Clin Mol Hepatol > Volume 31(Suppl); 2025 > Article
Hui, Mak, Fung, Seto, and Yuen: Prospect of emerging treatments for hepatitis B virus functional cure

ABSTRACT

Functional cure, defined as sustained hepatitis B surface antigen (HBsAg) seroclearance with unquantifiable hepatitis B virus (HBV) DNA at 24 weeks off treatment, is a favorable treatment endpoint in chronic hepatitis B (CHB). Nonetheless, functional cure is rarely attained with the current treatment modalities of nucleos(t)ide analogues (NUCs) and pegylated interferon alpha. Multiple novel virus-targeting agents and immunomodulators are under development for HBV with functional cure as the treatment goal. Among virus-targeting agents, antisense oligonucleotides and small-interfering RNAs are the most advanced in the developmental pipeline, and can induce potent and sustainable HBsAg suppression. The other virus-targeting agents have varying effects on HBsAg and HBV DNA, depending on the drug mechanism. In contrast, immunomodulators have modest effects on HBsAg and have limited roles in monotherapy. Multiple combination regimens incorporating RNA interference agents with immunomodulators have been studied through many ongoing clinical trials. These combination strategies demonstrate synergistic effects in inducing functional cure, and will likely be the future direction of development. Despite the promising results, research is warranted to optimize treatment protocols and to establish criteria for NUC withdrawal after novel therapies. Functional cure is now an attainable target in CHB, and the emerging novel therapeutics will revolutionize CHB management.

INTRODUCTION

Chronic hepatitis B (CHB) is a global health threat. Half of the global population lives in regions of high hepatitis B virus (HBV) endemicity, and over 820,000 patients die from HBV-related complications annually [1]. In the context of clinical trial design, functional cure is defined as sustained seroclearance of hepatitis B surface antigen (HBsAg) with unquantifiable HBV DNA at 24 weeks off treatment [2]. Functional cure is a desired endpoint in CHB, as it is associated with reduced liver fibrosis [3,4] and hepatocellular carcinoma (HCC) risks [5].
Currently approved drugs for HBV infection include nucleos(t)ide analogues (NUCs) and pegylated interferon alpha (Peg-IFNα), with NUCs being the predominant treatment option globally. NUCs can effectively suppress HBV replication with high barriers to resistance [6]. However, HBsAg seroclearance rarely occurs with NUC treatment. Yeo et al. [7] performed a meta-analysis on HBsAg seroclearance, demonstrating annual HBsAg seroclearance rates of 0.8% and 1.3% in NUC-treated and untreated patients, respectively. It is estimated that HBsAg reduces by 0.1 log IU/mL per year in NUC-treated patients, equating to HBsAg seroclearance after 36–52 years of treatment [8,9]. Most NUC-treated patients will not achieve functional cure and will require indefinite therapy.
The other approved HBV drug is Peg-IFNα—an immunomodulator that stimulates both innate and adaptive immune responses against HBV. Peg-IFNα is administered subcutaneously, usually for a finite course of 48 weeks [10]. The effects of Peg-IFNα on HBsAg seroclearance are more easily observed in patients with HBV genotype A, hence its role is limited in Asian patients with predominant non-genotype A HBV [10,11]. Furthermore, Peg-IFNα is contraindicated in decompensated liver disease, and is associated with autoimmune, hematological, and psychiatric side effects [12].
Given the limitations of current drugs, novel HBV treatment strategies aiming for functional cure are now under investigation. The first group of novel drugs are virus-targeting agents—which inhibit specific steps of the virus lifecycle, whereas the second group comprises immunomodulators—which augment host immune responses against HBV [12]. This article will review the novel treatment strategies for HBV functional cure, with a focus on agents that are currently under clinical development.

VIRUS-TARGETING AGENTS

HBV naturally exists as an enveloped virus with a partially double-stranded relaxed circular DNA (rcDNA) genome of 3,200 base pairs [13]. The HBV lifecycle is depicted in Figure 1. HBV first enters hepatocytes through interaction with sodium taurocholate cotransporting polypeptide (NTCP) receptors [14]. After cellular entry, the virus uncoats, and the rcDNA is transported into the host nucleus. Through host cell DNA repair machinery, rcDNA is ligated to form covalently closed circular DNA (cccDNA), which can subsequently transcribe messenger RNAs (mRNA) and pregenomic RNA (pgRNA) [15]. mRNAs are translated to viral proteins including hepatitis B surface antigen (HBsAg), hepatitis B core antigen (HBcAg), hepatitis B e antigen (HBeAg), HBV polymerase and the HBV X protein. HBcAg is a key component for the formation of viral capsids, whereas HBsAg is required for the formation of the viral envelope. HBsAg may also be released into the circulation as subviral particles [16].
Novel drugs have been developed to target the key steps of the viral lifecycle, and the main virus-targeting agents in clinical trials are summarized in Table 1.

Entry inhibitors

Entry inhibitors are competitive inhibitors of the HBsAg binding site on NTCP receptors. Through interfering with HBsAg binding, entry inhibitors block HBV entry into hepatocytes. Entry inhibition has primarily been studied in HBV and hepatitis D virus (HDV) coinfected patients, as HDV has limited therapeutic targets otherwise [12].
Bulevirtide is the first-in-class entry inhibitor that was approved by the European Medicines Agency for chronic HDV infection. In the MYR301 trial, patients were randomized to receive bulevirtide 2 mg daily for 144 weeks, bulevirtide 10 mg daily for 144 weeks, or to receive no treatment for 48 weeks followed by bulevirtide 10 mg daily for 96 weeks (control group). The primary endpoint was normalization of alanine aminotransferase (ALT) plus undetectable or 2 log reduction in HDV RNA at week 48. 45% and 48% of patients in the bulevirtide 2 mg and bulevirtide 10 mg groups achieved the primary endpoint, respectively, whereas only 2% of control patients reached this endpoint. Despite its potent effects on HDV, HBsAg seroclearance or HBsAg reduction by ≥1 log IU/mL did not occur in any patients after 48 weeks of bulevirtide treatment [17]. Given the availability of other novel agents, bulevirtide would likely have a limited role for patients with HBV mono-infection. Nonetheless, bulevirtide would remain as an important drug for patients with HBV and HDV coinfection.

Transcription inhibitors

Farnesoid X receptor (FXR) are nuclear receptors that regulate bile acid homeostasis [18]. FXR also interacts with the HBV enhancer II core promoter sequences and with the HBV X protein to regulate HBV transcription. FXR agonists can inhibit the transcription of cccDNA, inhibiting downstream translation and viral replication [19].
Vonafexor is a selective FXR agonist that has been studied in CHB patients. After administration of vonafexor 400 mg daily for 29 days, a statistically significant yet modest reduction of HBsAg by 0.1 log IU/mL was achieved. Vonafexor had no significant effects on other HBV markers after 29 days of treatment [20].

