast to other nucleoside analogues, this stable alteration
(a) this group is resistant to cellular esterases that otherwise might circumvents the need for the cellular addition of the alpha phosphate,
which is the rate-­limiting step in the synthesis of 2′-­deoxynucleoside
triphosphates, the active substrates for reverse transcriptase.
The phosphonate group of TFV differs from a normal phosphate
group in that a carbon is directly linked to the phosphorus. For a phos-
phate group, the phosphorus would be directly linked to an oxygen
atom in place of the carbon atom in the TFV diagram above. The phos-
phonate is referred to as acyclic because in naturally occurring nucle-
otides, the phosphate group is linked to a sugar moiety (cyclic, ribose
or deoxyribose), whereas this is absent in this drug. The nucleoside
analogues used for HBV treatment are cyclic nucleosides (ETV, lami-
vudine [LAM] and telbivudine). Adefovir (ADV), in spite of its different
characteristics, differs from TFV only by the lack of the methyl group
(Figure 1B).
More than 95% of persons treated with the highly potent TDF
and ETV achieve virological clearance. NAs are administered orally,
and tolerance is good. The safety of these drugs over lifelong ther-
F I G U R E 2 Schematic representation of mechanisms of HBV apy remains to be established. Regarding the risk of drug resistance,
replication and inhibition. The virus replication begins with the although common in the past with earlier, less potent NAs such as
attachment of the virion to hepatocyte cell surface receptor hNTCP. LAM and ADV, resistance has become extremely rare with TDF and
This step can be blocked with entry inhibitors such as myrcludex
ETV. Long-­term clinical data up to 6 years and beyond are emerging
B. Upon entry, the virion is released in the cytoplasm and the
for the newer NAs that are providing reassuring data on their efficacy
nucleocapsid travels to the nucleus where rcDNA enters the nucleus
and cccDNA is formed. Novel cccDNA formation, or maintenance and safety. There are large data that long-­term blockage of HBV repli-
of already formed cccDNA can theoretically be disrupted with small cation by the most potent drugs (ETV and TDF) results in an improved
molecule inhibitors or gene-­editing technology such as CRISPR/Cas9. long-­term survival with a decreased risk of progression to cirrhosis,
Viral mRNA and pregenomic RNA are transcribed from cccDNA. end-­stage liver disease and HCC. Furthermore, a study analysing liver
Inhibition of transcription suppresses viral gene expression. HBV DNA
histology in persons treated with TDF for 5 years demonstrated fibro-
also gets integrated into the host chromosome. Secretion inhibitors
can prevent the release of HBsAg, which can be independent of sis regression in the majority of patients and also cirrhosis reversal
the virus replication cycle. Capsid effectors/inhibitors can mislead (Figure 4).22,23 In addition, the cirrhosis reversal was observed during
proper nucleocapsid formation while nucleoside analogues inhibit treatment in 75% of patients with cirrhosis, probably associated with
reverse transcription inside the nucleocapsid. Secretion inhibitors a decreased risk of HCC and improved survival. Real-­world data with
can subsequently inhibit the release of enveloped virions. Finally,
immunomodulators such as therapeutic vaccines and TLR agonists
can enhance the immune response to chronic hepatitis B 12 11
Proportion of patients

10
AgHBe-positive (%)

10
8 6
6 5
T A B L E 1 Advantages and disadvantages of PEG-IFN and 4 3
2
nucleoside analogues 2
0
Nucleoside pegIFN LAM ADV ETV LdT TDF
Peg-­IFN analogues 3-4 years 2 years 4-5 years 2 years 3 years 5 years
12 11
Proportion of patients
AgHBe-negative (%)

Advantages Finite duration High efficacy 10


Higher rates of HBs loss Favourable 8
6 5
and/or seroconversion tolerability
4
No resistance Oral 2 0 0 0,5 0*
administration 0
pegIFN LAM ADV ETV LdT TDF
Disadvantages Poor tolerability Long life 3-4 years 2 years 4-5 years 2 years
duration
Moderate efficacy Unknown F I G U R E 3 HBsAg loss after therapy. Loss of HBsAg is the most
long-­term reliable indicator to measure a functional cure of hepatitis B. In
toxicity HBeAg(+) patients, HBsAg loss was around 11% with Peg-­IFN after
Risk of adverse events Costs 4 y, and around 10% after 5 y of TDF. In HBeAg(−) patients, HBsAg
loss was still around 11% with Peg-­IFN. However, no HBsAg loss was
Subcutaneous injection
observed after 2 y of TDF or ETV treatment15-17,22,23
14783231, 2018, S1, Downloaded from https://onlinelibrary.wiley.com/doi/10.1111/liv.13656 by Cochrane Israel, Wiley Online Library on [02/12/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
106 | SCHINAZI et al.

ETV and TDF in routine clinical practice are confirming the favourable open-­label, active-­controlled study, 740 patients with CHB were ran-
safety and excellent efficacy of these therapies. domly assigned to receive TDF plus PEG-­IFN for 48 weeks (group A),
TDF plus PEG-­IFN for 16 weeks followed by TDF for 32 weeks (group
B), TDF for 120 weeks (group C) or PEG-­IFN for 48 weeks (group D).
4 | TENOFOV IR ALAFENAMIDE (TAF) At week 72, 9.1% of subjects in group A had HBsAg loss compared
with 2.8% of subjects in group B, none of the subjects in group C and
TAF, as a single-­agent treatment, has been developed for CHB. It is a 2.8% of subjects in group D. A significantly higher proportion of sub-
novel prodrug of TFV with potent antiviral activity against HBV. The jects in group A had HBsAg loss than in group C (P < .001) or group
drug is more stable in plasma with more than 90% reduction in cir- D (P = .003; Figure 6). However, the proportions of subjects with
24
culating TFV level compared to TDF. The dose is once-­daily, oral, HBsAg loss did not differ significantly between group B and group
25 mg tablet. Chemical structures of TAF is provided in Figure 1B. In C (P = .47) or group D (P = .88). HBsAg loss in group A occurred in
the phase III clinical trials,25-27 treatment with TAF through 72 weeks HBeAg-­positive and HBeAg-­negative patients with all major viral gen-
demonstrated: viral suppression (HBV DNA < 29 IU/mL) similar to otypes. Finally, a significantly greater proportion of subjects receiving
TDF; higher rates of alanine aminotransferase (ALT) normalization; TDF plus PEG-­IFN for 48 weeks had HBsAg loss than those receiving
no resistance development in either treatment group at Week 48; TDF or PEG-­IFN alone.
rates of HBeAg loss and seroconversion similar to TDF in Study 110;
good tolerance in HBeAg-­negative and -­positive subjects; treatment-­
6 | NOVEL THERAPEUTIC STRATEGIES
emergent AEs similar to TDF; significantly less declines in hip and
IN DEVELOPMENT
spine bone mineral density (BMD) compared to TDF with improved
bone biomarkers; and significantly smaller decreases in estimated
­
6.1 | Entry inhibitors
glomerular filtration rate by Cockcroft-­Gault (eGFRCG) compared to
TDF, with improved markers of renal tubular function (Figure 5). The discovery of human sodium taurocholate cotransporting poly-
peptide (hNTCP) as the cellular receptor that mediates HBV fusion
has facilitated drug development efforts with regard to inhibition of
5 | RECENT DEVELOPMENTS REGARDING
HBV entry. Myrcludex B represents the most advanced therapeutic
APPROVED THERAPIES
agent that targets hNTCP-­mediated HBV entry into the cell (Table 2).
It is a synthetic 47 amino acid, N-­myristoylated lipopeptide that is de-
5.1 | Combination therapy (TDF plus PEG-­IFN)
rived from the amino acid sequence of L surface protein. By binding
In a recent study, HBsAg loss was evaluated in patients receiving to this receptor in a competitive manner, Myrcludex B excludes viral
28
the combination of TDF and PEG-­IFN for a finite duration. In an HBsAg binding, and thus inhibiting virus entry.29 Although Mycludex
B showed positive effects on virus control and intracellular cccDNA
p<0.001 accumulation,29 some serious adverse events were observed in phase
p<0.001 I clinical trials with healthy volunteers.30 Myrcludex B has also shown
100 Ishak score promise for the treatment of hepatitis D virus, which uses the same
6
90
hNTCP receptor as HBV.31 Another molecule of interest for inhibition
5
80
4 of HBV entry is irbesartan.32 Originally, this molecule was developed
70 3 against hypertension and received FDA approval in 1997. Because
60 2
irbesartan interferes with hNTCP receptors, it was also shown to be
Pa ents (%)

