Next Article in Journal
Vaccines against Major Poultry Viral Diseases: Strategies to Improve the Breadth and Protective Efficacy
Previous Article in Journal
Hepatitis C Virus Epidemiology in Lithuania: Situation before Introduction of the National Screening Programme
Previous Article in Special Issue
Early Treatment Consideration in Patients with Hepatitis B ‘e’ Antigen-Positive Chronic Infection: Is It Time for a Paradigm Shift?
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Editorial

Targeting Subviral Particles: A Critical Step in Achieving HBV Functional Cure but Where Are We with Current Agents in Clinical Development?

Replicor Inc., 6100 Royalmount Avenue, Montreal, QC H4P 2R2, Canada
Viruses 2022, 14(6), 1193; https://doi.org/10.3390/v14061193
Submission received: 9 May 2022 / Revised: 27 May 2022 / Accepted: 30 May 2022 / Published: 31 May 2022
The recent review [1] by Drs. Moini and Fung on HBsAg loss as an important treatment endpoint for functional cure provides a good overview of the different compounds in development for the treatment of chronic HBV. However, some clarifications are warranted based on the evolution of clinical data in the field with a variety of investigational agents over recent months.
ALG-010133 is a nucleic acid polymer (NAP) that was under development by Aligos Therapeutics. This NAP is a locked nucleic acid (LNA)-modified version of the NAP REP 2165 [2] previously developed by Replicor Inc. The LNA modification was abandoned more than a decade ago during the development of nucleic acid polymers (NAPs) at Replicor. Predictably, ALG-010133 monotherapy is accompanied by significant injection site reactions [3] and has no activity in HBV infected patients at the 400 mg dose [4], whereas active NAPs (REP 2139 and REP 2055) achieve up to 7 log10 IU/mL HBsAg reduction from baseline with monotherapy at similar doses and dosing duration [5]. The development of all LNA-modified NAPs has been halted [4]. The mechanistic reasons for the failure for ALG-010133 and why the LNA modification was discarded early in the development of NAPs have been recently detailed [2]. Development of the HBV LNA antisense oligonucleotide (ASO) ALG-020572 has also been canceled due to hepatoxicity [6], an expected complication of the LNA modification that was employed in this ASO [2].
A recent study demonstrating the ability of cccDNA to establish and maintain a stable cccDNA copy number in the absence of the core antigen [7] draws into question the potential clinical impact of capsid assembly modulators (CAMs) in chronic HBV infection. Several class I and class II capsid assembly modulators (CAMs) have been evaluated in monotherapy. As expected, all have elicited strong declines in HBV DNA and HBV RNA in agreement with their mechanism of action; however, none have shown any effects on HBsAg in the absence of nucleos(t)ide analogs (NUCs). Moreover, the lack of effect of CAM monotherapy on cccDNA activity (HBeAg and HBcrAg), despite the rapid turnover of active cccDNA [8] is consistent with the lack of requirement of HBcAg for cccDNA maintenance that was previously described [7]. On the other hand, all NUCs, either approved or in development (ETV, TDF, TAF, or the protide version of clevudine [ATI-2173]), have well-described secondary effects in stimulating innate immunity [9,10,11,12,13,14,15,16]. Moreover, in the clinic, NUC monotherapy is accompanied by mild declines in HBsAg and stronger declines and/or clearance of HBeAg, HBV RNA and HBcrAg [17,18,19,20,21]. As such, the universal rebound of infection following long term combination therapy with NUCs and vebicorvir [22] and the subsequent halt of its development draws into question the contribution that CAMs can make in the pursuit of a functional cure. The field is anticipating results with more potent CAMs in development.
It should be noted that nucleic acid polymers (NAPs) are accompanied by a reduction in HBsAg that is far greater than the typical 2–3 log10 IU/mL reduction from baseline that was observed with RNAi when combined with NUCs and CAMs [23]. NAPs achieve HBsAg reductions of up to 8 log10 IU/mL from baseline [24] with 70% of patients achieving HBsAg < 1 IU/mL, 60% of whom achieve HBsAg loss (<0.005 IU/mL) [21,24]. These effects of NAPs on circulating HBsAg are attributed to their ability to selectively target the assembly and secretion of HBV subviral particles (SVP) [25] which constitute >99.99% of circulating HBsAg [26]. This antiviral effect was recently validated in clinical studies where the small isoform of HBsAg was selectively cleared during therapy with NAPs, which is diagnostic of targeting of SVPs [27]. Importantly, NAPs have no effect on the production and or release of Dane particles or HBeAg [25].
Contrary to what is stated by Moini et al. [1], quantitative HBsAg is not well correlated with intrahepatic cccDNA [28,29,30,31] even in HBeAg positive patients [29], due to the contribution of SVP to the HBsAg pool. SVPs are produced from both active cccDNA and integrated HBV DNA [32]; integration is an early and cumulative event over the natural progression of chronic HBV infection. HBV DNA integration is present in HBeAg positive infection [33], albeit at lower levels than in HBeAg negative infection.