RNA interference agents

RNA interference (RNAi) agents can be subdivided into antisense oligonucleotides (ASOs) and small-interfering RNAs (siRNAs). Both types of drugs are designed with complementary nucleotide sequences to conserved regions of the HBV genome, enabling potent post-transcriptional gene silencing of all HBV transcripts [21]. As HBsAg can be produced from both HBV cccDNA and integrated HBV DNA [22], RNAi agents are designed to target sequences upstream from integration hotspots. This in turn enables knockdown of RNA transcribed from both cccDNA and integrated HBV DNA [21]. Aside from their direct effect in suppressing viral protein production and viral replication, RNAi agents also indirectly enhance host immune reconstitution through reducing host exposure to viral proteins.
Despite their similar action, ASOs and siRNAs have distinct structures. ASOs are single-stranded DNA that can enter hepatocytes in the unconjugated form. ASOs accumulate in cytoplasm, and operate through RNA splicing by RNAse-H. siRNAs are double-stranded RNA which enter hepatocytes through conjugated carriers. siRNAs accumulate in endosomes, and operate through RNA splicing by the RNA-induced silencing complex [21].
Bepirovirsen is an unconjugated ASO that is the most advanced novel agent in the developmental pipeline now. In the phase IIb B-Clear trial on bepirovirsen, the primary outcome was undetectable HBsAg with unquantifiable HBV DNA at 24 weeks post end-of-treatment (EOT). In patients who received subcutaneously injected bepirovirsen 300 mg weekly for 24 weeks with a loading dose, 9% of NUCtreated patients and 10% of non-NUC patients achieved the primary outcome, respectively. Among the patients who received 24 weeks of 300 mg weekly bepirovirsen, baseline HBsAg ≤3 log IU/mL was associated with higher response rates to bepirovirsen (NUC-treated patients: primary outcome in 16% of patients with baseline HBsAg ≤3 log IU/mL vs. 6% of patients with baseline HBsAg >3 log IU/mL; non-NUC patients: primary outcome in 25% of patients with baseline HBsAg ≤3 log IU/mL vs. 7% of patients with baseline HBsAg >3 log IU/mL) [23]. It is noteworthy that the intrinsic Toll-like receptor (TLR) 8 activity of bepirovirsen may provide additional control on HBV. Long-term monitoring of bepirovirsen responders is underway, with interim results suggesting sustainable drug effects for more than 9 months [24]. Bepirovirsen has now entered phase III trials.
GSK3389404 is an alternate form of bepirovirsen that is conjugated with N-acetylgalactosamine (GalNAc). GSK3389404 was studied in a phase II trial, yielding HBsAg suppression by 0.13–0.75 log IU/mL after 85 days of treatment. No incidences of HBsAg seroclearance were observable [25]. GSK3389404 has weaker HBsAg suppression effects than bepirovirsen, and its development has hence been halted. It is hypothesized that GalNAc conjugation enhanced hepatocyte uptake and limited GSK3389404 uptake in liver sinusoidal cells and Kupffer cells, which in turn reduced immune signaling and overall anti-HBV effects [26].
A range of siRNAs including daplusiran/tomligisiran, elebsiran, imdusiran, RBD-1016 and xalnesiran have entered phase II trials. In general, the siRNAs have 1 or more triggers targeting conserved sequences in the HBV genome. Daplusiran/ tomligisiran has 2 triggers targeting the S and X open reading frames (ORFs), xalnesiran has 1 trigger targeting the S ORF, while elebsiran, imdusiran and RBD-1016 are all designed with 1 trigger targeting the X ORF. As a class, siRNAs can potently suppress HBsAg, with over 90% of patients achieving HBsAg reduction by ≥1 log IU/mL and over 50% of patients achieving HBsAg suppression to <100 IU/mL [27-31]. Importantly, siRNA effects are sustainable after EOT. Mak et al. [32] followed up siRNA treated patients (15 patients with 4 doses of ARC-520, and 38 patients with 3 doses of daplusiran/ tomligisiran) for a mean time of 52.5 months, demonstrating sustainable HBsAg suppression for up to 6 years in some patients.

Capsid assembly modulators

The HBV nucleocapsid is essential for the HBV lifecycle, as it encapsidates HBV pgRNA and HBV polymerase, acting as a site for reverse transcription. Nucleocapsids containing mature nucleic acids can be enveloped by HBsAg to form new virions or may be recycled to replenish the cccDNA pool inside the hepatocyte nucleus [33].
Capsid assembly modulators (CAMs) are oral drugs that target HBcAg to interfere with nucleocapsids. CAMs are further divided into CAM-As—which induce aberrant nucleocapsid formation, and CAM-Es—which induce empty nucleocapsid formation [34]. Through its direct action on nucleocapsids, CAMs enable potent suppression of viral replication. As a class, CAMs can suppress both HBV DNA and HBV RNA to different degrees depending on individual drug potency. However, the early generation CAMs had minimal effects on viral proteins such as HBsAg [35-37].
Aside from its direct effect on nucleocapsids, CAMs also have secondary effects in inhibiting nucleocapsid recycling, in turn blocking cccDNA replenishment. Newer generation CAMs such as ALG-000184 have stronger secondary effects and are being actively studied [33]. In the ongoing ALG-000184-201 phase I trial, ALG-000184 (+/- entecavir) is administered for 96 weeks in HBeAg positive patients. In the week 72 interim analysis, a maximum mean HBV DNA reduction of 7 log IU/mL was achieved. 90% of patients on ALG-000184 300 mg monotherapy and 50% of patients on ALG-000184 300 mg+entecavir achieved unquantifiable HBV DNA at week 72. More importantly, significant reductions in HBV antigens were observed at week 72, with maximum mean reduction of HBsAg, HBeAg and HBrAg by 1.0 log IU/mL, 1.9 PEI log U/mL, and 2.1 log U/mL, respectively [38]. Based on the positive phase I data, ALG-000184 will be further studied in phase II trials.

Protein export inhibitors

Nucleic acid polymers (NAPs) are phosphorothioate oligonucleotides that inhibit HBV subviral particle transport from hepatocytes into the circulation. REP2139 (and its analogue REP2165) are NAPs administered by weekly intravenous infusions. In a phase II trial, REP2139/ REP2165 for 48 weeks led to HBsAg suppression by more than 1 log IU/mL in 87.5% of patients (35 out of 40), with 60% of patients (24 out of 40) having HBsAg suppressed to <0.05 IU/mL. At 48 weeks after EOT, 35% of patients (14 out of 40) had sustained response with HBsAg <0.05 IU/mL [39]. S antigen Transport-inhibiting Oligonucleotide Polymers (STOPS) are structurally altered forms of NAPs [40]. However, the development of STOPS has been discontinued after a negative phase I trial.

IMMUNOMODULATORS

Host immune tolerance is a critical pathogenetic factor contributing to HBV persistence. With long-term exposure to viral proteins (such as HBsAg), the host immune system develops tolerance towards HBV. HBV has evolved evasion mechanisms against the innate immune system [41], and can also induce T-cell dysfunction [42]. In contrast to virus-targeting agents, immunomodulators aim to reverse immune tolerance to generate host anti-HBV responses. The currently approved Peg-IFNα is an immunomodulator targeting the innate immune system, which can induce downstream cytokine release to trigger lymphocyte responses. Figure 2 depicts the main modes of action of novel HBV immunomodulators, and the immunomodulators in clinical trials are summarized in Table 2.

Toll-like receptor agonists

TLRs are important pattern recognition receptors in the innate immune system. TLRs detect pathogen molecular patterns and trigger downstream interferon and cytokine pathways to initiate host antiviral responses [43].
Selgantolimod is an oral TLR8 agonist that has entered phase II clinical trials. In 39 NUC-treated CHB patients who received selgantolimod for 24 weeks, 18% and 26% of patients had >0.1 log IU/mL HBsAg decline at EOT and at 24 weeks after EOT, respectively. 5% of patients (2 out of 39) had HBsAg seroclearance at 24 weeks after EOT [44]. Another phase II trial studied the use of selgantolimod+tenofovir alafenamide (TAF) in viremic CHB patients. Selgantolimod induced significant changes in serum cytokines, chemokines and immune cell subsets, yet no significant HBsAg suppression was noted. None of the viremic patients treated with selgantolimod achieved HBsAg decline by 1 log IU/mL or more at EOT. The mean HBsAg reduction at week 48 was 0.12, 0.16 and 0.12 log IU/mL in the selgantolimod 3 mg+TAF arm, the selgantolimod 1.5 mg+TAF arm, and the placebo+TAF arm, respectively [45].
TLR7 agonists have also been studied in CHB. While TLR7 agonists have shown target engagement with downstream immune signaling, the HBsAg reduction by TLR7 agonist monotherapy is modest. Ruzotolimod (RO7020531) treatment for 6 weeks led to mean HBsAg decline ranging from 0.07–0.15 log IU/mL at 6 weeks post-EOT [46]. Vesatolimod, another TLR7 agonist, showed no significant effects on HBsAg after 12 weeks of treatment [47,48].

T-cell modulation

T-cell dysfunction and deficit are important contributing factors to adaptive immunity dysfunction in CHB [42]. The programmed cell death protein 1 (PD1)/programmed cell death ligand 1 (PDL1) pathway is inhibitory towards T-cells and is overexpressed in CHB. Anti-PD1 or anti-PDL1 agents have hence drawn interests as novel immunomodulators for CHB.
Envafolimab is the only subcutaneously administered anti-PDL1 checkpoint inhibitor, while others are administered intravenously. In a phase II trial, lower baseline HBsAg level was significantly associated with higher magnitude of HBsAg reduction. After 24 weeks of envafolimab, the EOT HBsAg reduction was 1.38, 0.19, and 0.09 log IU/mL in patients with baseline HBsAg ≤100, 100–1,000, and >1,000 IU/mL, respectively [49]. In an extension trial studying 19 patients with baseline HBsAg <100 IU/mL, 21.1% of patients (4 out of 19) achieved HBsAg seroclearance after 24 weeks of envafolimab [50]. Cemiplimab and nivolumab are other anti-PD1/anti-PDL1 checkpoint inhibitors that have entered phase II trials.
Inhibitors of apoptosis (IAP) antagonist and immune mobilizing monoclonal T-cell receptors against virus (ImmTAV) are other novel T-cell modulating techniques that have shown anti-HBV effects in preclinical studies [51,52]. Both IAP antagonists and ImmTAV are being studied in ongoing clinical trials.