1
50
0 able to inhibit HBV virus entry in HepG2 cell culture.33 It remains to
40
be determined whether the addition of entry inhibitors to the current
30
arsenal of HBV therapeutics would provide any additional benefits,
20

10
since at least in theory, this class of inhibitors should be able to pro-
0 tect uninfected cells from HBV entry and subsequent establishment
Baseline Year 1 Year 5 of HBV cccDNA pool.34

F I G U R E 4 Fibrosis regression under TDF. Advanced liver fibrosis


and particularly cirrhosis were previously considered to be largely 6.2 | RNA interference
irreversible. Data from small studies suggest this may be possible.
The histological benefits of 5 y of TDF therapy in the largest study RNA interference is a relatively novel strategy that targets virus rep-
of paired liver biopsies in HBV over 5 y of therapy are shown. lication at the level of gene expression (Table 2). Briefly, short RNA
The histogram shows that the proportion of patients with Ishak
molecules are designed to target specific viral mRNA sequences.
stages 0-­2 increased over the study period, whereas, in contrast,
Upon entry into the cell, they hybridize to viral mRNA and the re-
the proportion with Ishak stages 4-­6 decreased, thus dramatically
illustrating a marked overall decrease in liver scarring for this sulting double-­stranded RNA is targeted for degradation.35 ARC-­520
cohort22,23 represents a leading siRNA molecule that when conjugated with
14783231, 2018, S1, Downloaded from https://onlinelibrary.wiley.com/doi/10.1111/liv.13656 by Cochrane Israel, Wiley Online Library on [02/12/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
SCHINAZI et al. | 107

cholesterol, can be delivered to the liver upon intravenous adminis- significantly controlled upon zinc finger nuclease delivery. However,
tration. Experiments on chimpanzees showed that ARC-­520 is highly they were unable to show specific mutations within cccDNA in a
effective at reducing circulating HBV DNA levels as well as serum reproducible manner.41 The efficacy of TALENs in reducing HBV
35,36
HBeAg and HBsAg. ARC-­520 has shown to be relatively well levels in cell culture was first demonstrated by Bloom et al in 2013
tolerated in a phase I clinical trial, although some hypersensitivity whereby targeting the S and C regions in cccDNA resulted in disrup-
reactions were observed. Therefore, the authors recommend future tion of 35% of cccDNA molecules in the HepG2 cell line.48 In agree-
37
co-­administration with antihistamines. Based on these promising ment with these findings, it was demonstrated that TALENs could
results, ARC-­520 has advanced to phase II trials. However, these also reduce the circulation of viral particle equivalents in the murine
trials were recently terminated because of formulation toxicities.38 hydrodynamic injection model48 and cause reductions in secreted
Interestingly, a recent study has identified a novel small molecule HBeAg levels.49 Use of CRISPR/Cas9 for treatment of HBV was first
inhibitor that can reduce viral messenger RNA (mRNA) and pgRNA evaluated in 2014.42 Through the simultaneous administration of
39
expression. Despite strong evidence that RG7834 does not func- multiple small guiding RNAs specific for various regions of the HBV
tion in as RNAi, it is not yet clear how this therapeutic agent can genome, several studies have demonstrated successful inhibition of
suppress HBV transcription in a virus-­specific manner. As well, it has virus replication and production of viral markers such as HBsAg.50-53 One
been shown that RG7834 can indeed reduce levels of viral antigens study also showed a marked decrease in cccDNA levels in HepAD38.
in the humanized mouse model, although it is still not clear whether This reduction was associated with rampant presence of mutations
this small molecule would also reduce HBsAg levels expressed from within viral DNA.40 Overall, preclinical studies with gene-­editing
integrated HBV DNA as seen in individuals with chronic hepatitis B. technologies have been promising. However, several challenges still
remain. For one, in vivo delivery methods that would maximize ef-
ficacy and minimize immune response to therapeutic gene editors
6.3 | Gene-­editing strategies
are still being developed. There is also a concern for viral escape
Recent advancement in novel sequence-­specific nuclease technology mutants as a result of treatment with sequence-­specific zinc finger
has led to the development of several novel therapeutic strategies nucleases, TALENs or CRISPR/Cas9 systems. Finally, adverse events
against HBV (Table 2). These include zinc finger nucleases, TALENs need to be carefully investigated with these agents, both in terms
(transcription activator-­like effector nucleases) and, CRISPR/Cas9 sys- of non-­specific cleavage of host chromosomes, and chromosome
tems, which through viral gene editing, have shown some success in destabilization as a result of cleavage of viral DNA integrants.44
reducing viral DNA and cccDNA levels in both cell culture and animal Despite these concerns, some gene-­editing agents such as EBT106
models.40-42 Most antiviral gene-­editing therapeutics act as mutagenic (Excision Biotherapeutics) are cautiously advancing towards clinical
agents that, upon cell entry, cause DNA damage at sequence-­specific development (https://www.excisionbio.comBiotherapeutics 2017,
sites within cccDNA. DNA repair via non-­homologous recombination posting date. [Online]).
at these sites is error-­prone and this in turn, results in generation of
insertion/deletions within cccDNA.43
6.4 | Secretion inhibitors
Zinc finger nucleases have successfully been used in tissue
culture to reduce HBV levels.44-47 For example Weber et al devel- In addition to infectious virion secretion, a large number of incomplete
oped a zinc finger nuclease that targeted the core and pol regions viral particles are released from infected hepatocytes. These include
of cccDNA. Using an adeno-­associated virus (AAV) delivery system, spherical and filamentous particles made up of HBsAg, also known as
they showed that HBV production from HepAD38 cells could be Australia antigen. It is hypothesized that these particles are released

HBeAg- HBeAg+

TAF: 92.6%
TDF: 92.1%
Proporon of Paents, % (95% CI)

TAF: 71.6%
TDF: 71.9%

Treatment difference: −0.9 (−7.0, 5.2); p=0.78


Treatment difference +0.6 (-5.3, +6.4); p=0.84

Weeks Weeks
F I G U R E 5 Efficacy of TAF in phase III
clinical trials. In the phase III clinical trials, ALT Normalizaon (AASLD Criteria)¤
in both HBeAg-­positive and HBeAg-­
negative chronic hepatitis B, treatment
Paents with ALT
Normalizaon, %

‡ ‡ †
‡ ‡
‡ ‡ ‡ TAF: 49.1%
with TAF through 72 wk demonstrated ‡ ‡
‡ ‡
TDF: 39.0%
† *p<0.05
comparable viral suppression (HBV †
†p<0.005
*
DNA < 29 IU/mL) to TDF and improved ‡p<0.001
rates of ALT normalization25,26
14783231, 2018, S1, Downloaded from https://onlinelibrary.wiley.com/doi/10.1111/liv.13656 by Cochrane Israel, Wiley Online Library on [02/12/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
108 | SCHINAZI et al.