The targeting of HBsAg loss must include the efficient removal of SVP. In this regard, several fundamental misconceptions still exist regarding the mechanisms by which HBsAg reduction is achieved during therapy with RNAi or ASO compounds that are designed to target the degradation of HBV mRNA. HBV infection has a very high degree of genetic plasticity (thousands of quasispecies exist in any particular patient) [34], a result of the lack of proofreading activity of the HBV reverse transcriptase, the rapid turnover of cccDNA and the constant immune pressure that is exerted on the virus. Given that single point mutations abolish the ability of RNAi or ASOs to target specific mRNA cleavage [35], the theoretical basis for the consideration of RNAi or ASO approaches is not clear. This was borne out by the failure of ARB-1467 (TKM-HBV), an LNP formulated RNAi with three siRNA triggers: two in HBsAg and one in HBx (thus covering cccDNA and integrated HBV DNA). LNP formulation of siRNA is the most efficient delivery method for RNAi compounds, resulting in the functional uptake of RNAi compounds, exclusively in hepatocytes [36,37]. In NUC-suppressed chronic HBV infection, ARB-1467 had minimal effects on HBsAg and, importantly, initial mild declines in HBV RNA and HBcrAg rapidly rebounded during treatment [38]. Additionally, a recent HBsAg isoform analysis (using the same assay platform that was used to evaluate NAPs) during therapy with AB-729 and a GalNAc-conjugated single RNAi trigger in HBx revealed no targeting of SVP, but rather the reverse, selective targeting of virions [39]. Both observations exclude mRNA degradation as a potential mechanism for HBsAg reduction by compounds designed as RNAi or ASO for chronic HBV infection.
What then is the mechanism underlying the HBsAg reductions that were observed with RNAi and the lone remaining HBV ASO compound in development (bepirovirsen)? All oligonucleotides have off-target immunoreactive properties. In the case of single stranded DNA, it is the stimulation of the innate immune response via TRL9 by CpG motifs [40]. Bepirovirsen (GSK 3228836) has a cryptic class II CpG motif in its 5′ end [26]. Unconjugated bepirovirsen accumulates mainly in Kupffer cells (as is the case for all phosphorothioate oligonucleotides), while GalNAc-conjugated bepirovirsen (GSK3389404) accumulates primarily in hepatocytes [41]. Unconjugated bepirovirsen therapy in chronic HBV infection leads to strong HBsAg reduction but only in patients with low HBsAg (<1000 IU/mL) at baseline, an observation that is inconsistent with an antisense effect [42]. This HBsAg decline was accompanied by the strong activation of immune responses consistent with the stimulation of TLR9 [43]. The GalNAc-conjugated variant of bepirovirsen (GSK 3389404) has none of these activities in chronic HBV infection [44], again excluding an antisense effect.
For any RNAi, stimulation of TLR3 is a foregone conclusion: the stimulation of TLR3 occurs with any double-stranded RNA (dsRNA) and is sequence independent [40]. It is possible to quench the TLR reactivity of dsRNA with extensive modification but this blocks the RISC-loading that is required for an RNAi effect [45]. RISC-mediated mRNA cleavage with RNAi reliably elicits a rapid effect on protein levels with a variety of liver proteins, yielding a 1 log10 reduction in protein levels within 15 days following the first RNAi dose. For all GalNAc-RNA evaluated to date in chronic HBV infection, many subjects did not experience any significant or reductions fare weaker than a 1 log10 reduction in HBsAg from baseline for 4 weeks after the first dose. Given the rapid turnover rate of SVP with a ½ life of 1–6 days [46,47], the absence of a 1 log10 decline in HBsAg two weeks after the first administered dose of all GalNAc-RNAi (in addition to the HBsAg isoform response analysis during AB-729 treatment mentioned above) strongly indicates that an RNAi effect is absent in these patients.
In switching from LNP formulation to GalNAc conjugation for RNAi delivery, the functional accumulation of RNAi (relative to LNP) in Kupffer cells becomes substantially higher, allowing for enhanced stimulation of TLR3. For all GalNAc-RNAi evaluated to date in chronic HBV infection, a delayed response in HBsAg occurs universally one month after the first dose of GalNAc-RNAi, gradually becoming saturated at 1.5–2 log10 reduction from baseline. This HBsAg response is remarkably similar to the delayed HBsAg response to TLR3 stimulation by poly I:C in the HDI HBV mouse model [48]. All of the current HBsAg response data with ASOs and LNP- or GalNAc-RNAi in human trials are inconsistent with effects on HBsAg decline expected from the degradation of mRNA; however, HBsAg responses (as well as markers of cccDNA activity) are entirely consistent with the inactivation/degradation of cccDNA-mediated by TLR9/TLR3-stimulated innate immune responses from these compounds.
A critical milestone in the achievement of functional cure is the clearance of HBsAg, which in turn requires elimination of SVP from the blood. Understanding the mechanisms, advantages and limitations of various agents is critical to designing optimal combination regimens which can achieve high rates of functional cure with finite therapy.