Therapeutic vaccination

Therapeutic vaccination utilizes host active immunity to generate anti-HBV responses. HBsAg-based therapeutic vaccines have entered development for almost two decades, yet older HBsAg-based therapeutic vaccines had limited antiviral effects [53]. Emerging data showed that the incorporation of non-HBsAg viral proteins may enhance vaccinal effects, and this has since reignited interest in HBV therapeutic vaccines [54].
VTP-300 is a prime-boost therapeutic vaccine targeting the polymerase, core and S regions of HBV. In a phase II trial, VTP-300 led to HBsAg decline by >0.5 log IU/mL in 16.7% of patients (3 out of 18 patients), with sustained HBsAg suppression at 8 months post-EOT. Of note, the three patients with HBsAg decline >0.5 log IU/mL all had baseline HBsAg levels <50 IU/mL [55].
BRII-179 is a therapeutic vaccine that targets all three HBV surface proteins (HBsAg, pre-S1 and pre-S2). In a phase II trial on BRII-179, >30% of patients developed anti-HBs responses with BRII-179 treatment, yet no significant HBsAg reductions were observed [56].

Monoclonal antibodies

Passive immunity through administration of anti-HBV monoclonal antibodies has been studied as a novel immunomodulation strategy for CHB. In the VIR-3434-1002 trial, a single dose of the monoclonal antibody tobevibart was administered. In an interim analysis up to week 8, patients who received tobevibart 300 mg achieved mean HBsAg and HBV DNA decline from baseline to nadir by 1.83 log IU/mL and 2.03 log IU/mL, respectively [57].
Lenvervimab is another monoclonal antibody that has entered clinical trials. Lenvervimab was administered at weekly doses for 4 doses in a phase I trial on patients with baseline HBsAg <1,000 IU/mL. 10.3% of patients (3 out of 29) who received multiple lenvervimab doses achieved undetectable HBsAg during treatment, although the HBsAg levels subsequently rebounded after EOT [58].

COMINBATION STRATEGIES

As reviewed in the prior sections, the novel HBV therapeutics are unable to consistently induce HBsAg seroclearance at high rates. Some virus-targeting agents, such as RNAi agents, have demonstrated potent effects in HBsAg suppression. Nonetheless, baseline HBsAg level appears to heavily influence treatment responses [23]. In contrast, novel immunomodulators all have modest effects on HBsAg, and have limited roles in monotherapy. It is hypothesized that combination strategies with novel HBV therapeutics may yield synergistic antiviral effects by targeting multiple pathways simultaneously [59]. Table 3 depicts the novel combination strategies that have entered clinical trials.

Combination of virus-targeting agents

The combination of daplusiran/tomligisiran (siRNA)+ bersacapavir (CAM) has been studied in the REEF-1 and REEF-2 trials. In the REEF-1 trial, the primary outcome was the proportion of patients reaching a predetermined criteria for stopping NUCs (ALT below 3x upper limit normal, unquantifiable HBV DNA, negative HBeAg, and HBsAg <10 IU/mL). After 48 weeks of treatment, the primary outcome occurred in 19% of patients on daplusiran/tomligisiran 200 mg monotherapy, 16% of patients on daplusiran/ tomligisiran 100 mg monotherapy, 9% of patients on daplusiran/ tomligisiran 100 mg+bersacapavir 250 mg, and 0% of patients on bersacapavir 250 mg monotherapy. Patients receiving daplusiran/tomligisiran 100 mg+bersacapavir 250 mg achieved HBsAg decline by 1.8 log IU/mL at EOT. In comparison, the treatment arms on daplusiran/tomligisiran 200 mg and daplusiran/tomligisiran 100 mg achieved EOT HBsAg decline by 2.6 log IU/mL and 2.1 log IU/mL, respectively. No patients in the combination arm achieved HBsAg seroclearance, whereas two patients on daplusiran/ tomligisiran 200 mg and one patient on daplusiran/ tomligisiran 100 mg achieved HBsAg seroclearance at 24 weeks post-EOT [60].
In the REEF-2 trial, the combination of daplusiran/tomligisiran 200 mg+bersacapavir 250 mg was compared against placebo in NUC-treated patients. 46.9% of patients on combination therapy achieved HBsAg <100 IU/mL at 48 weeks post-EOT, yet no patients achieved HBsAg seroclearance. All treatment including NUCs were withdrawn after 48 weeks of therapy. Patients in the treatment arm, when compared with controls, were less likely to have post-EOT biochemical flares (3.6% in active arm vs. 28.6% in control arm), and were less likely to require NUC resumption (9.1% in active arm vs. 26.8% in control arm) [61].
Imdusiran (siRNA)+vebicorvir (CAM) is another virus-targeting agent combination that has been studied. A phase II trial compared NUC-treated patients on imdusiran+ vebicorvir, imdusiran monotherapy, and vebicorvir monotherapy, respectively. An outcome of interest was the proportion of patients reaching a predetermined criteria to stop NUCs at week 48 (ALT below 2x upper limit normal, unquantifiable HBV DNA, and HBsAg <100 IU/mL). This stop NUC criteria was achieved by 80.0% of patients on imdusiran monotherapy, 61.5% of patients on combination therapy, and 0% of patients on vebicorvir monotherapy [62].
These studies suggest that the combination of siRNA+ CAM does not have synergistic effects on HBsAg suppression. The HBsAg reduction in this combination regimen appears to be primarily driven by siRNAs.

Combination of immunomodulators

The combination of therapeutic vaccination with anti-PD1 checkpoint inhibitors can simultaneously augment the function and abundance of anti-HBV T-cells. This strategy was studied in a phase II trial on VTP-300 (therapeutic vaccine)+nivolumab (anti-PD1). VTP-300 is a prime-boost vaccine with the prime and boost doses given at baseline and day 28, respectively. The trial was hence designed with treatment arms of 1) nivolumab administered with the day 28 boost dose only, or 2) nivolumab administered at baseline with the prime dose and at day 28 with the boost dose. In patients who received VTP-300+day 28 nivolumab, mean HBsAg decline of0.76 log IU/mL and 0.80 log IU/mL were achieved at 2 months post-EOT and 7 months post-EOT, respectively. 11.1% of patients (2 out of 18 patients) in the VTP-300+day 28 nivolumab arm achieved HBsAg seroclearance, and this response was sustained at 7 months post-EOT. In contrast, patients who received VTP-300+nivolumab at both baseline and day 28 did not achieve significant HBsAg decline or HBsAg seroclearance [55]. A potential explanation is that nivolumab with the prime dose vaccine may interfere with initial T-cell activation, which in turn negates vaccinal effects.
An ongoing open label trial is studying different regimens of VTP-300+nivolumab in patients with baseline HBsAg <200 IU/mL. The trial includes arm 1—vaccine boost with nivolumab on day 29, arm 2—vaccine boost with nivolumab on day 29 and day 85, and arm 3—vaccine boost on day 29 and day 85 with nivolumab on day 36. 43% of patients in arm 2 (3 out of 7) and 14% in arm 3 (1 out of 7) achieved undetectable HBsAg, and two of these patients from arm 2 had sustained undetectable HBsAg at day 169 [63].
Another immunotherapy combination that has been studied is BRII-179 (therapeutic vaccine) with or without IFNα. This regimen was able to mount antibody responses to HBsAg, although there was no significant HBsAg reduction observed [56].