in copious amounts to induce immune tolerance and exhaustion.54 Recent developments with regard to toll-­like receptor (TLR) ago-
Based on this, several inhibitors such as REP 2139 and REP 2165 have nists have also been shown to be effective in modulating the innate
been developed that inhibit the secretion of HBsAg containing sub- immune response against HBV. Activation of virus-­specific TLRs
particles (Table 2). Phase I clinical trials have shown positive results can lead to production of IFN-­α and -­β, both of which play import-
with regard to safety, tolerability and control of HBV viraemia. Based ant roles with regard to suppression of virus replication (See review
55
on these findings, Phase II clinical trials are underway. 59). Specifically, TLR-­7 plays a critical role in activating the immune
response within the liver environment without causing systemic in-
flammation.60-62 Stimulation of TLR-­8 as a result of viral infection was
6.5 | Polymerase inhibitors
shown to result in IFN-­γ production and subsequent virus control.63
Currently, nucleoside analogue inhibitors are the backbone of chronic At the same time, it was found that TLR-­8 function was impaired in
HBV treatment. As discussed elsewhere, when administered as mono- patients with chronic HBV infection.64 Based on this, several thera-
therapy, nucleoside analogues are quite successful at suppressing viral peutic strategies have been developed to harness the innate immune
replication, but do not eliminate the virus from the human body. There response in response to chronic hepatitis B. GS-­9620 is a TLR-­7 ag-
are currently several nucleoside analogue inhibitors in development onist that showed promising results in animal studies with chimpan-
that aim to provide therapeutic, pharmacological and tolerability im- zee65 and woodchuck models.66 Treatment with GS-­9620 resulted in
56
provements on the existing drug regimens. These include CMX-­157, reduction in circulating HBV DNA and woodchuck surface antigen.66
a novel prodrug of TAF and MIV-­210, a nucleoside analogue with Conversely, when GS-­9620 was administered to patients with chronic
57
antiviral activity in woodchucks. Another novel acyclic nucleoside hepatitis B in a phase II trial, it was found that the T cell response was
analogue is besifovir (LB80380), which in comparison to entecavir, improved in a transient manner, but no significant reduction in HBsAg
was shown to achieve similar therapeutic results in treatment-­naïve levels was detected. It was hypothesized that variations in dosing may
chronic hepatitis B patients.58 It remains to be seen whether these account for this discrepancy.67,68
novel nucleoside analogues, as mono or combination therapy, provide Development of therapeutic vaccines represents another recent
substantial advantages over current FDA-­approved therapies. strategy against chronic hepatitis B. It is hypothesized that the stimu-
lation of T cells with HBV antigens may result in the generation of an
effective and HBV-­specific adaptive immune response that may elimi-
6.6 | Immunomodulators
nate the chronic infection. Based on this, several therapeutic vaccines
An important characteristic of chronic hepatitis B is the development are currently being evaluated in the clinic. Examples include GS-­4774,
of immune exhaustion as a result of continuous exposure of T cells which consists of highly immunogenic recombinant HBcAg, HBsAg
to high levels of viral antigens. Overexpression of several inhibitory and HBx epitopes, ABX-­203 consisting of HBcAg and HBsAg epi-
receptors such as PD1 and CTLA-­4 has been shown to be associated topes,69 and INO-­1800 consisting of DNA plasmids coding for HBcAg
with T-­cell exhaustion. As such, it has been hypothesized that small and HBsAg. Other therapeutic vaccines currently in clinical trials are
molecules that inhibit PD-­1 expression, may result in T-­cell reactiva- listed in Table 2. Despite promising outcomes of recent immune ther-
tion, and subsequent mounting of an efficient immune response that apies, several potential challenges still remain. For one, manipulation
54
may be able to eliminate chronic hepatitis B. It is worth noting that of the innate or adaptive immune response may be accompanied by
most immune checkpoint inhibitors have the potential to be used in related adverse events in either a local, or systemic manner. This was
any number of diseases where immune reactivation is desired. These most acutely observed in recent clinical trials involving PD-­1 agonists
include important human diseases such as melanoma, lymphoma and and other immune-­modulatory therapies whereby the liver and the en-
54
other cancers. A list of current PD-­1 and other immune checkpoint docrine system were negatively affected.70,71
inhibitors is provided in Table 2.

HBsAg loss
0.15
48 weeks 72 weeks
0.14
0.13
0.12 F I G U R E 6 Combination therapy with
Patients with HBsAg loss

0.11
0.10
TDF and PEG-­IFN. At week 72, 9.1%
Kaplan-Meier (%)

TDF + PEG 48 weeks. 9.0%


0.09 of subjects in group A had HBsAg loss
0.08 compared with 2.8% of subjects in group
0.07 p = 0.003
0.06
B, none of the subjects in group C and
0.05 PEG 48 weeks. 2.8% p < 0.001 2.8% of subjects in group D. A significantly
0.04 TDF + PEG 16 weeks. →TDF 32 weeks 2.8%
p = NS higher proportion of subjects in group A
0.03
0.02 p = NS
had HBsAg loss than in group C (P < .001)
0.01 TDF 120 weeks. 0%
or group D (P = .003). However, the
0.00 proportions of subjects with HBsAg loss
0 4 8 12 16 20 24 28 32 36 40 44 48 52 56 60 64 68 72 did not differ significantly between group B
Weeks and group C (P = .47) or group D (P = .88)28
14783231, 2018, S1, Downloaded from https://onlinelibrary.wiley.com/doi/10.1111/liv.13656 by Cochrane Israel, Wiley Online Library on [02/12/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
SCHINAZI et al. | 109

virions or amplified from encapsidated rcDNA from the hepatocyte


6.7 | Inhibitors of cccDNA formation
cytoplasm. Because hepatocytes have a long half-­life (>6-­months or
The major strategy to “cure” HBV is to silence or eliminate cccDNA even years), the half-­life of cccDNA is also relatively long; therefore,
from the hepatocyte nucleus. cccDNA is the main element of viral elimination of cccDNA by hepatocyte turnover is not a major means
replication cycle as it serves as the template for transcription of all of clearance.72 Reverse transcriptase nucleoside analogue (NA) inhibi-
viral RNAs including pgRNA, and subsequently the progeny HBV DNA tors have a negligible effect on cccDNA formation, stability or ampli-
genomes are formed. cccDNA is either originated from new incoming fication, and therefore HBV commonly rebounds after cessation of

TABLE 2 Summary of recent anti-­HBV antiviral agents in clinical and preclinical development

Comments and
Drug class Antiviral agent Status Developer references

Entry inhibitors Myrcludex B Phase II MYR GmbH/Hepatera Bogomolov et al (2016)87


Irbesartan (Avapro) FDA-­approved Sanofi Research Approved for hyperten-
sion. Wang et al (2015)
33