Funding

This research received no external funding.

Conflicts of Interest

AV is an employee and shareholder of Replicor Inc.

References

  1. Moini, M.; Fung, S. HBsAg Loss as a Treatment Endpoint for Chronic HBV Infection: HBV Cure. Viruses 2022, 14, 657. [Google Scholar] [CrossRef] [PubMed]
  2. Vaillant, A. Editorial: In vitro mechanistic evaluation of nucleic acid polymers: A cautionary tale. Mol. Ther. Nucleic Acids 2022, 28, 168–174. [Google Scholar] [CrossRef] [PubMed]
  3. Gane, E.; Yuen, M.F.; Yogaratnam, J.; Le, K.; Vuong, J.; Christopher, W.; Gohil, V.; Schwabe, C.; Agarwal, K.; Jucov, A.; et al. Safety, Tolerability and Pharmacokinetics (PK) of Single and Multiple Doses of ALG-010133, an S-antigen Transport Inhibiting Oligonucleotide Polymer (STOPS) for the Treatment of Chronic Hepatitis B. J. Hepatol. 2021, 75, S741. [Google Scholar]
  4. Aligos. Aligos Halting Further Development of STOPS™ Drug Candidate, ALG-010133. Press Release. 2022. Available online: https://investor.aligos.com/news-releases/news-release-details/aligos-halting-further-development-stopstm-drug-candidate-alg/ (accessed on 8 May 2022).
  5. Vaillant, A. REP 2139: Antiviral Mechanisms and Applications in Achieving Functional Control of HBV and HDV Infection. ACS Infect. Dis. 2019, 5, 675–687. [Google Scholar] [CrossRef]
  6. Aligos. Aligos Discontinues Development of its Antisense Oligonucleotide Drug Candidate ALG-020572 in Subjects with Chronic Hepatitis B and Pivots Internal Strategic Emphasis to its Small Molecule Portfolio. 2022. Available online: https://www.eatg.org/hiv-news/aligos-discontinues-development-of-its-drug-candidate-alg-020572-for-the-treatment-of-hepatitis-b/ (accessed on 31 May 2021).
  7. Tu, T.; Zehnder, B.; Qu, B.; Urban, S. De novo synthesis of hepatitis B virus nucleocapsids is dispensable for the maintenance and transcriptional regulation of cccDNA. JHEP Rep. 2021, 3, 100195. [Google Scholar] [CrossRef]
  8. Huang, Q.; Zhou, B.; Cai, D.; Zong, Y.; Wu, Y.; Liu, S.; Mercier, A.; Guo, H.; Hou, J.; Colonno, R.; et al. Rapid Turnover of Hepatitis B Virus Covalently Closed Circular DNA Indicated by Monitoring Emergence and Reversion of Signature-Mutation in Treated Chronic Hepatitis B Patients. Hepatology 2021, 73, 41–52. [Google Scholar] [CrossRef] [Green Version]
  9. Kmonickova, E.; Potmesil, P.; Holy, A.; Zidek, Z. Purine P1 receptor-dependent immunostimulatory effects of antiviral acyclic analogues of adenine and 2,6-diaminopurine. Eur. J. Pharmacol. 2006, 530, 179–187. [Google Scholar] [CrossRef]
  10. Potmesil, P.; Krecmerova, M.; Kmonickova, E.; Holy, A.; Zidek, Z. Nucleotide analogues with immunobiological properties: 9-[2-Hydroxy-3-(phosphonomethoxy)propyl]-adenine (HPMPA), -2,6-diaminopurine (HPMPDAP), and their N6-substituted derivatives. Eur. J. Pharmacol. 2006, 540, 191–199. [Google Scholar] [CrossRef]