Combination of virus-targeting agents and immunomodulators

The combination of virus-targeting agents with immunomodulators targets both the host and the virus, which should yield maximal synergistic effects. Given the promising results seen in monotherapy with RNAi agents, they have become the backbone in most combination regimens with immunomodulators. The RNAi agents can also induce immune reconstitution through reducing viral antigens, which should theoretically augment immunomodulator effects.
The phase II B-Together trial studied the combination of bepirovirsen with Peg-IFNα in NUC-treated patients. The trial enrolled 55 patients in treatment arm 1—bepirovirsen 300 mg weekly with loading dose for 24 weeks followed by 24 weeks of Peg-IFNα, and 53 patients in treatment arm 2—bepirovirsen 300 mg weekly with loading dose for 12 weeks followed by 24 weeks of Peg-IFNα. Overall, 9% of patients in treatment arm 1 and 15% of patients in treatment arm 2 achieved HBsAg seroclearance at 24 weeks post-EOT. While Peg-IFNα did not enhance the magnitude of HBsAg suppression on top of bepirovirsen, it was effective in reducing post-EOT relapse [64].
Peg-IFNα has also been studied in combination with multiple siRNAs including imdusiran, elebsiran, and daplusiran/tomligisiran, respectively. Patients generally had excellent HBsAg suppression with siRNA+Peg-IFNα. In the combination of imdusiran+Peg-IFNα, 28% of patients who received 24 weeks of Peg-IFNα achieved HBsAg seroclearance with anti-HBs development at EOT [65]. Whereas with the elebsiran (6–13 doses)+Peg-IFNα (44–48 weeks) regimen, 25.8% of patients achieved HBsAg seroclearance at EOT, with 16.1% of patients having sustained HBsAg seroclearance at 24 weeks post-EOT [66]. Daplusiran/tomligisiran+Peg-IFNα has been studied in immunotolerant patients with baseline HBV DNA >4 log IU/mL, and the combination treatment led to sustained HBsAg seroclearance in 14.8% of patients [67].
Another commonly studied combination strategy involves siRNA+therapeutic vaccines. In the trial on imdusiran+ VTP-300, 94.7% on combination therapy achieved HBsAg <100 IU/mL at EOT, while 84.2% in the imdusiran monotherapy arm reached this endpoint [68]. Daplusiran/tomligisiran+JNJ-0535 (therapeutic vaccine) and elebsiran+BRII-179 (therapeutic vaccine) are other combinations that have entered phase II trials, demonstrating mean EOT HBsAg reduction of 2.18 log IU/mL and 1.75 log IU/mL, respectively [69,70].
The phase II PIRANGA trial studied the combination of xalnesiran+ruzotolimod (TLR7 agonist) or Peg-IFNα in NUC-treated patients. Among patients with baseline HBsAg <3 log IU/mL, patients on xalnesiran+Peg-IFNα appeared to have the best response, with 23% achieving HBsAg seroclearance at 24 weeks post-EOT. In contrast, this endpoint was achieved by 12% of patients on xalnesiran+ ruzotolimod, and in 3% of patients on xalnesiran monotherapy [71].
In the phase II trial on daplusiran/tomligisiran+nivolumab (anti-PD1), EOT HBsAg reduction of 2.0–2.1 log IU/mL was achieved, with 35.3–50.0% of patients achieved HBsAg <10 IU/mL at EOT, depending on different nivolumab administration regimens [72].
Finally, the phase II MARCH trial studied the combination of elebsiran+tobevibart (anti-HBV monoclonal antibody) +/- Peg-IFNα. The combination of elebsiran+tobevibart+PegIFNα appeared to enhance the sustainability of HBsAg suppression, as 14.3% of patients on this triple regimen had HBsAg seroclearance at EOT with 9.5% sustaining HBsAg negativity at 12 weeks post-EOT. In contrast, 15.0% on elebsiran+tobevibart had HBsAg seroclearance at EOT, with no patients sustaining this response at 12 weeks post-EOT [73].

SAFETY OF NOVEL TREATMENT STRATEGIES

As the novel agents in development have all been meticulously evaluated in animal studies and phase I trials, they are generally well-tolerated with favorable safety profiles. Nonetheless, several agents have indeed shown unexpected adverse effects, requiring termination of development. Terminated agents include inarigivir (pattern recognition receptor agonist) – with drug-induced mortality in patients, ARC-520 (siRNA)—with mortality in non-human primates due to the drug excipient, and ABI-H2158 (CAM)—with severe drug-induced hepatotoxicity [12]. It is hence critical to collect long-term safety data from novel agents before they are routinely implemented in clinical practice.
As reviewed above, Peg-IFNα is commonly utilized as an adjunct in combination regimens. Peg-IFNα is contraindicated in patients with decompensated cirrhosis. Furthermore, Peg-IFNα is associated with a diverse range of adverse effects. Clinicians should be vigilant with Peg-IFNα use, and some patients will inevitably require regimens without Peg-IFNα.
Ongoing trials frequently include NUC discontinuation phases to assess for sustained HBsAg seroclearance after NUC withdrawal. This is necessary for achieving functional cure, yet the evidence on NUC discontinuation after novel therapies is limited. Furthermore, NUC discontinuation is not without risks. In the REEF-2 trial (daplusiran/tomligisiran+bersacapavir), a patient enrolled in the control arm (receiving NUC alone) developed severe HBV flare with liver failure after NUC withdrawal [74]. Future research should aim to study viral marker kinetics after NUC discontinuation, and universal criteria for safe NUC withdrawal after novel therapies should be established [75].

DISCUSSION

The field of novel HBV therapeutics is rapidly growing. Due to varying patient populations and trial designs, it is difficult to directly compare the efficacy of different novel therapeutics. Nonetheless, RNAi agents appear to be the most potent in HBsAg suppression and in inducing HBsAg seroclearance. Among patients who received 300 mg weekly bepirovirsen for 24 weeks, the best performance was achieved in patients with HBsAg ≤3 log IU/mL (16% of NUC-treated patients and 25% of non-NUC patients achieving sustained undetectable HBsAg at 24 weeks post-EOT) [23]. Whereas for combination regimens of ASOs or siRNAs+Peg-IFNα, sustained HBsAg seroclearance at 24 weeks post-EOT was achievable in ≥15% of patients in specific treatment arms [64,66,67,71]. Long-term data has also been accumulated in siRNAs, demonstrating sustainable HBsAg responses for up to 6 years [32]. The maintenance of treatment responses after EOT is critical for novel therapeutics, as sustained HBsAg seroclearance is required for the definition of functional cure. Viral rebound after EOT is suboptimal as it represents a state of inadequate viral control, and treatment withdrawal may not be safe.
Aside from RNAi agents, newer-generation CAMs can also effectively suppress HBsAg. The efficacy in HBsAg suppression likely corresponds to the targeted pathways by the virus-targeting agents. RNAi agents target cccDNA transcriptional activity, whereas newer-generation CAMs prevent cccDNA replenishment, highlighting the importance of targeting cccDNA-related pathways in HBsAg suppression.
Among immunomodulators, incidences of HBsAg seroclearance have been reported in trials for specific agents, including selgantolimod, envafolimab, and lenvervimab. Nonetheless, immunomodulators generally have modest effects on HBsAg, and are unlikely to be used as monotherapy.
Various combination strategies including combination of virus-targeting agents, combination of immunomodulators, and combination of virus-targeting agents with immunomodulators have been tested. The combinations involving RNAi agents+immunomodulators appear to have the most promising results, as immunomodulators ride on the RNAi-induced HBsAg reduction and increase durability. It is important to study the influence of dosing sequence and timing in combination regimens, as these factors can induce unforeseen negative effects on treatment responses (such as in the combination of VTP-300+nivolumab) [55]. Safety, costs, and patient acceptability are also important considerations before these novel combination strategies can be widely utilized.
Current clinical trials on novel therapeutics focused on highly selected HBV patient cohorts, and our understanding on treatment response predictors remains limited. Lower baseline HBsAg is associated with HBsAg seroclearance with different novel agents [23,55,63], and will likely be a key factor in patient selection. Age is also negatively correlated with HBsAg reduction in siRNA therapy [32]. Ongoing studies are assessing immune-based markers in both peripheral blood and in the liver as predictors of treatment response [76,77]. Ultimately, a better understanding of response predictors will guide personalized application of novel therapeutics. Patients with low likelihood of HBsAg seroclearance may require combination regimens with specific tailoring of treatment duration and dosing. In contrast, patients with high likelihood of HBsAg seroclearance may only require a single course of RNAi therapy to achieve functional cure.
With progressive advancements in novel HBV therapeutics, functional cure is now an attainable treatment goal. However, functional cure should not be the final target in HBV therapy. The persistence of integrated HBV DNA and cccDNA after functional cure remains as risk factors for liver related complications [2]. In particular, potentially hepatocarcinogenic HBV integrations are present in all HBV disease phases, even after HBsAg seroclearance [78]. Complete HBV cure—the sterilization of cccDNA and integration HBV DNA from the host, should hence be the ultimate treatment target. Complete cure should theoretically eliminate risks of HBV-related complications. Preclinical studies have utilized gene editing and epigenetic techniques in targeting integrated HBV DNA and cccDNA to achieve complete cure [79-81]. The results from these novel treatment approaches are keenly anticipated, and the development of these novel techniques may enable complete HBV cure in the future.
While the current clinical trials have shown exciting results on virological responses and HBsAg seroclearance, it remains uncertain whether novel agents can reduce liver-related complications. Future studies must collect long-term data to determine whether virological responses to novel therapies can be translated to clinically relevant outcomes.
From a population health perspective, development of effective novel therapeutics would dramatically boost our progress in hepatitis elimination. However, practical issues such as linkage to care and drug provision are equally important in hepatitis care. It is estimated that only 10.3% of CHB patients worldwide know of their infection status [82], while only 8.2% of treatment-eligible patients actually receive therapy [83]. Public health interventions to optimize HBV care pathways will be necessary to maximize the beneficial effects from novel therapeutics.
To conclude, multiple novel virus-targeting agents and immunomodulators are under active development. Combination strategies have also emerged, demonstrating potent and sustainable effects in inducing HBsAg seroclearance. Functional cure is now an attainable treatment goal in CHB. Multiple clinical trials are ongoing, and the emerging novel agents will revolutionize CHB management.