Inhibitors of viral mRNA ALN-­HBV Phase I-­II Alnylam siRNA


ARC-­520 Terminated Arrowhead pharmaceuticals siRNA
ARB-­1467 Phase II Arbutus Biopharma siRNA88,89
ARB-­1740 Preclinial Arbutus Biopharma siRNA90
RO7020322 (RG7834) Phase I Roche Small molecule mRNA
inhibitor. Mueller et al
(2017)39
Ionis HBVRx Phase I Ionis Pharma/GSK Antisense molecule
(GSK3228836)
IONIS-­HBVLRx Phase I Ionis Pharma/GSK Antisense molecule91
(GSK33389404)
AB-­452 Preclinical Arbutus Biopharma RNA destabilizer92
Gene editing EBT106 Preclinical Excision Biotherapeutics CRISPR/Cas9 system93
Anti-­HBV sgRNA Preclinical CRISPR/Cas9 system.
Kennedy et al (2015)40
TALEN Preclinical Dryer et al (2016)94
Zinc finger nuclease Preclinical Weber et al (2014)41
HBsAg secretion inhibitor REP2139 & REP2165 Phase II Replicor Bazinet et al (2017)55,95
BM601 Preclinical Xu et al (2014)96
Nucleoside analogue LB80380 (besifovir, Phase III/Approved in S. Illong Pharmaceutical Yuen et al (2015)58
inhibitors besivo) Korea
CMX-­157 Phase IIa ContraVir Prodrug of tenofovir.
Painter et al (2007)56
Capsid assembly inhibitors/ GLS-­4 Phase II HEC Pharm, Sunshine China-­CFDA
modulators NVR 3-­778 Phase Ia Novira Pharmaceuticals/ Klumpp et al (2017)78
Janssen
BAY41-­4109 Phase I Berke et al (2017)79
JNJ56136379 Phase I JnJ Janssen NCT02662712. Zoulim
et al (2017)97
Core protein allosteric Phase I (ABI-­H0731) Assembly Biosciences ABI-­H0731 -­
modulators (CpAMs) IND enabling (ABI-­ NCT02908191
H2158) Huang et al AASLD
Clinical candidate (2017)98
(ABI-­Nx)
AB-­423 Phase I Arbutus Biopharma Eley et al AASLD (2017)99
AB-­506 IND enabling Arbutus Biopharma Mani et al AASLD
(2017)80

(Continues)
14783231, 2018, S1, Downloaded from https://onlinelibrary.wiley.com/doi/10.1111/liv.13656 by Cochrane Israel, Wiley Online Library on [02/12/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
110 | SCHINAZI et al.

TABLE 2 (Continued)

Comments and
Drug class Antiviral agent Status Developer references

cccDNA inhibitor Disubstituted Preclinical Cai et al (2012)100


sulphonamides
Therapeutic vaccine GS-­4774 Phase II GlobeImmune Recombinant X, S & C
epitopesa,101
ABX-­203 Phase II/III Abivax Recombinant S & C
epitopes
TG-­1050 Phase I Transgene Non-­replicative
adenovirus. C, pol & S
epitopes. Levin (2017)102
INO-­1800 Phase I Inovio DNA plasmids encoding S
&C
TLR agonists GS-­9620 Phase II Gilead Sciences TLR-­7 agonist. Boni et al
(2016)68
RO6864018 (RG7795) Phase II Roche TLR-­7 agonist
GS-­9688 Phase I Gilead Sciences TLR-­8 agonist
Immuno-­modulators PD-­1/PDL-­2 mAb Preclinical Merck Sharp and Dohme;
Bristol-­Myers Squibb
CTLA-­4 mAb Preclinical
SB9200 (Iranigivir) Phase II Spring Bank Pharmaceuticals RIG-­I and NOD-­2
agonist103
a
X = HBx, S = HBsAg, C = HBcAg.

treatment with NA.73 In addition, the presence of HBsAg in serum of (epidrugs) may represent a novel class of antivirals for treatment of
chronic hepatitis B (CHB) individuals with undetectable levels of HBV chronic hepatitis B. HBx binds to cccDNA and plays an important role
DNA after or during NA therapy strongly suggest that this protein is on activation of viral gene transcription, leading to high level of virus
transcribed either from cccDNA or integrated HBV DNA in the nu- replication. In contrast, inhibition of HBx binding to cccDNA leads to
cleus of hepatocytes. Enormous advances in understanding the HBV suppression of its transcription and virus replication by modifying the
replication cycle have facilitated the development of novel antiviral epigenetic regulation of cccDNA function with host restriction fac-
therapies targeting multiple steps of viral replication with numerous tors, including Smc5/6 complex and protein arginine methyltrans-
mechanistic actions. Both in vitro and in vivo models for testing these ferase PRMT1. HBV core inhibitors targeting cccDNA-­bound HBc
novel antiviral therapies have also significantly improved and mutually may potentially alter the epigenetics of cccDNA and in combination
open a new era for extinction of hepatitis B chronic infection.74 with HBx inhibitors could in theory lead to a stronger synergistic ef-
Several in vitro studies demonstrate that it is possible to degrade fect on cccDNA transcriptional silencing and HBV replication.76
cccDNA in the nucleus of hepatocytes with minimal hepatotoxicity
effect. As discussed above, programmable RNA-­guided DNA endonu-
6.9 | Nucleocapsid assembly inhibitors or modulators
cleases, including CRISPR/Cas9 system have shown in both cell and
mouse models the potential to serve as effective tool for the deple- Novel direct acting core assembly inhibitors or modulators have an
tion of the cccDNA pool in HBV-­infected hepatocytes (Table 2).40,44,52 important impact in HBV replication cycle. They can interfere with
Moreover, up-­regulation of APOBEC3A/B deaminase by several cyto- several steps of the HBV replication cycle including capsid assem-
kines including IFN-­α, IFN-­γ, tumour necrosis factor-­α and lymphotox- bly, reverse transcription, as well as pgRNA and polymerase protein
in-­β receptor agonist can cause partial degradation of cccDNA without packaging. These molecules may also affect intracellular trafficking
hepatotoxicity.75 These targets aim to prevent cccDNA formation by of relaxed circular DNA in the nucleus, virus assembly by disrupt-
damaging or destroying it. However, more research needs to be done ing its interaction with envelope proteins, and core binding to cc-
regarding off-­target effects of gene-­editing applications or the in vivo cDNA.77 Two main classes of core assembly modulators have been
delivery efficiency before they advance to humans. developed, including class I heteroarylpyrimidines (HAP), and class II
phenylpropenimides (PP) or sulfamoylbenzamide (SBA) and deriva-
tives (Table 2). NVR 3-­778 is a SBA compound developed by Novira
6.8 | Silencing cccDNA with epigenetic drugs
(later acquired by Johnson & Johnson, New Brunswick, NJ, USA) that
Viral cccDNA is a mini-­chromosome that is inherently stable and si- is in phase Ia clinical trial (NCT02112799 & NCT02401737), and
lencing its activity by epigenetic modifications with epigenetic drugs has shown synergistic effect when in combination with PEG-IFN.
14783231, 2018, S1, Downloaded from https://onlinelibrary.wiley.com/doi/10.1111/liv.13656 by Cochrane Israel, Wiley Online Library on [02/12/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
SCHINAZI et al. | 111

TABLE 3 Who to treat?

Phase Immune tolerant HBeAg-­positive CHB Inactive carrier HBeAg-­negative CHB

HBeAg status Positive Positive Negative Negative


ALT Normal Elevated Normal Elevated (fluctuating)
HBV DNA Very high > 200 000 IU/mL >2000 IU/mL <2000 IU/mL >2000 IU/mL (fluctuating)
Histology Normal or mild inflammation & Inflammation & fibrosis: Normal or mild Inflammation and fibrosis:
mild fibrosis moderate to severe inflammation moderate to severe
Disease progression Low Moderate to high No, very low Moderate to high
Treatment Not indicated Indicated Not indicated Indicated