  11. Murata, K.; Asano, M.; Matsumoto, A.; Sugiyama, M.; Nishida, N.; Tanaka, E.; Inoue, T.; Sakamoto, M.; Enomoto, N.; Shirasaki, T.; et al. Induction of IFN-lambda3 as an additional effect of nucleotide, not nucleoside, analogues: A new potential target for HBV infection. Gut 2018, 67, 362–371. [Google Scholar] [CrossRef]
  12. Murata, K.; Tsukuda, S.; Suizu, F.; Kimura, A.; Sugiyama, M.; Watashi, K.; Noguchi, M.; Mizokami, M. Immunomodulatory Mechanism of Acyclic Nucleoside Phosphates in Treatment of Hepatitis B Virus Infection. Hepatology 2020, 71, 1533–1545. [Google Scholar] [CrossRef] [PubMed]
  13. Lee, J.; Chuang, T.H.; Redecke, V.; She, L.; Pitha, P.M.; Carson, D.A.; Raz, E.; Cottam, H.B. Molecular basis for the immunostimulatory activity of guanine nucleoside analogs: Activation of Toll-like receptor 7. Proc. Natl. Acad. Sci. USA 2003, 100, 6646–6651. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Davenne, T.; Bridgeman, A.; Rigby, R.E.; Rehwinkel, J. Deoxyguanosine is a TLR7 agonist. Eur. J. Immunol. 2020, 50, 56–62. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Kurihara, M.; Tsuge, M.; Murakami, E.; Mori, N.; Ohishi, W.; Uchida, T.; Fujino, H.; Nakahara, T.; Abe-Chayama, H.; Kawaoka, T.; et al. The association between serum cytokine and chemokine levels and antiviral response by entecavir treatment in chronic hepatitis B patients. Antivir. Ther. 2018, 23, 239–248. [Google Scholar] [CrossRef] [Green Version]
  16. Vollmer, J.; Janosch, A.; Laucht, M.; Ballas, Z.K.; Schetter, C.; Krieg, A.M. Highly immunostimulatory CpG-free oligodeoxynucleotides for activation of human leukocytes. Antisense Nucleic Acid Drug Dev. 2002, 12, 165–175. [Google Scholar] [CrossRef]
  17. Mak, L.Y.; Wong, D.K.; Cheung, K.S.; Seto, W.K.; Fung, J.; Yuen, M.F. First-line oral antiviral therapies showed similar efficacies in suppression of serum HBcrAg in chronic hepatitis B patients. BMC Gastroenterol. 2021, 21, 123. [Google Scholar] [CrossRef] [PubMed]
  18. Liu, S.; Liu, Z.; Li, W.; Zhou, B.; Liang, X.; Fan, R.; Deng, R.; Hou, J.; Sun, J. Factors associated with the biphasic kinetics of serum HBV RNA in patients with HBeAg-positive chronic hepatitis B treated with nucleos(t)ide analogues. Aliment. Pharmacol. Ther. 2020, 52, 692–700. [Google Scholar] [CrossRef]
  19. van Bommel, F.; Bartens, A.; Mysickova, A.; Hofmann, J.; Kruger, D.H.; Berg, T.; Edelmann, A. Serum hepatitis B virus RNA levels as an early predictor of hepatitis B envelope antigen seroconversion during treatment with polymerase inhibitors. Hepatology 2015, 61, 66–76. [Google Scholar] [CrossRef]
  20. van Campenhout, M.J.; Brouwer, W.P.; van Oord, G.W.; Xie, Q.; Zhang, Q.; Zhang, N.; Guo, S.; Tabak, F.; Streinu-Cercel, A.; Wang, J.; et al. Hepatitis B core-related antigen levels are associated with response to entecavir and peginterferon add-on therapy in hepatitis B e antigen-positive chronic hepatitis B patients. Clin. Microbiol. Infect. 2016, 22, 571.e5–571.e9. [Google Scholar] [CrossRef] [Green Version]
  21. Bazinet, M.; Pantea, V.; Placinta, G.; Moscalu, I.; Cebotarescu, V.; Cojuhari, L.; Jimbei, P.; Iarovoi, L.; Smesnoi, V.; Musteata, T.; 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 Naive to Nucleos(t)ide Therapy. Gastroenterology 2020, 158, 2180–2194. [Google Scholar] [CrossRef]