FOOTNOTES

Authors’ contribution
RWHH was involved in data interpretation and drafting of the manuscript. LYM, JF and WKS were involved in critical revision of the manuscript. MFY was involved in study concept, critical revision of the manuscript, and overall study supervision. All authors have seen and approved the final version of the manuscript.
Acknowledgements
Generate AI and AI-assisted technologies were not used in preparation of this manuscript.
Conflicts of Interest
MF Yuen is an advisory board member and/or received research funding from AbbVie, Arbutus Biopharma, Assembly Biosciences, Bristol Myer Squibb, Dicerna Pharmaceuticals, GlaxoSmithKline, Gilead Sciences, Janssen, Merck Sharp and Dohme, Clear B Therapeutics, Springbank Pharmaceuticals; and received research funding from Arrowhead Pharmaceuticals, Fujirebio Incorporation and Sysmex Corporation. WK Seto received speaker’s fees from AstraZeneca and Echosens, is an advisory board member and received speaker’s fees of Abbott, received research funding from Alexion Pharmaceuticals, Boehringer Ingelheim, Pfizer and Ribo Life Science, and is an advisory board member, received speaker’s fees and researching funding from Gilead Sciences. The remaining authors have no conflict of interests.

Figure 1.
Novel virus-targeting agents for hepatitis B. cccDNA, covalently-closed circular DNA; HBcAg, hepatitis B core antigen; HBsAg, hepatitis B surface antigen; HBV, hepatitis B virus; mRNA, messenger RNA; NTCP, sodium taurocholate cotransporting polypeptide; pgRNA, pregenomic RNA; rcDNA, relaxed circular DNA.

cmh-2024-0855f1.jpg
Figure 2.
Novel immunomodulators for hepatitis B. HBV, hepatitis B virus.

cmh-2024-0855f2.jpg
Table 1.
Virus-targeting agents in clinical development
Class Mechanism Agent Delivery Dosing* Clinical trial number Clinical trial phase
Entry inhibitor NTCP receptor competitive antagonist Bulevirtide SC 2 mg or 10 mg daily for 144 weeks NCT03852719 Phase III
Transcription inhibitor FXR agonist Vonafexor Oral 400 mg daily for 29 days NCT04365933 Phase II
RNA interference agents ASO Bepirovirsen SC 300 mg or 150 mg weekly with loading dose for up to 24 weeks NCT05630820 Phase III
siRNA Daplusiran/tomligisiran SC Ascending dosing (25–400 mg) with varying dosing intervals (weekly, 2-weekly, 4-weekly) for 3 doses NCT03365947 Phase II
Elebsiran SC 200 mg monthly for 6 months NCT02826018 and NCT03672188 Phase II
Imdusiran SC 60 mg or 90 mg at varying dosing intervals (4-weekly, 8-weekly, 12-weekly) for 48 weeks NCT06154278 Phase II
RBD-1016 SC Ascending dosing (0.3–3.0 mg/kg) at days 1 and 29 NCT05961098 Phase II
Xalnesiran SC Ascending dosing (1.5–6.0 mg/kg) monthly for 4 months NCT03772249 Phase II
CAMs CAM-A GLS4 Oral 120 mg three times daily for 96 weeks NCT04147208 Phase II
RO7049389 Oral 600 mg daily for 48 weeks NCT04225715 Phase II
CAM-E ALG-000184 Oral 300 mg daily for 96 weeks NCT04536337 Phase I
Bersacapavir Oral 250 mg daily for 48 weeks NCT04439539 Phase II
Protein export inhibitor NAPs REP2139 IV 250 mg weekly for 48 weeks NCT0256719 Phase II
REP2165 IV 250 mg weekly for 48 weeks NCT0256719 Phase II

ASO, antisense oligonucleotide; CAM, capsid assembly modulator; FXR, farnesoid X receptor; IV, intravenous; NAP, nucleic acid polymer; NTCP, sodium-taurocholate co-transporting polypeptide; SC, subcutaneous; siRNA, Small-interfering RNA.

* Dosing in latest clinical trials at the time of writing.

Arm 1: Bepirovirsen 300 mg weekly for 24 weeks with loading dose; arm 2: bepirovirsen 300 mg weekly with loading dose for 12 weeks, followed by 150 mg weekly for 12 weeks; arm 3: bepirovirsen 300 mg weekly with loading dose for 12 weeks, followed by placebo for 12 weeks, arm 4: placebo for 12 weeks, followed by bepirovirsen 300 mg weekly without loading dose for 12 weeks.

Table 2.
Immunomodulators in clinical development
Class Mechanism Agent Delivery Dosing* Clinical trial number Clinical trial phase
Toll-like receptor agonist Toll-like receptor 8 agonist Selgantolimod Oral 1.5 mg or 3.0 mg weekly for 24 weeks NCT03491553 and NCT03615066 Phase II
Toll-like receptor 7 agonist Ruzotolimod Oral 150 mg or 170 mg alternate day for 6 weeks NCT04225715 Phase II
Vesatolimod Oral Ascending dosing (1–4 mg) weekly for 4–12 weeks NCT02166047 Phase II
T-cell modulation Anti-PD1 or anti-PDL1 Envafolimab SC Ascending dosing (0.3–2.5 mg/kg) every 2 weeks for up to 24 weeks NCT04465890 Phase II
Cemiplimab IV Ascending dosing (0.3–3.0 mg/kg) for 2 doses in 6 weeks NCT04046107 Phase II
Nivolumab IV 0.3 mg/kg monthly for up to 24 weeks NCT04891770 Phase II
Inhibitors of apoptosis antagonist APG1387 IV Ascending dosing (12–30 mg) weekly for 12 weeks NCT04568265 Phase II
Immune mobilizing monoclonal T-cell receptors against virus IMC-I109V IV Ascending dosing weekly for up to 24 weeks NCT05867056 Phase I
Therapeutic vaccination Targeting polymerase, HBcAg and HBsAg VTP-300 IM Active component (ChAdOx1-HBV 2.5×1010 vp) with Ankara boost (MVA-HBV 1×108 pfu) once NCT04778904 Phase II
Targeting HBsAg, Pre-S1 and Pre-S2 BRII-179 IM 20 ug or 40 ug monthly for 12 weeks NCT04749368 Phase II
Targeting HBcAg and HBsAg GSK3528869A SC Active component (ChAd155-hli-HBV 5×1010 vp) with Ankara boost (MVA-HBV 2×108 pfu) once NCT03866187 & NCT05276297 Phase II
Monoclonal antibodies IgG1-type recombinant human hepatitis B immunoglobulin Lenvervimab IV Ascending dosing (80,000–24,000 IU) weekly for 4 weeks NCT03801798 Phase II
IgG1-lambda anti HBsAg human monoclonal antibody Tobevibart SC Single scending dose (6–300 mg) NCT05612581 & NCT04856085 Phase II

HBcAg, hepatitis B core antigen; HBsAg, hepatitis B surface antigen; IgG, immunoglobulin G; IM, intramuscular; IV, intravenous; PD1, programmed cell-death 1; PDL1, programmed cell-death ligand 1; SC, subcutaneous.

* Dosing in latest clinical trials at the time of writing.

Table 3.
Combination regimens in clinical development
Mode of combination Combination regimen Clinical trial number Clinical trial phase
Combination of virus-targeting agents Daplusiran/tomligisiran (siRNA)+bersacapavir (CAM) NCT03982186 & NCT04129554 Phase II
Imdusiran (siRNA)+vebicorvir (CAM) NCT04820686 Phase II
Combination of immunomodulators VTP-300 (Therapeutic vaccine)+nivolumab (Anti-PD1) NCT04778904 Phase II
BRII-179 (Therapeutic vaccine)+IFNα NCT04749368 Phase II
Combination of virus-targeting agents with immunomodulators Bepirovirsen (ASO)+Peg-IFNα NCT04676724 Phase II
Imdusiran (siRNA)+Peg-IFNα NCT04980482 Phase II
Elebsiran (siRNA)+Peg-IFNα NCT04412863 Phase II
Daplusiran/tomligisiran (siRNA)+bersacapavir (CAM)+Peg-IFNα NCT04439539 Phase II
Imdusiran (siRNA)+VTP-300 (Therapeutic vaccine) ACTRN12622000317796 Phase II
Elebsiran (siRNA)+BRII-179 (Therapeutic vaccine) NCT04749368 Phase II
Daplusiran/tomligisiran (siRNA)+JNJ-0535 (Therapeutic vaccine) NCT05123599 Phase I
Xalnesiran (siRNA)+ruzotolimod (Toll-like receptor 7 agonist) or Peg-IFNα NCT04225715 Phase II
Daplusiran/tomligisiran (siRNA)+nivolumab (anti-PD1) ISRCTN15803686 Phase II
Elebsiran (siRNA)+tobevibart (Monoclonal antibody) NCT04856085 Phase II

ASO, antisense oligonucleotide; CAM, capsid assembly modulator; PD-1, programmed cell-death 1; Peg-IFNα, pegylated interferon alpha; siRNA, small-interfering RNA.