Evaluation of NVR 3-­788 with PEG-­IFN in mice with humanized liver vs >3-­fold the upper limit of normal). PS2 analysed data on only age
confirmed inhibition of both HBV DNA replication and HBV RNA pro- and level of hepatitis B surface antigen. Our system differentiated
78
duction. Recent in vitro studies with JNJ-­632 (SBA) or Bay41-­409 persons with no or mild fibrosis (F0-­F1) from those with marked or
(HAP) compounds in human primary hepatocytes showed that both severe (F2-­F4) fibrosis with a high positive predictive value (PPV).
compounds inhibited early and late steps of viral replication by (a) The high level of specificity for the identification of non-­severe
preventing formation of cccDNA, and (b) reducing intracellular HBV fibrosis (F0-­F2) limits the risk of overlooking patients with severe
RNA levels and HBe and HBs antigens production when these com- fibrosis (F3-­F4).
pounds were added at the time of infection.79 Clinical studies are In another study, the expression of 13 fibrosis-­related microRNAs
being conducted with 3 other potent core assembly modulators, (miRNAs; miR-­20a, miR-­21, miR-­27a, miR-­27b, miR-­29a, miR-­29c,
including morphothiadine mesilate GLS4 (HAP) in phase II (HEC miR-­92a, miR-­122, miR-­146a, miR-­155, miR-­221, miR-­222 and miR-­
Pharm/Sunshie, Guangdong,­ China-­CFDA), JNJ56136379 in phase 224) was analysed in 194 serums and 177 liver biopsies of patients
I (JnJ Janssen, New Brunswick, NJ, USA -­ NCT02662712), and the with either CHB or chronic hepatitis C (CHC) to develop models to
first-­generation Core protein Allosteric Modifier (CpAM) in phase I diagnose advanced fibrosis and cirrhosis (Metavir F3-­F4).82 In CHB
Assembly Biosciences (NCT02662712). New improved second gen- subjects, the model (serum miR-­122, serum miR-­222, platelet count
eration of CpAMs that induce formation of aberrant capsids were re- and alkaline phosphatase) was more accurate than APRI and FIB-­4 to
cently developed that show favourable preclinical profile in inhibiting discriminate in between mild and moderate fibrosis (F1-­F2) and F3-­
4 HBV markers, including secretion of HBeAg and HBsAg, pgRNA F4 (AUC of CHB model: 0.85 vs APRI: 0.70 and FIB-­4: 0.81). In CHC
and cccDNA (Table 2). A new generation of capsid assembly inhibi- persons, the model (hepatic miR-­122, hepatic miR-­224, platelet count,
tor AB-­506 (Arbutus Biopharma, Burnaby, BC, Canada) was devel- albumin and alanine aminotransferase) was more accurate than both
oped that increases thermal stability to core protein compared to APRI and FIB-­4 to discriminate in between patients with F3-­F4 and
first-­generation inhibitors, and it also inhibits pgRNA encapsidation F1-­F2 (AUC of the CHC model = 0.93 vs APRI: 0.86 and FIB-­4: 0.79).
in HepAD38 cells and demonstrates potent in vivo activity in mouse Most of the miRNAs tested were differentially expressed in patients
80
model. AB-­423 (Arbutus Biopharma) is another inhibitor of capsid with CHB and CHC. In particular, serum miR-­122 was 28-­fold higher
assembly that has demonstrated potent inhibition of HBV replication in patients with CHB than in those with CHC. Both CHB and CHC
by blocking pgRNA encapsidation. This compound is in phase I clinical models may help for the diagnosis of advanced fibrosis and cirrhosis
study (Table 2).81 (F3-­F4).

7 | WHO TO TREAT: THE IMPORTANCE OF 8 | HBSAG QUANTIFICATION: A NEW


FIBROSIS AS PROGNOSIS FACTOR TOOL FOR HBV MONITORING

Fibrosis is the most important prognosis predictor of survival. Quantifying HBsAg is certainly an important new tool for predict-
Therefore, subjects at risk of fibrosis progression or with advanced ing the severity of disease and distinguishing inactive carriers from
fibrosis may be prioritized for treatment (Table 3). Several new persons with HBeAg-­negative chronic active hepatitis. In this way,
markers have been developed to assess fibrosis. For instance, our HBsAg measurement helps tailor follow-­up and treatment manage-
group in France developed a simple scoring system to determine ment.83,84 In addition, a decline in quantitative HBsAg during therapy
the severity of fibrosis in persons with genotypes B or C HBV infec- is a strong predictor of SVR after PEG-­IFN therapy and of the prob-
tion who are HBeAg-­positive.28 We developed 2 prediction scoring ability of HBsAg loss, which is the ultimate goal of therapy. In patients
systems (PSs). PS1 analysed data on HBV genotype (B vs C), patient treated with nucleoside analogues, a decline in HBsAg levels is also a
age (<30 vs ≥30 years), level of hepatitis B surface antigen (≤17 500 predictor of HBsAg loss, allowing therapy to be discontinued. A low
vs >17 500 IU/mL) and level of alanine aminotransferase (≤3-­fold HBsAg level is predictive of favourable response. Baseline HBsAg
14783231, 2018, S1, Downloaded from https://onlinelibrary.wiley.com/doi/10.1111/liv.13656 by Cochrane Israel, Wiley Online Library on [02/12/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
112 | SCHINAZI et al.