  22. Gane, E.; Sulkowski, M.; Ma, X.; Nguyen, T.; Hann, H.W.; Hassanein, T.; Elkhashab, M.; Chan, S.; Nahass, R.; Bennett, M.; et al. Viral Response and Safety Following Discontinuation of Treatment with the Core Inhibitor Vebicorvir and a Nucleos (t)ide Reverse Transcriptase Inhibitor in Patients with HBeAg Positive or Negative Chronic Hepatitis B Virus Infection. J. Hepatol. 2021, 75, S736. [Google Scholar]
  23. Yuen, M.F.; Asselah, T.; Jacobson, I.M.; Brunetto, M.; Janssen, H.; Takehara, T.; Hou, J.L.; Kakuda, T.N.; Lambrecht, T.; Beaumont, M.; et al. Efficacy and Safety of the siRNA JNJ-3989 and/or the Capsid Assemb;y Modulator JNJ-6739 for the Treatment of Chronic Hepatitis B Virus Infection: Results From the Phase 2b REEF-1 Study. Hepatology 2021, 74, 1390A–1391A. [Google Scholar]
  24. Bazinet, M.; Anderson, M.; Pantea, V.; Placinta, G.; Moscalu, I.; Cebotarescu, V.; Cojuhari, L.; Jimbei, P.; Iarovoi, L.; Smesnoi, V.; et al. Analysis of HBsAg Immunocomplexes and cccDNA Activity During and Persisting After NAP-Based Therapy. Hepatol. Commun. 2021, 5, 1873–1887. [Google Scholar] [CrossRef] [PubMed]
  25. Boulon, R.; Blanchet, M.; Lemasson, M.; Vaillant, A.; Labonte, P. Characterization of the antiviral effects of REP 2139 on the HBV lifecycle in vitro. Antivir. Res. 2020, 183, 104853. [Google Scholar] [CrossRef] [PubMed]
  26. Vaillant, A. HBsAg, Subviral Particles, and Their Clearance in Establishing a Functional Cure of Chronic Hepatitis B Virus Infection. ACS Infect. Dis. 2020, 7, 1351–1368. [Google Scholar] [CrossRef] [PubMed]
  27. Bazinet, M.; Anderson, M.; Pantea, V.; Placinta, G.; Moscalu, I.; Cebotarescu, V.; Cojuhari, L.; Jimbei, P.; Iarovoi, L.; Smesnoi, V.; et al. HBsAg isoform dynamics during NAP-based therapy of HBeAg-negative chronic HBV and HBV/HDV infection. Hepatol. Commun. 2022. [Google Scholar] [CrossRef] [PubMed]
  28. Lai, C.L.; Wong, D.; Ip, P.; Kopaniszen, M.; Seto, W.K.; Fung, J.; Huang, F.Y.; Lee, B.; Cullaro, G.; Chong, C.K.; et al. Reduction of covalently closed circular DNA with long-term nucleos(t)ide analogue treatment in chronic hepatitis B. J. Hepatol. 2017, 66, 275–281. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  29. Ghany, M.G.; King, W.C.; Lisker-Melman, M.; Lok, A.S.F.; Terrault, N.; Janssen, H.L.A.; Khalili, M.; Chung, R.T.; Lee, W.M.; Lau, D.T.Y.; et al. Comparison of HBV RNA and Hepatitis B Core Related Antigen With Conventional HBV Markers Among Untreated Adults With Chronic Hepatitis B in North America. Hepatology 2021, 74, 2395–2409. [Google Scholar] [CrossRef]
  30. Peiffer, K.H.; Kuhnhenn, L.; Jiang, B.; Mondorf, A.; Vermehren, J.; Knop, V.; Susser, S.; Walter, D.; Dietz, J.; Carra, G.; et al. Divergent preS Sequences in Virion-Associated Hepatitis B Virus Genomes and Subviral HBV Surface Antigen Particles From HBV e Antigen-Negative Patients. J. Infect. Dis. 2018, 218, 114–123. [Google Scholar] [CrossRef] [Green Version]