Abbreviations

ALT
alanine aminotransferase
ASO
antisense oligonucleotide
CAM
capsid assembly modulator
cccDNA
covalently closed circular DNA
CHB
chronic hepatitis B
EOT
end-of-treatment
FXR
Farnesoid X receptor
GalNAc
N-acetylgalactosamine
HBcAg
hepatitis B core antigen
HBeAg
hepatitis B e antigen
HBsAg
hepatitis B surface antigen
HBV
hepatitis B virus
HCC
hepatocellular carcinoma
HDV
hepatitis D virus
IAP
Inhibitors of apoptosis
ImmTAV
Immune mobilizing monoclonal T-cell receptors against virus
mRNA
messenger RNA
NAP
nucleic acid polymer
NTCP
sodium taurocholate cotransporting polypeptide
NUC
nucleos(t)ide analogue
PD1
programmed cell death protein 1
PDL1
programmed cell death ligand 1
Peg-IFNα
pegylated interferon alpha
pgRNA
pregenomic RNA
rcDNA
relaxed circular DNA
RNAi
RNA interference
STOPS
S antigen Transport-inhibiting Oligonucleotide Polymers
TAF
tenofovir alafenamide
TLR
toll-like receptor

REFERENCES

1. World Health Organization (WHO). Guidelines for the prevention, diagnosis, care and treatment for people with chronic hepatitis B infection. WHO web site, <https://www.who.int/publications/i/item/9789240090903>. Accessed 25 Sep 2024.

2. Ghany MG, Buti M, Lampertico P, Lee HM; 2022 AASLD-EASL HBV-HDV Treatment Endpoints Conference Faculty. Guidance on treatment endpoints and study design for clinical trials aiming to achieve cure in chronic hepatitis B and D: report from the 2022 AASLD-EASL HBV-HDV Treatment Endpoints Conference. Hepatology 2023;78:1654-1673.
crossref pmid
3. Mak LY, Seto WK, Hui RW, Fung J, Wong DK, Lai CL, et al. Fibrosis evolution in chronic hepatitis B e antigen-negative patients across a 10-year interval. J Viral Hepat 2019;26:818-827.
crossref pmid pdf
4. Mak LY, Hui RW, Chung MSH, Wong DK, Fung J, Seto WK, et al. Regression of liver fibrosis after HBsAg loss: A prospective matched case-control evaluation using transient elastography and serum enhanced liver fibrosis test. J Gastroenterol Hepatol 2024;39:2826-2834.
crossref pmid pmc
5. Hui RW, Mak LY, Cheung TT, Lee VH, Seto WK, Yuen MF. Clinical practice guidelines and real-life practice on hepatocellular carcinoma: the Hong Kong perspective. Clin Mol Hepatol 2023;29:217-229.
crossref pmid pmc pdf
6. Hui RW, Mak LY, Seto WK, Yuen MF, Fung J. Chronic hepatitis B: a scoping review on the guidelines for stopping nucleos(t)ide analogue therapy. Expert Rev Gastroenterol Hepatol 2023;17:443-450.
crossref pmid
7. Yeo YH, Ho HJ, Yang HI, Tseng TC, Hosaka T, Trinh HN, et al. Factors associated with rates of HBsAg seroclearance in adults with chronic HBV infection: a systematic review and meta-analysis. Gastroenterology 2019;156:635-646.e9.
crossref pmid
8. Zoutendijk R, Hansen BE, van Vuuren AJ, Boucher CA, Janssen HL. Serum HBsAg decline during long-term potent nucleos(t)ide analogue therapy for chronic hepatitis B and prediction of HBsAg loss. J Infect Dis 2011;204:415-418.
crossref pmid
9. Chevaliez S, Hézode C, Bahrami S, Grare M, Pawlotsky JM. Long-term hepatitis B surface antigen (HBsAg) kinetics during nucleoside/nucleotide analogue therapy: finite treatment duration unlikely. J Hepatol 2013;58:676-683.
crossref pmid
10. Buster EH, Flink HJ, Cakaloglu Y, Simon K, Trojan J, Tabak F, et al. Sustained HBeAg and HBsAg loss after long-term follow-up of HBeAg-positive patients treated with peginterferon alpha-2b. Gastroenterology 2008;135:459-467.
crossref pmid
11. Wong VW, Wong GL, Yan KK, Chim AM, Chan HY, Tse CH, et al. Durability of peginterferon alfa-2b treatment at 5 years in patients with hepatitis B e antigen-positive chronic hepatitis B. Hepatology 2010;51:1945-1953.
crossref pmid
12. Hui RW, Mak LY, Seto WK, Yuen MF. Assessing the developing pharmacotherapeutic landscape in hepatitis B treatment: a spotlight on drugs in phase II clinical trials. Expert Opin Emerg Drugs 2022;27:127-140.
crossref pmid
13. Yuen MF, Lai CL. Hepatitis B in 2014: HBV research moves forward--receptors and reactivation. Nat Rev Gastroenterol Hepatol 2015;12:70-72.
crossref pmid pdf
14. Watashi K, Urban S, Li W, Wakita T. NTCP and beyond: opening the door to unveil hepatitis B virus entry. Int J Mol Sci 2014;15:2892-2905.
crossref pmid pmc
15. 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;72:972-983.
crossref pmid pmc
16. Mak LY, Hui RW, Fung J, Seto WK, Yuen MF. The role of different viral biomarkers on the management of chronic hepatitis B. Clin Mol Hepatol 2023;29:263-276.
crossref pmid pmc pdf
17. Wedemeyer H, Aleman S, Brunetto MR, Blank A, Andreone P, Bogomolov P, et al.; MYR 301 Study Group. A phase 3, randomized trial of bulevirtide in chronic hepatitis D. N Engl J Med 2023;389:22-32.
crossref pmid
18. Ali AH, Carey EJ, Lindor KD. Recent advances in the development of farnesoid X receptor agonists. Ann Transl Med 2015;3:5.
pmid pmc
19. Oehler N, Volz T, Bhadra OD, Kah J, Allweiss L, Giersch K, et al. Binding of hepatitis B virus to its cellular receptor alters the expression profile of genes of bile acid metabolism. Hepatology 2014;60:1483-1493.
crossref pmid
20. Erken R, Andre P, Roy E, Kootstra N, Barzic N, Girma H, et al. Farnesoid X receptor agonist for the treatment of chronic hepatitis B: a safety study. J Viral Hepat 2021;28:1690-1698.
crossref pmid pmc pdf
21. Hui RW, Mak LY, Seto WK, Yuen MF. RNA interference as a novel treatment strategy for chronic hepatitis B infection. Clin Mol Hepatol 2022;28:408-424.
crossref pmid pmc pdf
22. Wooddell CI, Yuen MF, Chan HL, Gish RG, Locarnini SA, Chavez D, et al. RNAi-based treatment of chronically infected patients and chimpanzees reveals that integrated hepatitis B virus DNA is a source of HBsAg. Sci Transl Med 2017;9:eaan0241.
crossref pmid pmc
23. Yuen MF, Lim SG, Plesniak R, Tsuji K, Janssen HLA, Pojoga C, G, et al.; B-Clear Study Group. Efficacy and safety of bepirovirsen in chronic hepatitis B infection. N Engl J Med 2022;387:1957-1968.
crossref pmid
24. Gadano A, Arbune M, Atsukawa M, Fujiyama S, Gankina NUGU, John B, et al. Evidence of durable response to bepirovirsen in B-Clear responders: B-Sure first annual report. J Hepatol 2023;78(Supplement 1):S110-S111.
crossref
25. Yuen MF, Heo J, Kumada H, Suzuki F, Suzuki Y, Xie Q, et al. Phase IIa, randomised, double-blind study of GSK3389404 in patients with chronic hepatitis B on stable nucleos(t)ide therapy. J Hepatol 2022;77:967-977.
crossref pmid
26. Mak LY, Hui RW, Fung J, Seto WK, Yuen MF. Bepirovirsen (GSK3228836) in chronic hepatitis B infection: an evaluation of phase II progress. Expert Opin Investig Drugs 2023;32:971-983.
crossref pmid
27. Yuen MF, Locarnini S, Lim TH, Strasser SI, Sievert W, Cheng W, et al. Combination treatments including the small-interfering RNA JNJ-3989 induce rapid and sometimes prolonged viral responses in patients with CHB. J Hepatol 2022;77:1287-1298.
crossref pmid
28. Gane E, Lim YS, Kim JB, Jadhav V, Shen L, Bakardjiev AI, et al. Evaluation of RNAi therapeutics VIR-2218 and ALN-HBV for chronic hepatitis B: results from randomized clinical trials. J Hepatol 2023;79:924-932.
crossref pmid
29. Gane EJ, Kim W, Lim TH, Tangkijvanich P, Yoon JH, Sievert W, et al. First-in-human randomized study of RNAi therapeutic RG6346 for chronic hepatitis B virus infection. J Hepatol 2023;79:1139-1149.
crossref pmid
30. Seto WK, Liang Z, Gan LM, Fu J, Yuen MF. Safety and antiviral activity of RBD1016, a RNAi therapeutic, in Chinese subjects with chronic hepatitis B virus (HBV) infection. J Hepatol 2023;78(Supplement 1):S1152.
crossref
31. Yuen MF, Holmes J, Strasser SI, Leerapun A, Sukeepaisarnjaroen W, Tangkijvanich P, et al. 48 weeks of AB-729+nucleos(t) ide analogue (NA) therapy results in profound, sustained HBsAg declines in both HBeAg+ and HBeAg- subjects which are maintained in HBeAg- subjects who have discontinued all therapy. Paper presented at: Global Hepatitis Summit; 2023; Paris, France.

32. Mak LY, Wooddell CI, Lenz O, Schluep T, Hamilton J, Davis HL, et al. Long-term hepatitis B surface antigen response after finite treatment of ARC-520 or JNJ-3989. Gut 2025;74:440-450.
crossref pmid
33. Hui RWH, Mak LY, Seto WK, Yuen MF. Role of core/capsid inhibitors in functional cure strategies for chronic hepatitis B. Curr Hepatol Rep 2020;19:293-301.
crossref pdf
34. Mak LY, Hui RW, Seto WK, Yuen MF. Novel drug development in chronic hepatitis B infection: Capsid assembly modulators and nucleic acid polymers. Clin Liver Dis 2023;27:877-893.
crossref pmid
35. Yuen MF, Agarwal K, Ma X, Nguyen TT, Schiff ER, Hann HL, et al. Safety and efficacy of vebicorvir in virologically suppressed patients with chronic hepatitis B virus infection. J Hepatol 2022;77:642-652.
crossref pmid
36. Yuen MF, Fung S, Ma X, Nguyen TT, Hassanein T, Hann HW, et al. Long-term open-label vebicorvir for chronic HBV infection: Safety and off-treatment responses. JHEP Rep 2024;6:100999.
crossref pmid pmc
37. Yuen MF, Zhou X, Gane E, Schwabe C, Tanwandee T, Feng S, et al. Safety, pharmacokinetics, and antiviral activity of RO7049389, a core protein allosteric modulator, in patients with chronic hepatitis B virus infection: a multicentre, randomised, placebo-controlled, phase 1 trial. Lancet Gastroenterol Hepatol 2021;6:723-732.
crossref pmid
38. Yuen MF, Gane EJ, Agarwal K, Niu J, Ren H, Ding Y, et al. Extended treatment of HBeAg+ CHB subjects with the Capsid assembly modulator ALG-000184 with or without Entecavir is associated with reductions in viral markers and favorable anti-HBeAb trends. J Hepatol 2024;80:S807-S808.

39. 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;158:2180-2194.
crossref pmid
40. 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 2021;27:335-348.
crossref pmid pmc
41. Zheng P, Dou Y, Wang Q. Immune response and treatment targets of chronic hepatitis B virus infection: innate and adaptive immunity. Front Cell Infect Microbiol 2023;13:1206720.
crossref pmid pmc
42. Park JJ, Wong DK, Wahed AS, Lee WM, Feld JJ, Terrault N, et al. Hepatitis B virus--Specific and global T-cell dysfunction in chronic hepatitis B. Gastroenterology 2016;150:684-695.e5.
pmid
43. Ma Z, Zhang E, Yang D, Lu M. Contribution of Toll-like receptors to the control of hepatitis B virus infection by initiating antiviral innate responses and promoting specific adaptive immune responses. Cell Mol Immunol 2015;12:273-282.
crossref pmid pmc pdf
44. Gane EJ, Dunbar PR, Brooks AE, Zhang F, Chen D, Wallin JJ, et al. Safety and efficacy of the oral TLR8 agonist selgantolimod in individuals with chronic hepatitis B under viral suppression. J Hepatol 2023;78:513-523.
crossref pmid
45. Janssen HL, Lim YS, Kim HJ, Sowah L, Tseng CH, Coffin CS, et al. Safety, pharmacodynamics, and antiviral activity of selgantolimod in viremic patients with chronic hepatitis B virus infection. JHEP Rep 2023;6:100975.
crossref pmid pmc
46. Yuen MF, Balabanska R, Cottreel E, Chen E, Duan D, Jiang Q, et al. TLR7 agonist RO7020531 versus placebo in healthy volunteers and patients with chronic hepatitis B virus infection: a randomised, observer-blind, placebo-controlled, phase 1 trial. Lancet Infect Dis 2023;23:496-507.
crossref pmid
47. 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;68:431-440.
crossref pmid
48. Agarwal K, Ahn SH, Elkhashab M, Lau AH, Gaggar A, Bulusu A, et al. Safety and efficacy of vesatolimod (GS-9620) in patients with chronic hepatitis B who are not currently on antiviral treatment. J Viral Hepat 2018;25:1331-1340.
crossref pmid pdf
49. Wang G, Cui Y, Xie Y, Mao Q, Xie Q, Gu Y, et al. ALT flares were linked to HBsAg reduction, seroclearance and seroconversion: interim results from a phase IIb study in chronic hepatitis B patients with 24-week treatment of subcutaneous PD-L1 Ab ASC22 (Envafolimab) plus nucleos (t) ide analogs. J Hepatol 2022;77:S70.
crossref
50. Wang G, Cui Y, Hu G, Fu L, Lu J, Chen Y, et al. HBsAg loss in chronic hepatitis B patients after 24-week treatment with subcutaneously administered PD-L1 antibody ASC22 (Envafolimab): Interim results from a phase IIb extension cohort. Paper presented at: The Liver Meeting 2023; Boston, MA, USA.

51. Zhang X, Huang K, Zhu W, Zhang G, Chen J, Huang X, et al. Targeting inhibitor of apoptosis proteins (IAPs) enhances intrahepatic antiviral immunity to clear hepatitis B virus infection in vivo. J Hepatol 2020;73:S5-S6.
crossref
52. Fergusson JR, Wallace Z, Connolly MM, Woon AP, Suckling RJ, Hine DW, et al. Immune-mobilizing monoclonal T cell receptors mediate specific and rapid elimination of hepatitis B-infected cells. Hepatology 2020;72:1528-1540.
crossref pmid pmc pdf
53. Vandepapelière P, Lau GK, Leroux-Roels G, Horsmans Y, Gane E, Tawandee T, et al. Therapeutic vaccination of chronic hepatitis B patients with virus suppression by antiviral therapy: a randomized, controlled study of co-administration of HBsAg/AS02 candidate vaccine and lamivudine. Vaccine 2007;25:8585-8597.
crossref pmid
54. Cargill T, Barnes E. Therapeutic vaccination for treatment of chronic hepatitis B. Clin Exp Immunol 2021;205:106-118.
crossref pmid pmc pdf
55. Tak WY, Chuang WL, Chen CY, Tseng KC, Lim YS, Lo GH, et al. Phase Ib/IIa randomized study of heterologous ChAdOx1-HBV/MVA-HBV therapeutic vaccination (VTP-300) as monotherapy and combined with low-dose nivolumab in virally-suppressed patients with CHB. J Hepatol 2024;81:949-959.
crossref pmid
56. Ma H, Lim TH, Leerapun A, Weltman M, Jia J, Lim YS, et al. Therapeutic vaccine BRII-179 restores HBV-specific immune responses in patients with chronic HBV in a phase Ib/IIa study. JHEP Rep 2021;3:100361.
crossref pmid pmc
57. Agarwal K, Yuen MF, Wedemeyer H, Cloutier D, Shen L, Arizpe A, et al. Reduction in hepatitis B viral DNA and hepatitis B surface antigen following administration of a single dose of VIR3434, an investigational neutralizing monoclonal antibody: First experience in a population with viremia. Hepatology 2022;76:S303-S304.

58. Lee HW, Park JY, Hong T, Park MS, Ahn SH. Efficacy of lenvervimab, a recombinant human immunoglobulin, in treatment of chronic hepatitis B virus infection. Clin Gastroenterol Hepatol 2020;18:3043-3045.e1.
crossref pmid
59. Hui RWH, Mak LY, Cheung KS, Fung J, Seto WK, Yuen MF. Novel combination strategies with investigational agents for functional cure of chronic hepatitis B infection. Curr Hepatol Rep 2022;21:59-67.
crossref pdf
60. Yuen MF, Asselah T, Jacobson IM, Brunetto MR, Janssen HLA, Takehara T, et al. Efficacy and safety of the siRNA JNJ-73763989 and the capsid assembly modulator JNJ-56136379 (bersacapavir) with nucleos(t)ide analogues for the treatment of chronic hepatitis B virus infection (REEF-1): a multicentre, double-blind, active-controlled, randomised, phase 2b trial. Lancet Gastroenterol Hepatol 2023;8:790-802.
pmid
61. Agarwal K, Buti M, van Bömmel F, Lampertico P, Janczewska E, Bourliere M, et al. JNJ-73763989 and bersacapavir treatment in nucleos(t)ide analogue-suppressed patients with chronic hepatitis B: REEF-2. J Hepatol 2024;81:404-414.
crossref pmid
62. MacQuillan G, Elkhashab M, Antonov K, Valaydon Z, Davidson S, Fung SK, et al. Preliminary off-treatment responses following 48 weeks of vebicorvir, nucleos (t) ide reverse transcriptase inhibitor, and AB-729 combination in virologically suppressed patients with hepatitis B e antigen negative chronic hepatitis B: analysis from an open-label phase 2 study. Hepatology 2023;78(S1):S47-S48.

63. Tait D, Bussey L, Kolenovska R, Downs M, Anderson K, Vardeu A, et al. VTP-300 immunotherapeutic, plus low dose PD-1 inhibitor, nivolumab, continues to show meaningful, sustained reductions in HBsAg levels. J Hepatol 2024;80:S811.

64. Buti M, Heo J, Tanaka Y, Andreone P, Atsukawa M, Cabezas J, et al. Pegylated interferon reduces relapses following bepirovirsen treatment in participants with chronic hepatitis b virus infection on nucleos (t) ide analogues: end of study results from the phase 2b B-Together study. Hepatology 2023;78:S49-S53.
crossref
65. Yuen MF, Heo J, Nahass RG, Wong G, Burda T, Bhamidimarri KR, et al. Imdusiran (AB-729) administered every 8 weeks in combination with 24 weeks of pegylated interferon alfa-2a in virally suppressed, HBeAg-negative subjects with chronic HBV infection leads to HBsAg loss in some subjects at end of IFN treatment. J Hepatol 2024;80(S1):S809-S810.

66. Yuen MF, Lim YS, Yoon KT, Lim TH, Heo J, Tangkijvanich P, et al. VIR-2218 (elebsiran) plus pegylated interferon-alfa-2a in participants with chronic hepatitis B virus infection: a phase 2 study. Lancet Gastroenterol Hepatol 2024;9:1121-1132.
crossref pmid
67. Kennedy PT, Fung SK, Buti M, Yilmaz G, Chuang WL, Asselah T, et al. Efficacy and safety of siRNA JNJ-73763989, capsid assembly modulator-E (CAM-E) JNJ-56136379, and nucleos (t) ide analogs (NA) with pegylated interferon alpha-2a (PEGIFN-α2a) added in immune-tolerant patients with chronic hepatitis B virus (HBV) infection: interim results from the phase 2 REEF-IT study. Hepatology 2023;78(S1):S568-S569.

68. Agarwal K, Yuen MF, Roberts S, Lo GH, Hsu CW, Chuang WL, et al. Imdusiran (AB-729) administered every 8 weeks for 24 weeks followed by the immunotherapeutic VTP-300 maintains lower HBV surface antigen levels in NA-suppressed CHB subjects than 24 weeks of imdusiran alone. J Hepatol 2024;80(S1):S26-S27.

69. Bourgeois S, Buti M, Gane EJ, Agarwal K, Janczewska E, Hsu YCH, et al. Efficacy, safety, tolerability, and immunogenicity of JNJ-0535 following a reduction of viral antigen levels through administration of siRNA JNJ-3989 in patients with chronic HBeAg negative hepatitis B-interim data of the OSPREY study. J Hepatol 2024;80:S79-S80.

70. Ji Y, Le Bert N, Lee A, Zhu C, Tan Y, Liu W, et al. Therapeutic vaccine (BRII-179) induced immune response associated with HBsAg reduction in a subset of chronic hepatitis B participants. J Hepatol 2024;80:S36.

71. Hou J, Xie Q, Zhang W, Hua R, Tang H, Gane EJ, et al. Efficacy and safety of xalnesiran with and without an immunomodulator in virologically suppressed participants with chronic hepatitis B: end of study results from the phase 2, randomized, controlled, adaptive, open-label platform study (PIRANGA). J Hepatol 2024;80:S26.

72. Asselah T, Fung SK, Akhan S, Chuang WL, Buti M, Brunetto M, et al. A phase 2 open-label study to evaluate safety, tolerability, efficacy, and pharmacodynamics of JNJ-73763989, nucleos (t) ide analogs, and a low-dose PD-1 inhibitor in patients with chronic hepatitis B-Interim results of the OCTOPUS-1 study. J Hepatol 2024;80(S1):S27-S28.

73. Gane EJ, Jucov A, Dobryanksa M, Lim TH, Arizpe A, Cloutier D, et al. Safety and antiviral activity of short-duration combinations of the investigational small interfering ribonucleic acid VIR-2218 with the neutralizing, vaccinal monoclonal antibody VIR-3434: post-treatment follow-up from the Phase 2 MARCH trial. J Hepatol 2023;78:S34-S35.
crossref
74. Agarwal K, Lok J, Carey I, Shivkar Y, Biermer M, Berg T, et al. A case of HBV-induced liver failure in the REEF-2 phase II trial: Implications for finite treatment strategies in HBV ‘cure’. J Hepatol 2022;77:245-248.
crossref pmid
75. Leung RH, Hui RW, Mak LY, Mao X, Liu KS, Wong DK, et al. ALT to qHBsAg ratio predicts long-term HBsAg seroclearance after entecavir cessation in Chinese patients with chronic hepatitis B. J Hepatol 2024;81:218-226.
crossref pmid
76. Hogan T, Castaneda EG, Garcia EP, Barnard R, Ray A, Paff M, et al. High-dimensional analysis of flow cytometry data reveals differences in post-treatment frequencies of naive B cells, CD56dim natural killer cells, and terminally differentiated effector memory CD8+ T cells in responders versus nonresponders to bepirovirsen. J Hepatol 2024;80:S807.

77. Singh J, Salaun B, You S, Corson S, Paff M, Theodore D. Cell-mediated immunity analysis to assess the characteristics of immune response to bepirovirsen: Examples from the B-Clear study. J Hepatol 2023;78:S1167-S1169.
crossref
78. Hui RW, Wong DK, Ho DW, Lyu X, Mak LY, Fung J, et al. HBV DNA integration profiles in the natural history of chronic hepatitis B. Hepatol Int 2024;18(Suppl1):50-51.
pmid
79. Kostyushev D, Kostyusheva A, Brezgin S, Ponomareva N, Zakirova NF, Egorshina A, et al. Depleting hepatitis B virus relaxed circular DNA is necessary for resolution of infection by CRISPR-Cas9. Mol Ther Nucleic Acids 2023;31:482-493.
crossref pmid pmc
80. Gorsuch CL, Nemec P, Yu M, Xu S, Han D, Smith J, et al. Targeting the hepatitis B cccDNA with a sequence-specific ARCUS nuclease to eliminate hepatitis B virus in vivo. Mol Ther 2022;30:2909-2922.
crossref pmid pmc
81. Tune Therapeutics. Epigenetic editing for the treatment of HBV 2023. Tune Therapeutics web site, <https://tunetx.com/wp-content/uploads/2023/12/HEPDART_Dec5_Web.pdf >. Accessed 25 Sep 2024.

82. Cui F, Blach S, Manzengo Mingiedi C, Gonzalez MA, Sabry Alaama A, Mozalevskis A, et al. Global reporting of progress towards elimination of hepatitis B and hepatitis C. Lancet Gastroenterol Hepatol 2023;8:332-342.
crossref pmid
83. Polaris Observatory Collaborators. Global prevalence, cascade of care, and prophylaxis coverage of hepatitis B in 2022: a modelling study. Lancet Gastroenterol Hepatol 2023;8:879-907.
pmid

Editorial Office
The Korean Association for the Study of the Liver
Room A1210, 53 Mapo-daero(MapoTrapalace, Dowha-dong), Mapo-gu, Seoul, 04158, Korea
TEL: +82-2-703-0051   FAX: +82-2-703-0071    E-mail: cmh_journal@ijpnc.com
Copyright © The Korean Association for the Study of the Liver.         
COUNTER
TODAY : 1605
TOTAL : 2560085
Close layer