levels are reliable predictors of sustained response and several data 9. Blondot M-L, Bruss V, Kann M. Intracellular transport and egress of
confirm this result. Baseline HBsAg levels are significantly lower in hepatitis B virus. J Hepatol. 2016;64:S49‐S59.
10. Alter H, Block TM, Brown N, et al. A research agenda for cur-
patients who achieve an SVR than in non-­responders, both in HBeAg-­
ing chronic hepatitis B virus infection. Hepatology. 2017; in press.
positive and HBeAg-­negative patients.85,86 https://doi.org/10.1002/hep.29509.
11. EASL. Clinical practice guidelines on the management of hepatitis B
virus infection. J Hepatol. 2017;67:370‐398.
9 | CONCLUSIONS 12. Terrault NA, Bzowej NH, Chang KM, et al. AASLD guidelines for
treatment of chronic hepatitis B. Hepatology. 2016;63:261‐283.
13. Lai CL, Chien RN, Leung NW, et al. A one-­year trial of lamivudine for
The recent success in eliminating hepatitis C virus infection from the chronic hepatitis B. Asia hepatitis lamivudine study group. N Engl J
liver using safe and potent combinations of direct antiviral agents Med. 1998;339:61‐68.
has raised hopes that this will also be possible for HBV infections. 14. Liaw YF, Sung JJ, Chow WC, et al. Lamivudine for patients with
chronic hepatitis B and advanced liver disease. N Engl J Med.
Lifetime use of current anti-­HBV agents can decrease and suppress
2004;351:1521‐1531.
the HBV replication to undetectable levels, but it does not produce
15. Lau GK, Piratvisuth T, Luo KX, et al. Peginterferon Alfa-­2a, lamivu-
a cure. To have a sustained clinical cure, the discovery and develop- dine, and the combination for HBeAg-­positive chronic hepatitis B. N
ment of novel combined modalities that prevent virus rebound on Engl J Med. 2005;352:2682‐2695.
discontinuation of therapy is needed. There is also a need to silence 16. Marcellin P, Lau GK, Bonino F, et al. Peginterferon alfa-­2a alone, lami-
vudine alone, and the two in combination in patients with HBeAg-­
or eliminate cccDNA, and decrease or eliminate HBsAg, HBeAg,
negative chronic hepatitis B. N Engl J Med. 2004;351:1206‐1217.
pgRNA and possibly other HBV markers to undetectable levels. In 17. Dienstag JL. Hepatitis B virus infection. N Engl J Med.
addition, restoring or increasing the host immune response should 2008;359:1486‐1500.
lead to a functional and perhaps an absolute cure. Once these per- 18. Wursthorn K, Jung M, Riva A, et al. Kinetics of hepatitis B surface
antigen decline during 3 years of telbivudine treatment in hepatitis B
sons are cured, theoretically they can be vaccinated against HBV to
e antigen-­positive patients. Hepatology. 2010;52:1611‐1620.
prevent reinfection. Only then, we will have all the tools necessary 19. Marcellin P, Bonino F, Yurdaydin C, et al. Hepatitis B surface antigen
to eradicate HBV globally. levels: association with 5-­year response to peginterferon alfa-­2a in
hepatitis B e-­antigen-­negative patients. Hepatol Int. 2013;7:88‐97.
20. De Clercq E, Holy A. Acyclic nucleoside phosphonates: a key class of
CO NFLI CTS OF I NTE RE ST antiviral drugs. Nat Rev Drug Discov. 2005;4:928‐940.
21. De Clercq E. The clinical potential of the acyclic (and cyclic) nucle-
Raymond Schinazi is the founder and major shareholder of Cocrystal oside phosphonates: the magic of the phosphonate bond. Biochem
Pharma, Inc. Tarik Asselah is a speaker and investigator for BMS, Pharmacol. 2011;82:99‐109.
Janssen, Gilead, Roche and Merck. Maryam Ehteshami and Leda 22. Marcellin P, Gane E, Buti M, et al. Regression of cirrhosis during
treatment with tenofovir disoproxil fumarate for chronic hepatitis B:
Bassit have no conflicts of interest to declare.
a 5-­year open-­label follow-­up study. Lancet. 2013;381:468‐475.
23. Marcellin P, Asselah T. Long-­term therapy for chronic hepatitis B:
hepatitis B virus DNA suppression leading to cirrhosis reversal. J
O RCI D
Gastroenterol Hepatol. 2013;28:912‐923.
Tarik Asselah http://orcid.org/0000-0002-0024-0595 24. Murakami E, Wang T, Park Y, et al. Implications of efficient hepatic
delivery by tenofovir alafenamide (GS-­7340) for hepatitis B virus
therapy. Antimicrob Agents Chemother. 2015;59:3563‐3569.
25. Buti M, Gane E, Seto WK, et al. Tenofovir alafenamide versus
REFERENCES
tenofovir disoproxil fumarate for the treatment of patients with
1. WHO. Fact sheet No. 204. http://www.who.int/mediacentre/fact- HBeAg-­negative chronic hepatitis B virus infection: a randomised,
sheets/fs204/en/. Accessed October 24, 2012. double-­blind, phase 3, non-­inferiority trial. Lancet Gastroenterol
2. Marcellin P, Asselah T, Boyer N. Fibrosis and disease progression in Hepatol. 2016;1:196‐206.
hepatitis C. Hepatology. 2002;36:S47‐S56. 26. Chan HL, Fung S, Seto WK, et al. Tenofovir alafenamide versus teno-
3. Yan H, Zhong G, Xu G, et al. Sodium taurocholate cotransporting fovir disoproxil fumarate for the treatment of HBeAg-­positive chronic
polypeptide is a functional receptor for human hepatitis B and D hepatitis B virus infection: a randomised, double-­blind, phase 3, non-­
virus. Elife. 2012;1:e00049. inferiority trial. Lancet Gastroenterol Hepatol. 2016;1:185‐195.
4. Schreiner S, Nassal M. A role for the host DNA damage response 27. Abdul Basit S, Dawood A, Ryan J, et al. Tenofovir alafenamide for
in hepatitis B virus cccDNA formation—and beyond? Viruses. the treatment of chronic hepatitis B virus infection. Expert Rev Clin
2017;9:125. Pharmacol. 2017;10:707‐716.
5. Zeisel MB, Lucifora J, Mason WS, et al. Towards an HBV cure: state-­ 28. Marcellin P, Ahn SH, Ma X, et al. Combination of tenofovir disoproxil
of-­the-­art and unresolved questions-­report of the ANRS workshop fumarate and peginterferon alpha-­2a increases loss of hepatitis B
on HBV cure. Gut. 2015;64:1314‐1326. surface antigen in patients with chronic hepatitis B. Gastroenterology.
6. Nassal M. HBV cccDNA: viral persistence reservoir and key obstacle 2016;150:134‐144 e110.
for a cure of chronic hepatitis B. Gut. 2015;64:1972‐1984. 29. Volz T, Allweiss L, Ben MM, et al. The entry inhibitor Myrcludex-­B ef-
7. Tu T, Budzinska MA, Shackel NA, et al. HBV DNA integration: molec- ficiently blocks intrahepatic virus spreading in humanized mice pre-
ular mechanisms and clinical implications. Viruses. 2017;9:75. viously infected with hepatitis B virus. J Hepatol. 2013;58:861‐867.
8. Yim HJ, Lok AS. Natural history of chronic hepatitis B virus infec- 30. Blank A, Markert C, Hohmann N, et al. First-­in-­human application of
tion: what we knew in 1981 and what we know in 2005. Hepatology. the novel hepatitis B and hepatitis D virus entry inhibitor myrcludex
2006;43:S173‐S181. B. J Hepatol. 2016;65:483‐489.
14783231, 2018, S1, Downloaded from https://onlinelibrary.wiley.com/doi/10.1111/liv.13656 by Cochrane Israel, Wiley Online Library on [02/12/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
SCHINAZI et al. | 113

31. Rizzetto M, Smedile A. Pegylated interferon therapy of chronic hep- 52. Seeger C, Sohn JA. Targeting Hepatitis B Virus With CRISPR/Cas9.
atitis D: in need of revision. Hepatology. 2015;61:1109‐1111. Mol Ther Nucleic Acids. 2014;3:e216.
32. Verrier ER, Colpitts CC, Sureau C, et al. Hepatitis B virus receptors 53. Ramanan V, Shlomai A, Cox DB, et al. CRISPR/Cas9 cleavage of viral
and molecular drug targets. Hepatol Int. 2016;10:567‐573. DNA efficiently suppresses hepatitis B virus. Sci Rep. 2015;5:10833.
33. Wang XJ, Hu W, Zhang TY, et al. Irbesartan, an FDA approved drug 54. Arends JE, Lieveld FI, Ahmad S, et al. New viral and immunological
for hypertension and diabetic nephropathy, is a potent inhibitor for targets for hepatitis B treatment and cure: a review. Infect Dis Ther.
hepatitis B virus entry by disturbing Na(+)-­dependent taurocholate 2017;6:461‐476.
cotransporting polypeptide activity. Antiviral Res. 2015;120:140‐146. 55. Bazinet M, Pantea V, Cebotarescu V, et al. Safety and efficacy of REP
34. Boucle S, Bassit L, Ehteshami M, et al. Toward elimination of hepati- 2139 and pegylated interferon alfa-­2a for treatment-­naive patients
tis B virus using novel drugs, approaches, and combined modalities. with chronic hepatitis B virus and hepatitis D virus co-­infection (REP
Clin Liver Dis. 2016;20:737‐749. 301 and REP 301-­LTF): a non-­randomised, open-­label, phase 2 trial.
35. Gish RG, Yuen MF, Chan HL, et al. Synthetic RNAi triggers and their Lancet Gastroenterol Hepatol. 2017;2:877‐889.
use in chronic hepatitis B therapies with curative intent. Antiviral Res. 56. Painter GR, Almond MR, Trost LC, et al. Evaluation of hexadecyloxy­
2015;121:97‐108. propyl-­9-­R-­[2-­(Phosphonomethoxy)propyl]-­ adenine, CMX157,
36. Sebestyen MG, Wong SC, Trubetskoy V, et al. Targeted in vivo de- as a potential treatment for human immunodeficiency virus type
livery of siRNA and an endosome-­releasing agent to hepatocytes. 1 and hepatitis B virus infections. Antimicrob Agents Chemother.
Methods Mol Biol. 2015;1218:163‐186. 2007;51:3505‐3509.
37. Schluep T, Lickliter J, Hamilton J, et al. Safety, tolerability, and phar- 57. Michalak TI, Zhang H, Churchill ND, et al. Profound antiviral effect
macokinetics of ARC-­520 injection, an RNA interference-­based of oral administration of MIV-­210 on chronic hepadnaviral infection
therapeutic for the treatment of chronic hepatitis B virus infection, in a woodchuck model of hepatitis B. Antimicrob Agents Chemother.
in healthy volunteers. Clin Pharmacol Drug Dev. 2017;6:350‐362. 2009;53:3803‐3814.
38. Peters MG, Locarnini S. New direct-­acting antiviral agents and immu- 58. Yuen MF, Ahn SH, Lee KS, et al. Two-­year treatment outcome of
nomodulators for hepatitis B virus infection. Gastroenterol Hepatol. chronic hepatitis B infection treated with besifovir vs. entecavir: re-
2017;13:348‐356. sults from a multicentre study. J Hepatol. 2015;62:526‐532.
39. Mueller H, Wildum S, Luangsay S, et al. A novel orally available small 59. Bertoletti A, Ferrari C. Innate and adaptive immune responses in
molecule that inhibits hepatitis B virus expression. J Hepatol. 2017; chronic hepatitis B virus infections: towards restoration of immune
in press. https://doi.org/10.1016/j.jhep.2017.10.014. control of viral infection. Gut. 2012;61:1754‐1764.
40. Kennedy EM, Bassit LC, Mueller H, et al. Suppression of hepatitis B virus 60. Funk E, Kottilil S, Gilliam B, et al. Tickling the TLR7 to cure viral hep-
DNA accumulation in chronically infected cells using a bacterial CRISPR/ atitis. J Transl Med. 2014;12:129.
Cas RNA-­guided DNA endonuclease. Virology. 2015;476:196‐205. 61. Gane EJ, Lim YS, Gordon SC, et al. The oral toll-­like receptor-­7 ag-
41. Weber ND, Stone D, Sedlak RH, et al. AAV-­mediated delivery of zinc onist GS-­9620 in patients with chronic hepatitis B virus infection. J
finger nucleases targeting hepatitis B virus inhibits active replication. Hepatol. 2015;63:320‐328.
PLoS One. 2014;9:e97579. 62. Rebbapragada I, Birkus G, Perry J, et al. Molecular determinants of
42. Lin SR, Yang HC, Kuo YT, et al. The CRISPR/Cas9 system facilitates GS-­9620-­dependent TLR7 activation. PLoS One. 2016;11:e0146835.
clearance of the intrahepatic HBV templates in vivo. Mol Ther Nucleic 63. Jo J, Tan AT, Ussher JE, et al. Toll-­like receptor 8 agonist and bacteria
Acids. 2014;3:e186. trigger potent activation of innate immune cells in human liver. PLoS
43. Bloom K, Ely A, Arbuthnot P. A T7 endonuclease I assay to detect Pathog. 2014;10:e1004210.
talen-­mediated targeted mutation of HBV cccDNA. Methods Mol 64. Deng G, Ge J, Liu C, et al. Impaired expression and function of TLR8
Biol. 2017;1540:85‐95. in chronic HBV infection and its association with treatment re-
44. Ely A, Moyo B, Arbuthnot P. Progress with developing use of gene sponses during peg-­IFN-­alpha-­2a antiviral therapy. Clin Res Hepatol
editing To cure chronic infection with hepatitis B virus. Mol Ther. Gastroenterol. 2017;41:386‐398.
2016;24:671‐677. 65. Lanford RE, Guerra B, Chavez D, et al. GS-­9620, an oral agonist
45. Cradick TJ, Keck K, Bradshaw S, et al. Zinc-­finger nucleases as a of Toll-­like receptor-­7, induces prolonged suppression of hepati-
novel therapeutic strategy for targeting hepatitis B virus DNAs. Mol tis B virus in chronically infected chimpanzees. Gastroenterology.
Ther. 2010;18:947‐954. 2013;144:1508‐1517.
46. Zimmerman KA, Fischer KP, Joyce MA, et al. Zinc finger proteins de- 66. Menne S, Tumas DB, Liu KH, et al. Sustained efficacy and seroconver-
signed to specifically target duck hepatitis B virus covalently closed sion with the Toll-­like receptor 7 agonist GS-­9620 in the Woodchuck
circular DNA inhibit viral transcription in tissue culture. J Virol. model of chronic hepatitis B. J Hepatol. 2015;62:1237‐1245.
2008;82:8013‐8021. 67. Janssen HLA, Kim YJ, Ferrari C, et al. Safety, efficacy and pharma-
47. Zhao X, Zhao Z, Guo J, et al. Creation of a six-­fingered artificial tran- codynamics of vesatolimod (GS-­9620) in virally-­suppressed pa-
scription factor that represses the hepatitis B virus HBx gene inte- tients with chronic hepatitis B. J Hepatol. 2017; in press. https://doi.
grated into a human hepatocellular carcinoma cell line. J Biomol Screen. org/10.1016/j.jhep.2017.10.027.
2013;18:378‐387. 68. Boni C, Vecchi A, Rossi M, et al. TLR-­7 agonist GS-­9620 can improve
48. Bloom K, Ely A, Mussolino C, et al. Inactivation of hepatitis B virus HBV-­specific T cell and NK cell responses in nucleos(t)ide suppressed
replication in cultured cells and in vivo with engineered transcription patients with chronic hepatitis B. Hepatology. 2016;64(S1):7A.
activator-­like effector nucleases. Mol Ther. 2013;21:1889‐1897. 69. Wedemeyer HA, Sukeepaisarnjaroen W, Tangkijvanich P, et al. LBP-­
49. Chen J, Zhang W, Lin J, et al. An efficient antiviral strategy for target- 518 therapeutic vaccination of chronic hepatitis B patients with
ing hepatitis B virus genome using transcription activator-­like effec- ABX203 (NASVAC) to prevent relapse after stopping NUCs: con-
tor nucleases. Mol Ther. 2014;22:303‐311. trasting timing rebound between tenofovir and entecavir. J Hepatol.
50. Zhen S, Hua L, Liu YH, et al. Harnessing the clustered regularly inter- 2017;66:S101.
spaced short palindromic repeat (CRISPR)/CRISPR-­associated Cas9 70. Corsello SM, Barnabei A, Marchetti P, et al. Endocrine side effects
system to disrupt the hepatitis B virus. Gene Ther. 2015;22:404‐412. induced by immune checkpoint inhibitors. J Clin Endocrinol Metab.
51. Wang J, Xu ZW, Liu S, et al. Dual gRNAs guided CRISPR/Cas9 2013;98:1361‐1375.
system inhibits hepatitis B virus replication. World J Gastroenterol. 71. Postow MA. Managing immune checkpoint-blocking antibody side
2015;21:9554‐9565. effects. Am Soc Clin Oncol Educ Book. 2015;76‐83.
14783231, 2018, S1, Downloaded from https://onlinelibrary.wiley.com/doi/10.1111/liv.13656 by Cochrane Israel, Wiley Online Library on [02/12/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
114 | SCHINAZI et al.

72. Allweiss L, Dandri M. The role of cccDNA in HBV maintenance. chronic hepatitis B infection. The Liver Meeting AASLD, Washington,
Viruses. 2017;9:156. DC, USA; 2017. Abstract 929.
73. Seto WK, Lam YF, Fung J, et al. Changes of HBsAg and HBV DNA 91. Han KKJ, Elston R, Oliver S, et al. Safety, tolerability and pharmaco-
levels in Chinese chronic hepatitis B patients after 5 years of enteca- kinetics of GSK3389404, an antisense oligonucleotide for the treat-
vir treatment. J Gastroenterol Hepatol. 2014;29:1028‐1034. ment of chronic hepatitis B (CHB) infection: a randomized, double
74. Lok AS, Zoulim F, Dusheiko G, et al. Hepatitis B cure: from discovery infection: a randomized, double-blind, placebo-controlled, dose-es-
to regulatory approval. J Hepatol. 2017;67:847‐861. calation, first time in human study. The Liver Meeting AASLD,
75. Lucifora J, Xia Y, Reisinger F, et al. Specific and nonhepato- Washington, DC, USA; 2017. Abstract 918.
toxic degradation of nuclear hepatitis B virus cccDNA. Science. 92. Gotchev D. Identification and characterization of HBV RNA destabi-
2014;343:1221‐1228. lizers in vitro and in vivo. The Liver Meeting, Washington, DC, USA;
76. Hong X, Kim ES, Guo H. Epigenetic regulation of hepatitis B virus 2017. Abstract 923.
covalently closed circular DNA: implications for epigenetic therapy 93. Moyo B, Bloom K, Scott T, et al. Advances with using CRISPR/Cas-­
against chronic hepatitis B. Hepatology. 2017;66:2066‐2077. mediated gene editing to treat infections with hepatitis B virus and
77. Mak LY, Wong DK, Seto WK, et al. Hepatitis B core protein as a ther- hepatitis C virus. Virus Res. 2017. https://doi.org/10.1016/j.virus-
apeutic target. Expert Opin Ther Targets. 2017;21:1153‐1159. res.2017.01.003 [Epub ahead of print].
78. Klumpp K, Shimada T, Allweiss L, et al. Efficacy of NVR 3-­778, Alone 94. Dreyer T, Nicholson S, Ely A, et al. Improved antiviral efficacy using
and in combination with pegylated interferon, vs entecavir in uPA/ TALEN-­mediated homology directed recombination to introduce
SCID mice with humanized livers and HBV infection. Gastroenterology. artificial primary miRNAs into DNA of hepatitis B virus. Biochem
2017. https://doi.org/10.1053/j.gastro.2017.10.017 [Epub ahead of Biophys Res Commun. 2016;478:1563‐1568.
print]. 95. Bazinet PV, Placinta G, Moscalu I, et al. Interim follow-up analysis in
79. Berke JM, Dehertogh P, Vergauwen K, et al. Capsid assembly mod- the REP 401 protocol: Functional control of HBeAg negative chronic
ulators have a dual mechanism of action in primary human hepato- HBV infection persists after withdrawal of combined therapy with
cytes infected with hepatitis B virus. Antimicrob Agents Chemother. REP 2139 or REP 2165, tenofovir disoproxil fumarate and pegylated
2017;61:e00560‐17. https://doi.org/10.1128/AAC.00560-00517. interferon alpha-2a. The Liver Meeting, Washinton, DC, USA; 2017.
80. Mani CA, Phelps JR, Abbott C, et al. Antiviral characterization of a Abstract LB-24.
next generation chemical series of HBV capsid inhibitors in vitro and 96. Xu YB, Yang L, Wang GF, et al. Benzimidazole derivative, BM601, a
in vivo. The Liver Meeting Washington, DC, USA; 2017. Abstract novel inhibitor of hepatitis B virus and HBsAg secretion. Antiviral Res.
953. 2014;107:6‐15.
81. Biopharma A. Posting date. https://investor.arbutusbio.com/stat- 97. Zoulim F, Vandenbossche JJ, Lenz O, et al. Safety, tolerability, pharma-
ic-files/dd17ac56-87b0-4aab-af43-2b8496785f2d. [Online.]. cokinetics, and antiviral activity of JNJ-56136379, a novel HBV cap-
Accessed December 20, 2017. sid assembly modulator, in non-cirrhotic, treatment-naïve patients
82. Appourchaux K, Dokmak S, Resche-Rigon M, et al. MicroRNA-­based with chronic hepatitis B. The Liver Meeting AASLD, Washington, DC,
diagnostic tools for advanced fibrosis and cirrhosis in patients with USA; 2017. Abstract LB-15.
chronic hepatitis B and C. Sci Rep. 2016;6:34935. 98. Huang Q, Turner W, Haydar S, et al. Preclinical profile of potent
83. Martinot-Peignoux M, Lapalus M, Asselah T, et al. HBsAg quantifica- second generation CpAMs capable of inhibiting the generation of
tion: useful for monitoring natural history and treatment outcome. HBsAg, HBeAg, pgRNA and cccDNA in HBV infected cells. The Liver
Liver Int. 2014;34(Suppl 1):97‐107. Meeting, Washington, DC, USA; 2017. Abstract 922.
84. Martinot-Peignoux M, Carvalho-Filho R, Lapalus M, et al. Hepatitis 99. Eley T. Single dose safety, tolerability, and pharmacokinetics of AB-
B surface antigen serum level is associated with fibrosis sever- 423 in healthy volunteers from the ongoing single and multiple as-
ity in treatment-­naive, e antigen-­positive patients. J Hepatol. cending dose study AB-423-001. The Liver Meeting, Washinton, DC,
2013;58:1089‐1095. USA; 2017. Abstract 917.
85. Martinot-Peignoux M, Lapalus M, Maylin S, et al. Baseline HBsAg 100. Cai D, Mills C, Yu W, et al. Identification of disubstituted sulfon-
and HBcrAg titres allow peginterferon-­based ‘precision medi- amide compounds as specific inhibitors of hepatitis B virus cova-
cine’ in HBeAg-­negative chronic hepatitis B patients. J Viral Hepat. lently closed circular DNA formation. Antimicrob Agents Chemother.
2016;23:905‐911. 2012;56:4277‐4288.
86. Martinot-Peignoux M, Asselah T, Marcellin P. HBsAg quantification 101. Gane EJ, Gaggar A, Nguyen AH, et al. A phase 1 study evaluating
to optimize treatment monitoring in chronic hepatitis B patients. anti-­PD-­1 treatment with or without GS-­4774 in HBeAg negative
Liver Int. 2015;35(Suppl 1):82‐90. chronic hepatitis B patients. J Hepatol. 2017;66:S26‐S27.
87. Bogomolov P, Alexandrov A, Voronkova N, et al. Treatment of chronic 102. Sprinzl M. Phase 1b clinical trial of TG1050, a novel HBV-targeted
hepatitis D with the entry inhibitor myrcludex B: first results of a immunotherapy, in NUC suppressed chronic hepatitis B patients:
phase Ib/IIa study. J Hepatol. 2016;65:490‐498. safety and early immunological data following single administration.
88. Agarwal K, Cheng W, Sievert W, et al. Bi-weekly dosing of ARB- The Liver Meeting, Washinton, DC, USA; 2017. Abstract 906.
1467 LNP siRNA in HBeAg negative, virally suppressed patients with 103. Locarnini S, Wong D, Jackson K, et al. Novel antiviral activity of
chronic HBV infection leads to deeper declines in HBsAg and po- SB 9200 (Inarigivir), a RIG-I agonost: results from cohort 1 of the
tential association with IL28b. The Liver Meeting, Washington, DC, Achieve trial. The Liver Meeting, Washington, DC, USA; 2017.
USA; 2017. Abstract LB-17. Abstract LB-25.
89. Eley T, Roberts R, Streinu-Cercel A, et al. Pharmacokinetics and ex-
ploratory exposure-response of siRNAs administered monthly as
ARB-001467 (ARB-1467) in a phase 2a study in HBeAg positive and
How to cite this article: Schinazi RF, Ehteshami M, Bassit L,
negative virally suppressed subjects with chronic hepatitis B. The
et al. Towards HBV curative therapies. Liver Int. 2018;
Liver Meeting, Washington, DC, USA; 2017. Abstract 42.
90. Thi EP, Pei L, Ye X, et al. In vivo study of a LNP siRNA investigational 38(Suppl. 1):102–114. https://doi.org/10.1111/liv.13656
agent applied sequentially with immunomodulatory treatments for

You might also like