  31. Hu, B.; Wang, R.; Fu, J.; Su, M.; Du, M.; Liu, Y.; Li, H.; Wang, H.; Lu, F.; Jiang, J. Integration of hepatitis B virus S gene impacts on hepatitis B surface antigen levels in patients with antiviral therapy. J. Gastroenterol. Hepatol. 2018, 33, 1389–1396. [Google Scholar] [CrossRef]
  32. Tu, T.; Budzinska, M.A.; Shackel, N.A.; Urban, S. HBV DNA Integration: Molecular Mechanisms and Clinical Implications. Viruses 2017, 9, 75. [Google Scholar] [CrossRef]
  33. Podlaha, O.; Wu, G.; Downie, B.; Ramamurthy, R.; Gaggar, A.; Subramanian, M.; Ye, Z.; Jiang, Z. Genomic modeling of hepatitis B virus integration frequency in the human genome. PLoS ONE 2019, 14, e0220376. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Domingo, E.; Sheldon, J.; Perales, C. Viral quasispecies evolution. Microbiol. Mol. Biol. Rev. 2012, 76, 159–216. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Vickers, T.A.; Koo, S.; Bennett, C.F.; Crooke, S.T.; Dean, N.M.; Baker, B.F. Efficient reduction of target RNAs by small interfering RNA and RNase H-dependent antisense agents. A comparative analysis. J. Biol. Chem. 2003, 278, 7108–7118. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. Woitok, M.M.; Zoubek, M.E.; Doleschel, D.; Bartneck, M.; Mohamed, M.R.; Kiessling, F.; Lederle, W.; Trautwein, C.; Cubero, F.J. Lipid-encapsulated siRNA for hepatocyte-directed treatment of advanced liver disease. Cell Death Dis. 2020, 11, 343. [Google Scholar] [CrossRef] [PubMed]
  37. Witzigmann, D.; Kulkarni, J.A.; Leung, J.; Chen, S.; Cullis, P.R.; van der Meel, R. Lipid nanoparticle technology for therapeutic gene regulation in the liver. Adv. Drug Deliv. Rev. 2020, 159, 344–363. [Google Scholar] [CrossRef] [PubMed]
  38. Agarwal, K.; Gane, E.; Cheng, C.; Sievert, W.; Roberts, S.K.; Ahn, S.H.; Kim, Y.J.; Streinu-Cercel, A.; Denning, J.; Symonds, W.; et al. HBcrAg, HBV-RNA declines in A Phase 2a Study Evaluating the Multi-dose Activity of ARB-1467 in HBeAg-Positive and Negative Virally Suppressed Subjects with Hepatitis B. Hepatology 2017, 66, 22A. [Google Scholar]
  39. Thi, E.P.; Yuen, M.F.; Gane, E.; Sevinsky, H.; Sims, K.; Anderson, M.; Lam, A.M.; Sofia, M.J.; Cloherty, G.; Picchio, G. Inhibition of Hepatitis B Surface Antigen by RNA Interference Therapeutic AB-729 in Chronic Hepatitis B Patients Correlates with Suppression of all HBsAg Isoforms and HBV RNA. J. Hepatol. 2021, 75, S760. [Google Scholar]
  40. Kawai, T.; Akira, S. The roles of TLRs, RLRs and NLRs in pathogen recognition. Int. Immunol. 2009, 21, 317–337. [Google Scholar] [CrossRef] [Green Version]
  41. Prakash, T.P.; Graham, M.J.; Yu, J.; Carty, R.; Low, A.; Chappell, A.; Schmidt, K.; Zhao, C.; Aghajan, M.; Murray, H.F.; et al. Targeted delivery of antisense oligonucleotides to hepatocytes using triantennary N-acetyl galactosamine improves potency 10-fold in mice. Nucleic Acids Res. 2014, 42, 8796–8807. [Google Scholar] [CrossRef] [Green Version]
  42. Yuen, M.F.; Heo, J.; Jang, J.W.; Yoon, J.H.; Kweon, Y.O.; Park, S.J.; Tami, Y.; You, S.; Yates, P.; Tao, Y.; et al. Safety, tolerability and antiviral activity of the antisense oligonucleotide bepirovirsen in patients with chronic hepatitis B: A phase 2 randomized controlled trial. Nat. Med. 2021, 27, 1725–1734. [Google Scholar] [CrossRef]
  43. You, S.; Singh, J.; Smith, S.; Jordan, W.; Remingler, K.; Joshi, S.; Ermler, M.; Delahaye, J.; Taylor, A.; Chakraborty, S.; et al. Treatment with GSK3228836 Leads to HBsAg Reduction and Induction of Interferon Gamma Related Proteins and Chemokines in a Phase 2a, Randomized, Double-Blind, Placebo-Controlled Study. J. Hepatol. 2021, 75, S455. [Google Scholar]
  44. Yuen, M.F.; Heo, J.; Kumada, H.; Suzuki, F.; Suzuki, Y.; Xie, Q.; Jia, J.; Karino, Y.; Hou, J.; Chayama, K.; et al. Results After 12 Weeks Treatment of Multiple Doses odf GSK3389404 in Chronic Hepatitis B (CHB) Subjects on Stable Nucleos(t)ide Therapy in a Phase 2a Double-Blind, Placebo-Controlled Study. Hepatology 2019, 70, 433A. [Google Scholar]
  45. Leuschner, P.J.; Ameres, S.L.; Kueng, S.; Martinez, J. Cleavage of the siRNA passenger strand during RISC assembly in human cells. EMBO Rep. 2006, 7, 314–320. [Google Scholar] [CrossRef] [PubMed]
  46. Loomba, R.; Decaris, M.; Li, K.W.; Shankaran, M.; Mohammed, H.; Matthews, M.; Richards, L.M.; Nguyen, P.; Rizo, E.; Andrews, B.; et al. Discovery of Half-life of Circulating Hepatitis B Surface Antigen in Patients With Chronic Hepatitis B Infection Using Heavy Water Labeling. Clin. Infect. Dis. 2019, 69, 542–545. [Google Scholar] [CrossRef] [PubMed]
  47. Shekhtman, L.; Cotler, S.J.; Hershkovich, L.; Uprichard, S.L.; Bazinet, M.; Pantea, V.; Cebotarescu, V.; Cojuhari, L.; Jimbei, P.; Krawczyk, A.; et al. Modelling hepatitis D virus RNA and HBsAg dynamics during nucleic acid polymer monotherapy suggest rapid turnover of HBsAg. Sci. Rep. 2020, 10, 7837. [Google Scholar] [CrossRef]
  48. Wu, J.; Huang, S.; Zhao, X.; Chen, M.; Lin, Y.; Xia, Y.; Sun, C.; Yang, X.; Wang, J.; Guo, Y.; et al. Poly(I:C) treatment leads to interferon-dependent clearance of hepatitis B virus in a hydrodynamic injection mouse model. J. Virol. 2014, 88, 10421–10431. [Google Scholar] [CrossRef] [Green Version]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Vaillant, A. Targeting Subviral Particles: A Critical Step in Achieving HBV Functional Cure but Where Are We with Current Agents in Clinical Development? Viruses 2022, 14, 1193. https://doi.org/10.3390/v14061193

AMA Style

Vaillant A. Targeting Subviral Particles: A Critical Step in Achieving HBV Functional Cure but Where Are We with Current Agents in Clinical Development? Viruses. 2022; 14(6):1193. https://doi.org/10.3390/v14061193

Chicago/Turabian Style

Vaillant, Andrew. 2022. "Targeting Subviral Particles: A Critical Step in Achieving HBV Functional Cure but Where Are We with Current Agents in Clinical Development?" Viruses 14, no. 6: 1193. https://doi.org/10.3390/v14061193

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop