Skip to main content
Global Health & Medicine logoLink to Global Health & Medicine
. 2023 Aug 31;5(4):199–207. doi: 10.35772/ghm.2023.01065

Biology of the hepatitis B virus (HBV) core and capsid assembly modulators (CAMs) for chronic hepatitis B (CHB) cure

William M McFadden 1,2, Stefan G Sarafianos 1,2,*
PMCID: PMC10461335  PMID: 37655181

Summary

Hepatitis B virus (HBV) is a hepadnavirus, a small DNA virus that infects liver tissue, with some unusual replication steps that share similarities to retroviruses. HBV infection can lead to chronic hepatitis B (CHB), a life-long infection associated with significant risks of liver disease, especially if untreated. HBV is a significant global health problem, with hundreds of millions currently living with CHB. Currently approved strategies to prevent or inhibit HBV are highly effective, however, a cure for CHB has remained elusive. To achieve a cure, elimination of the functionally integrated HBV covalently closed chromosomal DNA (cccDNA) genome is required. The capsid core is an essential component of HBV replication, serving roles when establishing infection and in creating new virions. Over the last two and a half decades, significant efforts have been made to find and characterize antivirals that target the capsid, specifically the HBV core protein (Cp). The antivirals that interfere with the kinetics and morphology of the capsid, termed capsid assembly modulators (CAMs), are extremely potent, and clinical investigations indicate they are well tolerated and highly effective. Several CAMs offer the potential to cure CHB by decreasing the cccDNA pools. Here, we review the biology of the HBV capsid, focused on Cp, and the development of inhibitors that target it.

Keywords: HBV, capsid assembly modulators (CAMs), hepatitis, hepadnaviridae, capsid core

Introduction

The hepatitis B virus (HBV) was first discovered as an infectious agent in the 1960s by Dr. Baruch Blumberg, who described the presence of what would later be named the HBV surface antigen (HBsAg) in the blood of patients with hepatitis (1). Blumberg's discovery subsequently led to the development of diagnostic screening tests and effective vaccines against HBV by targeting HBsAg, work for which he would later be awarded the 1976 Nobel Prize in Physiology or Medicine (2). As of 2019, the World Health Organization (WHO) estimates that approximately 1.5 million new HBV infections occur each year, with the highest burden of disease currently in the WHO Western Pacific Region and the WHO African Region (3). A fraction of adults (< 10%) with acute HBV infection will develop chronic hepatitis B (CHB), while acutely infected infants are at a much higher risk (~90%); a staggering 296 million individuals are estimated to be living with CHB (3,4). Despite the goals set by the WHO to decrease HBV-related mortality by 65% from 2015 to 2030, global HBV-related mortality is projected to increase by 39% during this time (5,6). Many factors contribute to this increased mortality (recently reviewed (6)) including, for example, economic disparities in HBV distribution between and within countries, comorbidities and viral coinfections like hepatitis D virus and human immunodeficiency virus (HIV), and societal stigmas that add barriers to receiving HBV healthcare (6,7).

Currently approved strategies to prevent or inhibit HBV are highly effective, including the HBV vaccine (90-95% effective), as well as repurposed nucleos(t) ide analogs (NUCs) from HIV reverse transcriptase (RT) inhibitors like tenofovir and lamivudine (3TC) (4,7,8). Although HBV is effectively inhibited by NUCs, caseation of antiviral therapy leads to unpredictable outcomes and often a rebound of viral load (9). Thus, the current standard of care for CHB is life-long treatment with NUCs. There is another approved class with curative potential, interferon-α or pegylated interferon-α (peg- IFNα), but it is not well tolerated and suppresses chronic infection in only 30% of patients (10). Currently, CHB has no reliable cure. Therefore, direct-acting agents that target outside the polymerase, alone or in combination, are likely required to achieve HBV eradication (11).

The HBV replication cycle

The mature, infectious HBV virion is a 42 nm particle comprised of a viral envelope with embedded HBsAg and contains the relaxed-circular DNA genome (rcDNA) that is covalently attached to the polymerase (P) and encapsulated by the assembled nucleocapsid core (12,13).

HBV belongs to the hepadnaviridae family of viruses, and as such HBV infects liver cells following HBsAg interactions with the sodium taurocholate cotransporting polypeptide (NTCP), a transmembrane protein specifically expressed in hepatocytes (14). The virus is internalized in an endosome, and the nucleocapsid is released into the cytoplasm by currently unresolved mechanisms (15). In infectious particles, the nucleocapsid is a T = 4 icosahedral core that is an assembly of the multimeric core protein (Cp), or HBV core antigen (HBcAg) (Figure 1). Once in the cytoplasm, the core is trafficked toward the nucleus to proceed through the nuclear pore complex (NPC) in an importin-dependent fashion (12,16). Following nuclear entry, the partially double-stranded and partially single-stranded rcDNA with the covalently attached P protein triggers host cell DNA repair mechanisms. This inevitably leads to the formation of the covalently closed chromosomal DNA (cccDNA), an episome indistinguishable to the host from its own genome, functionally integrating itself into the cell (17,18).

Figure 1.

Figure 1.

Structure of the hepatitis B Virus (HBV) capsid core. [Left] Rendition of released particles from HBV infection, including mature virions with assembled capsids (PDB: 6HTX) (96) and core protein (Cp) dimers (PDB: 6ECS) (97) as well as both non-infectious subviral particles and excreted HBV E Antigen (HBeAg, PDB: 3V6Z) (51). No full structure of HBV Surface Antigen (HBsAg) is deposited in the PDB; partial HBsAg (PDB: 7TUK) (98) and CD9 (PDB: 6K4J) (99) protein embedded in the default membrane structure from cellPAINT to represent the viral envelope (in grey). The mature capsid shown in blue and all other proteins in white. The P•pgRNA complex or P•rcDNA would be located inside the core. Made with cellPAINT (v2.0) (100). [Right] Two orientations of the HBV Capsid core (PDB: 6HTX) (96) with hexamers in cyan and pentamers in pink. One dimer of Cp dimers has the secondary structure shown with distinct colors in each Cp monomer to display protein·protein interactions. Made with ChimeraX (v1.5) (101).

The cccDNA genome is the template for multiple mRNA transcripts of varying size, one of which encodes the full pre-genomic RNA (pgRNA) that can be translated into HBcAg or HBeAg depending on the ORF used, and P (19-21). The HBV P protein is a large, multifunctional protein with Ribonuclease H (RNaseH), RT protein-priming, RNA-dependent DNA-polymerase (RdDP) and DNA-dependent DNA-polymerase (DdDP) activity (8,21). The interaction of P with pgRNA, specifically with the epsilon RNA stem loop, is required for encapsidation into assembling cores, which has fine-tuned assembly kinetics to make infectious particles (22). The pgRNA serves as the template for RT when creating the rcDNA after encapsidation (21,23). In addition to the infectious virions, referred to as "Dane particles", infected cells excrete non-infectious soluble core antigens (HBeAg) and excrete HBsAg-coated subviral particles (SVP). These SVPs can contain viral mRNA, erroneous cellular mRNA or be entirely devoid of nucleic acid (24). The mRNA translated from cccDNA can be alternatively spliced, leading to production of small/ medium and large HBsAg mRNA. The HBsAg precursor proteins are translated at the endoplasmic reticulum (ER) membrane, at different initiation codons, and adopt various conformations to enable the HBV envelope to form and bud (12,19).

The core protein (Cp) of HBV

The largest transcript from the cccDNA encodes the full HBV genome, the pgRNA. In addition to being packaged into assembling virions, as discussed above, the pgRNA can be used as the mRNA for multiple HBV proteins. pgRNA encodes for P, as well as two in-frame initiation codons that translate precore (p25) or Cp (p21) (12,19,21). When translated starting at the second in-frame initiation codon, Cp is cytosolically translated prior to its assembly into the HBV capsid core. Cp is a 21 kDa protein with 183-185 amino acids, depending on the virus isolate. The N-terminal Domain of Cp (CpNTD) is responsible for capsid assembly and the C-terminal domain of Cp (CpCTD) is responsible for binding nucleic acid and signaling (13,25-29) (Figure 2A).

Figure 2.

Figure 2.

Structure and Characteristics of the HBV core protein (Cp). (A). Sequence and structural motifs in p25; colors to the right (UniProt: P0C6H5). Arrows indicate start codon sites, with the second being used to translate Cp and the first for preCore (preC). The site of the Signal Peptide (SP; brown) cleavage shown with the yellow triangle. Residues are colored by charges at pH 7.0 (42). (B). Two Cp Dimers (PDB: 6HTX) (96), one in gray and the other colored based on the sequence annotation in (A). Made with ChimeraX (101). (C). Shows the local hydropathy of the Cp, with the CpNTD hydrophobic helices for assembling the core. (D). Displays the local charge of the Cp, exemplifying the CpCTD having a characteristic basic charge for nucleic acid binding. (E). Predictions of intrinsic disorder by IUPred2 (> 0.5 disordered region; < 0.5 ordered region) (102), exemplifying the disordered nature of the CpCTD. A, C-E made with idpr v 1.8.0 (42).

The CpNTD comprises residues 1-149 and, when expressed in E. coli, can assemble in vitro into particles indistinguishable from native HBV cores; however, the CpNTD could not assemble in mammalian cells or in vitro with rabbit reticulocyte lysate (29-33). This domain contains five α-helices (α1-α5), with α3 and α4 acting as the dimerization interface by making a 4-helix bundle (4HB) in Cp•Cp dimers, and α1, α2, and α5 sit perpendicular to the 4HB making the interface for dimer-dimer interactions (Figure 2B). Following translation, Cp monomers will immediately dimerize at the hairpins made by the amphipathic, antiparallel α3 and α4, forming a 4HB, which protrudes out of assembled cores like a spike (Figure 2C) (25,29,30,34-36). The 4HB is stabilized by a disulfide bond between the Cys61 in both monomers, but this linkage is not required for Cp assembly (21,37). Following dimerization, hydrophobic interactions between α5's and multiple contacts throughout Cp create dimer•dimer interactions to form the assembled capsid (36,38,39). The residues 140-149 are frequently referred to as a linker domain connecting CpNTD and CpCTD, and it has assembly-independent functions (40,41).

The CpCTD comprises residues 150-183 or 150-185 depending on subtype, and is characterized by intrinsic disorder, due in part to the presence of 16 positively charged Arg residues and 7 residues (6 Ser and 1 Thr) that can be phosphorylated in this 35-37 amino acid region (26-28,42) (Figure 2, C-E). These sequential Arg residues provide the nucleic acid binding functions of Cp that are needed for encapsulation of pgRNA during core assembly, as well as influencing rcDNA formation and Cp potentially being associated with nuclear cccDNA (17,23,27-29,41,43). The CpCTD additionally contains both nuclear localization signals (NLS) and nuclear export signals (NES) that are made of these sequential Arg residues (16). Further, the phosphorylation of CpCTD residues encode a complex regulatory system that influences many aspects of HBV replication, including cellular trafficking, disassembly at the nuclear pore complex and RNA encapsidation, which remains the target of ongoing investigations (27,31,32,44,45). For many years, the full-length Cp protein was challenging to recombinantly express and purify; truncated expression constructs made only of the CpNTD gave increased stability, and thus, many studies of Cp assembly have relied solely on the CpNTD. Now, bacterial codon-optimized expression vectors allow for the efficient production of full-length Cp in E. coli, especially helping in the translation of the Arg-rich CpCTD (28,45,46). While the CpCTD is not required for capsid lattice assembly, it is required for creating infectious HBV virions (47).

Translation of the preCore (preC) begins at the initiation codon upstream of the Cp initiation site and generates a precursor protein that contains a 29 residue N-terminal extension of the Cp, which contains a cotranslational ER localization signal (19,20). A segment of the preC N-terminus is processed inside the ER lumen, changing the protein from p25 to p22, and subsequent processing at the C-terminus into p17 prior to ER-Golgi- mediated secretion (Figure 2A; yellow triangle) (48,49). The p22 intermediate and Cp are nearly identical at the sequence level, except for the remaining 10 residue N-terminal extension in p22 (19,48,49). This short leader sequence contains an influential cysteine residue (Cys- 7), that forms a disulfide bond with Cys61, creating distinct quaternary structures that result in two HBV-related antigens: HBeAg and HBcAg for p17 and Cp, respectively (13,50,51). Of note, when denaturing Cp, the antigenic region of HBeAg is revealed (49,52). The function of HBeAg is not known and it is dispensable for infection; HBeAg is found in serum of patients shortly after infection and, as such, is used as a marker for active viral replication (53). Additionally, there is emerging interest in tracking the transcriptional activity of cccDNA using the hepatitis B virus core-related antigen (HBcrAg), a mixture of HBcAg, HBeAg, and other preC gene products (19).

Once the critical concentration of Cp has been surpassed, 90 or 120 Cp dimers will spontaneously self-assemble into T = 3 or T = 4 icosahedral capsids, respectively. In the assembled cores, the CpNTD faces outwards with the 4HB protrusions giving the capsid a spike-like appearance, and the CpCTD is generally localized to the interior and is highly flexible. Assembly is characterized by sigmoidal kinetics, similar to a crystal lattice with the rate-limiting step being the formation of a nucleation seed (54). The lag phase is characterized by the formation of a trimer of dimers; individual dimer-dimer interactions in vitro are weak (~3.5 kcal/ mol), which allows for the thermodynamic editing of improperly associated dimers. Once nuclei form, they rapidly assemble into icosahedral capsids, as free dimers remain. Cp dimers form at least 3 contacts with additive association energies that are enough to form stable capsids even though individual dimer•dimer interactions are weak (54-56). It is important to note that the in vivo assembly also can utilize RNA not possible in these CpNTD constructs; in purified capsid-like particles, those with the CpCTD and ssRNA require harsher denaturation conditions (45,47,57). T = 4 capsids are the primary result of assembly products (> 90%) and have a diameter of ~36 nm, while T = 3 capsids have a diameter ~32 nm (58). The ratio of these core sizes, as well as the kinetics of assembly, are impacted by ionic strength and protein concentration with mild conditions favoring T = 4 and few intermediates (28). In addition to the spiked icosohedreon, the capsid is characterized by many holes that are permeable to ions, metabolites and inhibitors (25,39,58).

Capsid assembly modulators (CAMs)

A class of molecules with curative potential are CAMs that bind Cp dimers. Also referred to as core protein allosteric modulators (CpAMs) and Capsid Inhibitors (CIs), these are an exciting development in the field of HBV therapeutics, as NUCs treat but do not cure CHB (59). These Cp-targeting HBV antivirals bind to the same site but are commonly subdivided into two classes: type I CAMs that create aggregated or aberrant capsids that are unable to function (CAM-A), and type II CAMs that enhance the assembly rates of Cp dimer•dimer interactions to make empty cores (CAM-E) (28,60,61). As the capsid has numerous roles throughout the replication cycle, its misassembly can impact multiple steps in acute or CHB infection.

Class I: CAM-A

The first CAM-A molecule described was BAY 41- 4109, belonging to the heteroaryldihydropyrimidines (HAP) family with HBV inhibition at submicromolar concentrations (33,62,63). Oral administration of BAY 41-4109 decreased HBV DNA in liver and plasma of HBV-transgenic mice, in line with 3TC treatment, but also showed a reduction of cytoplasmic HBcAg, a phenotype distinct from the NUC treatment (62). The observed Cp reduction is a result of proteosome activity (63), although an increase in Cp ubiquitylation has not been shown following HAP treatment (33). More recent studies have found nuclear aggregates of Cp, specifically associated with promyelocytic leukemia (PML) nuclear bodies, after multiple days of CAM-A (BAY 41-4109 and BAY 38-7690) but not CAM-E treatment (64,65). Shortly after BAY 41-4109 treatment, Cp aggregates were primarily cytoplasmic, but associated with nuclear envelope (66). Further, it was shown that after infection BAY 41-4109-induced Cp aggregates were perinuclear and targeted for p62-mediated macroautophagy and lysosomal degradation by the host factor STUB1, an E3 ubiquitin ligase (67). It should be noted that STUB1 is a co-chaperone of heat shock protein 70 (HSP70) and HSP90 (68), and both are considered proviral factors for HBV capsid assembly (67,69,70). Though the BAY compounds were not clinically developed, they have served as good probes to characterize the CAM-A chemotype.

Currently, the most promising HAP compounds are RO7049389 and GLS4. RO7049389 is currently in phase 2 trials (NCT04225715), with phase 1 results (NCT02952924) showing infrequent, mild adverse events and a reduction of HBV DNA in treatment-naïve CHB patients after 4 weeks of CAM-A administration comparable to NUC controls (71). Additionally, due to the high prevalence of HBV in Asia (3,6), the safety of RO7049389 was validated in healthy Chinese volunteers in a phase 1 clinical trial (NCT03570658). This showed the compound was well-tolerated in single (600 mg) and multiple (400 mg twice daily) doses, and the safety profiles were similar between Asian and non-Asian healthy volunteers, though with higher plasma exposure of RO7049389 observed in the Chinese participants (72). The other leading CAM-A molecule currently in phase 2 trials is GLS4 (NCT04147208), a derivative of Bay 39- 5493, with nanomolar potency and is fairly well-tolerated and effective when administered at 120 mg daily in combination with 100 mg ritonavir for 28 days (73,74). The safety and efficacy of these CAM-A compounds are exciting, but their long-term application and antiviral resistance remains to be seen.

Some other CAM-A molecules include KL060332 (75), a HAP molecule under a phase 1a trial in China, and preclinical compounds JNJ-890 (76), HAP_R01 (77), and ZW-1847 (78). A unique, non-HAP molecule with CAM-A phenotype is Ciclopirox, which inhibited HBV at 5 mg/kg in mice (79). Interestingly, ciclopirox olamine®, an antifungal agent, was tested for oral safety and tolerability as an anti-apoptotic gene suppressant for treating hematological malignancies (NCT00990587), indicating its safety for use in future trials investigating HBV inhibition in clinical trials (79,80).

Class II: CAM-E

Both CAM-A and CAM-E bind to the same hydrophobic pocket at Cp dimer•dimer interfaces, but the cores formed following only CAM-E treatment are noninfectious, vacant icosahedral capsids. CAM-E molecules enhance the rate dimer-dimer interactions, overcoming the rate-limiting step in assembly, and form icosahedral capsids that are devoid of P•pgRNA. Unlike the CAM-A class made primarily of the HAP chemotype that results in aggregates, CAM-E molecules are much more diverse and rapidly lead to native-like assemblies. Some CAM-E scaffolds include phenylpropenamides (PPAs), glyoxamoylpyrroloxamides (GLPs), sulfamoylbenzamides (SBAs), and sulfamoylpyrroloamides (SPAs).

One of first reported CAMs and the best studied CAM-E molecule is AT-130, which belongs to the PPA class (61,81). AT-130 inhibits HBV with submicromolar potency in cell culture but has not been tried in clinical trials. As the prototypical scaffold for the CAM-E chemotype, AT-130 has been shown to increase the kinetics of capsid assembly through interactions near CpNTD linker and α5 that alter the interface of dimer•dimer interactions, as well as distorting the 4HB, resulting in intermediates that are primed to assemble (82-84). These result in icosahedral capsids without the P•pgRNA needed for HBV replication, or partially completed assembly intermediates (84,85).

Another preclinical CAM-E class that has great potential is the GLP class, with the first GLP compound patented in 2015 (61,86). GLP compounds are exceptionally potent, with the compound GLP-26 having nanomolar potency in humanized mice (87). GLP-26 was further shown to be well tolerated and highly effective at inhibiting HBV in both non-human primate studies and primary human cardiomyocytes, indicating safety for further investigations (88).

Both the SBA and SPA classes are some of the most clinically developed CAMs. One of the first successful clinical investigations into CAM safety was the SBA compound NVR 3-778 (NCT02401737), which was tolerated but not as effective as approved NUCs and thus was discontinued (28,89). An SBA under clinical investigation is JNJ-64530440 (phase 1b, NCT03439488) with potent antiviral activity in patients with CHB and was well tolerated (90). Additionally, the SPA JNJ-6379 (JADE NCT03361956), recently reported phase 2 trials results of a reduction in HBV DNA and RNA when given in combination with a NUC. They note that multiple patients had the T33N mutation emerge during monotherapy (91). This underscores the likely importance of combination therapy in future HBV therapeutics.

Some other promising CAM-Es molecules include ALG-000184, a potent inhibitor with successful phase 1 trials (NCT04536337) (92), Canocapavir (ZM-H1505R) that was well tolerated (Phase 1b, NCT05470829) (93), and GST-HG141 (phase 1, NCT04386915 & NCT04868981) (94). Many CAMs have been reported and are under investigation in clinical trials without their exact CAM phenotype described (Recently reviewed (28,55)). Additionally, a novel HBV antiviral class has been reported as a cccDNA inhibitor; the molecule ccc_ R08 is hypothesized to target an unknown host factor and decreased preexisting cccDNA pools, unlike currently reported CAMs (95).

Conclusions

At the present, approximately 3.5% of the world's population lives with CHB, though most are unaware (3,6). Key to curing CHB is elimination of cccDNA, and the currently approved and widely used NUCs suppress HBV replication but do not achieve a cure (18,55). Due to their ability to clear cccDNA, CAMs that modify Cp dimer•dimer interactions have the potential to redefine the standard of care for treating HBV infection and could subsequently achieve a cure for CHB (11,59). The long-term effects of CAM treatment and likely emergence of CAM-induced antiviral resistance remain to be investigated. However, the progress in CAM development and successful investigations into HBV capsid biology over the last two and a half decades give promise for highly effective and well characterized treatments to potentially cure the hundreds of millions currently living with CHB.

Funding

This research was supported in part by the National Institutes of Health (R01 AI132833 and R01 AI121315 to SGS; WMM was supported in part by T32 GM135060 and F31 AI174951). SGS acknowledges funding from the Nahmias-Schinazi Distinguished Chair in Research.

Conflict of Interest

The authors have no conflicts of interest to disclose.

References

  • 1. Blumberg B, Sutnick A, London WT. Hepatitis and leukemia: their relation to Australia antigen. Bull N Y Acad Med. 1968; 44:1566-1586. [PMC free article] [PubMed] [Google Scholar]
  • 2. Suk-Fong Lok A. Hepatitis B: 50 years after the discovery of Australia antigen. J Viral Hepat. 2016; 23:5-14. [DOI] [PubMed] [Google Scholar]
  • 3. World Health Organization. Hepatitis B: WHO; 2022. https://www.who.int/news-room/fact-sheets/detail/hepatitis-b (accessed May 15, 2023).
  • 4. Indolfi G, Easterbrook P, Dusheiko G, Siberry G, Chang MH, Thorne C, Bulterys M, Chan PL, El-Sayed MH, Giaquinto C, Jonas MM, Meyers T, Walsh N, Wirth S, Penazzato M. Hepatitis B virus infection in children and adolescents. Lancet Gastroenterol Hepatol. 2019; 4:466-476. [DOI] [PubMed] [Google Scholar]
  • 5. Razavi-Shearer D, Blach S, Gamkrelidze I, Estes C, Mooneyhan E, Razavi-Shearer K, Razavi H. The disease burden of hepatitis B and hepatitis C from 2015 to 2030: the long and winding road. Journal of Hepatology. 2022; 77:S43. [Google Scholar]
  • 6. Hsu YC, Huang DQ, Nguyen MH. Global burden of hepatitis B virus: current status, missed opportunities and a call for action. Nat Rev Gastroenterol Hepatol. 2023. doi: 10.1038/s41575-023-00760-9. [DOI] [PubMed] [Google Scholar]
  • 7. Cooke GS, Andrieux-Meyer I, Applegate TL, et al. Accelerating the elimination of viral hepatitis. Lancet Gastroenterol Hepatol. 2019; 4:135-184. [DOI] [PubMed] [Google Scholar]
  • 8. Michailidis E, Kirby KA, Hachiya A, Yoo W, Hong SP, Kim SO, Folk WR, Sarafianos SG. Antiviral therapies: focus on hepatitis B reverse transcriptase. Int J Biochem Cell Biol. 2012; 44:1060-1071. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9. Pockros PJ, Mulgaonkar A. Cessation of nucleoside/ nucleotide analogue therapy in chronic hepatitis B HBeAg-negative patients. Gastroenterol Hepatol (N Y). 2022; 18:320-325. [PMC free article] [PubMed] [Google Scholar]
  • 10. Ye J, Chen J. Interferon and hepatitis B: current and future perspectives. Front Immunol. 2021; 12:733364. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11. Boucle S, Bassit L, Ehteshami M, Schinazi RF. Toward elimination of hepatitis B virus using novel drugs, approaches, and combined modalities. Clin Liver Dis. 2016; 20:737-749. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12. Tsukuda S, Watashi K. Hepatitis B virus biology and life cycle. Antiviral Res. 2020; 182:104925. [DOI] [PubMed] [Google Scholar]
  • 13. Selzer L, Zlotnick A. Assembly and release of hepatitis B virus. Cold Spring Harb Perspect Med. 2015; 5:a021394. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14. Yan H, Zhong G, Xu G, et al. Sodium taurocholate cotransporting polypeptide is a functional receptor for human hepatitis B and D virus. Elife. 2012; 1:e00049. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15. Herrscher C, Roingeard P, Blanchard E. Hepatitis B virus entry into cells. Cells. 2020; 9:1486. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16. Li HC, Huang EY, Su PY, Wu SY, Yang CC, Lin YS, Chang WC, Shih C. Nuclear Export and import of human hepatitis B virus capsid protein and particles. PLoS Pathog. 2010; 6:e1001162. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17. Bock CT, Schwinn S, Locarnini S, Fyfe J, Manns MP, Trautwein C, Zentgraf H. Structural organization of the hepatitis B virus minichromosome. J Mol Biol. 2001; 307:183-196. [DOI] [PubMed] [Google Scholar]
  • 18. Wei L, Ploss A. Mechanism of hepatitis B virus cccDNA formation. Viruses. 2021; 13:1463. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19. Hong X, Luckenbaugh L, Mendenhall M, Walsh R, Cabuang L, Soppe S, Revill PA, Burdette D, Feierbach B, Delaney W, Hu J. Characterization of hepatitis B precore/ core-related antigens. J Virol. 2021; 95:e01695-e01720. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20. Standring DN, Ou JH, Masiarz FR, Rutter WJ. A signal peptide encoded within the precore region of hepatitis B virus directs the secretion of a heterogeneous population of e antigens in Xenopus oocytes. Proc Natl Acad Sci U S A. 1988; 85:8405-8409. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21. Clark DN, Tajwar R, Hu J, Tavis JE. The hepatitis B virus polymerase. Enzymes. 2021; 50:195-226. [DOI] [PubMed] [Google Scholar]
  • 22. Bartenschlager R, Schaller H. Hepadnaviral assembly is initiated by polymerase binding to the encapsidation signal in the viral RNA genome. EMBO J. 1992; 11:3413-3420. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23. Nassal M. The arginine-rich domain of the hepatitis B virus core protein is required for pregenome encapsidation and productive viral positive-strand DNA synthesis but not for virus assembly. J Virol. 1992; 66:4107-4116. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24. Chai N, Chang HE, Nicolas E, Han Z, Jarnik M, Taylor J. Properties of subviral particles of hepatitis B virus. J Virol. 2008; 82:7812-7817. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25. Wynne SA, Crowther RA, Leslie AG. The crystal structure of the human hepatitis B virus capsid. Mol Cell. 1999; 3:771-780. [DOI] [PubMed] [Google Scholar]
  • 26. Chain BM, Myers R. Variability and conservation in hepatitis B virus core protein. BMC Microbiology. 2005; 5:33. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27. de Rocquigny H, Rat V, Pastor F, Darlix JL, Hourioux C, Roingeard P. Phosphorylation of the arginine-rich C-terminal domains of the hepatitis B virus (HBV) core protein as a fine regulator of the interaction between HBc and nucleic acid. Viruses. 2020; 12:738. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28. Niklasch M, Zimmermann P, Nassal M. The hepatitis B virus nucleocapsid-dynamic compartment for infectious virus production and new antiviral target. Biomedicines. 2021; 9:1577. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29. Birnbaum F, Nassal M. Hepatitis B virus nucleocapsid assembly: primary structure requirements in the core protein. J Virol. 1990; 64:3319-3330. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30. Wingfield PT, Stahl SJ, Williams RW, Steven AC. Hepatitis core antigen produced in escherichia coli: subunit composition, conformation analysis, and in vitro capsid assembly. Biochemistry. 1995; 34:4919-4932. [DOI] [PubMed] [Google Scholar]
  • 31. Ludgate L, Liu K, Luckenbaugh L, Streck N, Eng S, Voitenleitner C, Delaney WE 4th, Hu J. Cell-free hepatitis B virus capsid assembly dependent on the core protein C-terminal domain and regulated by phosphorylation. J Virol. 2016; 90:5830-5844. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32. Rat V, Pinson X, Seigneuret F, Durand S, Herrscher C, Lemoine R, Burlaud-Gaillard J, Raynal PY, Hourioux C, Roingeard P, Tramier M, de Rocquigny H. Hepatitis B virus core protein domains essential for viral capsid assembly in a cellular context. J Mol Biol. 2020; 432:3802-3819. [DOI] [PubMed] [Google Scholar]
  • 33. Kim H, Ko C, Lee JY, Kim M. Current progress in the development of hepatitis B virus capsid assembly modulators: chemical structure, mode-of-action and efficacy. Molecules. 2021; 26:7420. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34. Conway JF, Cheng N, Zlotnick A, Wingfield PT, Stahl SJ, Steven AC. Visualization of a 4-helix bundle in the hepatitis B virus capsid by cryo-electron microscopy. Nature. 1997; 386:91-94. [DOI] [PubMed] [Google Scholar]
  • 35. Böttcher B, Wynne SA, Crowther RA. Determination of the fold of the core protein of hepatitis B virus by electron cryomicroscopy. Nature. 1997; 386:88-91. [DOI] [PubMed] [Google Scholar]
  • 36. Zheng CL, Fu YM, Xu ZX, Zou Y, Deng K. Hepatitis B virus core protein dimer*dimer interface is critical for viral replication. Mol Med Rep. 2019; 19:262-270. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37. Bourne CR, Finn MG, Zlotnick A. Global structural changes in hepatitis B virus capsids induced by the assembly effector HAP1. J Virol. 2006; 80:11055-11061. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38. Zhao Z, Wang JC, Segura CP, Hadden-Perilla JA, Zlotnick A. The integrity of the intradimer interface of the hepatitis B virus capsid protein dimer regulates capsid self-assembly. ACS Chem Biol. 2020; 15:3124-3132. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39. Hadden JA, Perilla JR, Schlicksup CJ, Venkatakrishnan B, Zlotnick A, Schulten K. All-atom molecular dynamics of the HBV capsid reveals insights into biological function and cryo-EM resolution limits. Elife. 2018; 7:e32478. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40. Watts NR, Conway JF, Cheng N, Stahl SJ, Belnap DM, Steven AC, Wingfield PT. The morphogenic linker peptide of HBV capsid protein forms a mobile array on the interior surface. EMBO J. 2002; 21:876-884. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41. Xi J, Luckenbaugh L, Hu J. Multiple roles of PP2A binding motif in hepatitis B virus core linker and PP2A in regulating core phosphorylation state and viral replication. PLoS Pathog. 2021; 17:e1009230. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42. McFadden WM, Yanowitz JL. idpr: a package for profiling and analyzing intrinsically disordered proteins in R. PLoS One. 2022; 17:e0266929. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43. Zlotnick A, Cheng N, Stahl SJ, Conway JF, Steven AC, Wingfield PT. Localization of the C terminus of the assembly domain of hepatitis B virus capsid protein: implications for morphogenesis and organization of encapsidated RNA. Proc Natl Acad Sci U S A. 1997; 94:9556-9561. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44. Luo J, Xi J, Gao L, Hu J. Role of hepatitis B virus capsid phosphorylation in nucleocapsid disassembly and covalently closed circular DNA formation. PLoS Pathog. 2020; 16:e1008459. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45. Heger-Stevic J, Zimmermann P, Lecoq L, Böttcher B, Nassal M. Hepatitis B virus core protein phosphorylation: Identification of the SRPK1 target sites and impact of their occupancy on RNA binding and capsid structure. PLoS Pathog. 2018; 14:e1007488. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46. Heger-Stevic J, Kolb P, Walker A, Nassal M. Displaying whole-chain proteins on hepatitis B virus capsid-like particles. Methods Mol Biol. 2018; 1776:503-531. [DOI] [PubMed] [Google Scholar]
  • 47. Chua PK, Tang FM, Huang JY, Suen CS, Shih C. Testing the balanced electrostatic interaction hypothesis of hepatitis B virus DNA synthesis by using an in vivo charge rebalance approach. J Virol. 2010; 84:2340-2351. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48. Messageot F, Salhi S, Eon P, Rossignol JM. Proteolytic processing of the hepatitis B virus e antigen precursor. Cleavage at two furin consensus sequences. J Biol Chem. 2003; 278:891-895 [DOI] [PubMed] [Google Scholar]
  • 49. Ito K, Kim KH, Lok AS, Tong S. Characterization of genotype-specific carboxyl-terminal cleavage sites of hepatitis B virus e antigen precursor and identification of furin as the candidate enzyme. J Virol. 2009; 83:3507-3517. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50. Nassal M, Rieger A. An intramolecular disulfide bridge between Cys-7 and Cys61 determines the structure of the secretory core gene product (e antigen) of hepatitis B virus. J Virol. 1993; 67:4307-4315. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51. DiMattia MA, Watts NR, Stahl SJ, Grimes JM, Steven AC, Stuart DI, Wingfield PT. Antigenic switching of hepatitis B virus by alternative dimerization of the capsid protein. Structure. 2013; 21:133-142. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52. Takahashi K, Akahane Y, Gotanda T, Mishiro T, Imai M, Miyakawa Y, Mayumi M. Demonstration of hepatitis B e antigen in the core of Dane particles. J Immunol. 1979; 122:275-279. [PubMed] [Google Scholar]
  • 53. Guvenir M, Arikan A. Hepatitis B virus: from diagnosis to treatment. Pol J Microbiol. 2020; 69:391-399. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54. Zlotnick A. Theoretical aspects of virus capsid assembly. J Mol Recognit. 2005; 18:479-490. [DOI] [PubMed] [Google Scholar]
  • 55. Viswanathan U, Mani N, Hu Z, Ban H, Du Y, Hu J, Chang J, Guo JT. Targeting the multifunctional HBV core protein as a potential cure for chronic hepatitis B. Antiviral Res. 2020; 182:104917. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56. Ceres P, Zlotnick A. Weak protein-protein interactions are sufficient to drive assembly of hepatitis B virus capsids. Biochemistry. 2002; 41:11525-11531. [DOI] [PubMed] [Google Scholar]
  • 57. Porterfield JZ, Dhason MS, Loeb DD, Nassal M, Stray SJ, Zlotnick A. Full-length hepatitis B virus core protein packages viral and heterologous RNA with similarly high levels of cooperativity. J Virol. 2010; 84:7174-7184. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58. Crowther RA, Kiselev NA, Böttcher B, Berriman JA, Borisova GP, Ose V, Pumpens P. Three-dimensional structure of hepatitis B virus core particles determined by electron cryomicroscopy. Cell. 1994; 77:943-950. [DOI] [PubMed] [Google Scholar]
  • 59. Lam AM, Ren S, Espiritu C, Kelly M, Lau V, Zheng L, Hartman GD, Flores OA, Klumpp K. Hepatitis B virus capsid assembly modulators, but not nucleoside analogs, inhibit the production of extracellular pregenomic RNA and spliced RNA variants. Antimicrob Agents Chemother. 2017; 61:e00680-e00717. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60. McFadden WM, Sarafianos SG. Targeting the HIV-1 and HBV capsids, an encore. Viruses. 2023; 15:896. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61. Nijampatnam B, Liotta DC. Recent advances in the development of HBV capsid assembly modulators. Curr Opin Chem Biol. 2019; 50:73-79. [DOI] [PubMed] [Google Scholar]
  • 62. Weber O, Schlemmer KH, Hartmann E, Hagelschuer I, Paessens A, Graef E, Deres K, Goldmann S, Niewoehner U, Stoltefuss J, Haebich D, Ruebsamen-Waigmann H, Wohlfeil S. Inhibition of human hepatitis B virus (HBV) by a novel non-nucleosidic compound in a transgenic mouse model. Antiviral Res. 2002; 54:69-78. [DOI] [PubMed] [Google Scholar]
  • 63. Deres K, Schröder CH, Paessens A, et al. Inhibition of hepatitis B virus replication by drug-induced depletion of nucleocapsids. Science. 2003; 299:893-896. [DOI] [PubMed] [Google Scholar]
  • 64. Huber AD, Wolf JJ, Liu D, Gres AT, Tang J, Boschert KN, Puray-Chavez MN, Pineda DL, Laughlin TG, Coonrod EM, Yang Q, Ji J, Kirby KA, Wang Z, Sarafianos SG. The heteroaryldihydropyrimidine bay 38-7690 induces hepatitis B virus core protein aggregates associated with promyelocytic leukemia nuclear bodies in infected cells. mSphere. 2018; 3:e00131-e00218. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65. Corcuera A, Stolle K, Hillmer S, Seitz S, Lee JY, Bartenschlager R, Birkmann A, Urban A. Novel non-heteroarylpyrimidine (HAP) capsid assembly modifiers have a different mode of action from HAPs in vitro. Antiviral Res. 2018; 158:135-142. [DOI] [PubMed] [Google Scholar]
  • 66. Rat V, Seigneuret F, Burlaud-Gaillard J, Lemoine R, Hourioux C, Zoulim F, Testoni B, Meunier JC, Tauber C, Roingeard P, de Rocquigny H. BAY 41-4109-mediated aggregation of assembled and misassembled HBV capsids in cells revealed by electron microscopy. Antiviral Res. 2019; 169:104557. [DOI] [PubMed] [Google Scholar]
  • 67. Lin J, Yin L, Xu XZ, Sun HC, Huang ZH, Ni XY, Chen Y, Lin X. Bay41-4109-induced aberrant polymers of hepatitis b capsid proteins are removed via STUB1- promoted p62-mediated macroautophagy. PLoS Pathog. 2022; 18:e1010204. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68. Kundrat L, Regan L. Balance between folding and degradation for Hsp90-dependent client proteins: a key role for CHIP. Biochemistry. 2010; 49:7428-7438. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69. Lim SO, Park SG, Yoo JH, Park YM, Kim HJ, Jang KT, Cho JW, Yoo BC, Jung GH, Park CK. Expression of heat shock proteins (HSP27, HSP60, HSP70, HSP90, GRP78, GRP94) in hepatitis B virus-related hepatocellular carcinomas and dysplastic nodules. World J Gastroenterol. 2005; 11:2072-2079. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70. Seo HW, Seo JP, Jung G. Heat shock protein 70 and heat shock protein 90 synergistically increase hepatitis B viral capsid assembly. Biochem Biophys Res Commun. 2018; 503:2892-2898. [DOI] [PubMed] [Google Scholar]
  • 71. Yuen MF, Zhou X, Gane E, Schwabe C, Tanwandee T, Feng S, Jin Y, Triyatni M, Lemenuel-Diot A, Cosson V, Xue Z, Kazma R, Bo Q. 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. [DOI] [PubMed] [Google Scholar]
  • 72. Wu X, Feng S, Zhang J, Zhang W, Zhang Y, Zhu M, Triyatni M, Zhao N, Bo Q, Jin Y. Evaluation of the safety, tolerability, and pharmacokinetics of RO7049389 in healthy Chinese volunteers. Clin Transl Sci. 2022; 15:195-203. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73. Zhang H, Wang F, Zhu X, Chen Y, Chen H, Li X, Wu M, Li C, Liu J, Zhang Y, Ding Y, Niu J. Antiviral activity and pharmacokinetics of the hepatitis B virus (HBV) capsid assembly modulator GLS4 in patients with chronic HBV infection. Clin Infect Dis. 2021; 73:175-182. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74. Ren Q, Liu X, Luo Z, Li J, Wang C, Goldmann S, Zhang J, Zhang Y. Discovery of hepatitis B virus capsid assembly inhibitors leading to a heteroaryldihydropyrimidine based clinical candidate (GLS4). Bioorg Med Chem. 2017; 25:1042-1056. [DOI] [PubMed] [Google Scholar]
  • 75. Tai Z, Tian Q, Zhao X, Xie J, Lu Y, Tan Y, Zhao W, Ma X, Yuan X, Song H, Xue T, Wang J. Discovery of KL060332, a potential best-in-class HBV capsid inhibitor. https://aasld.confex.com/aasld/2020/meetingapp.cgi/Paper/19680 (accessed May 15, 2023). [Google Scholar]
  • 76. Lahlali T, Berke JM, Vergauwen K, Foca A, Vandyck K, Pauwels F, Zoulim F, Durantel D. Novel potent capsid assembly modulators regulate multiple steps of the hepatitis B virus life cycle. Antimicrob Agents Chemother. 2018; 62:e00835-e00918. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77. Qiu Z, Lin X, Zhang W, et al. Discovery and pre-clinical characterization of third-generation 4-H heteroaryldihydropyrimidine (HAP) analogues as hepatitis B virus (HBV) capsid inhibitors. J Med Chem. 2017; 60:3352-3371. [DOI] [PubMed] [Google Scholar]
  • 78. Senaweera S, Du H, Zhang H, Kirby KA, Tedbury PR, Xie J, Sarafianos SG, Wang Z. Discovery of new small molecule hits as hepatitis B virus capsid assembly modulators: structure and pharmacophore-based approaches. Viruses. 2021; 13:770. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79. Kang JA, Kim S, Park M, Park HJ, Kim JH, Park S, Hwang JR, Kim YC, Jun Kim Y, Cho Y, Sun Jin M, Park SG. Ciclopirox inhibits hepatitis B virus secretion by blocking capsid assembly. Nat Commun. 2019; 10:2184. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80. Minden MD, Hogge DE, Weir SJ, et al. Oral ciclopirox olamine displays biological activity in a phase I study in patients with advanced hematologic malignancies. Am J Hematol. 2014; 89:363-368. [DOI] [PubMed] [Google Scholar]
  • 81. Delaney WE, Edwards R, Colledge D, Shaw T, Furman P, Painter G, Locarnini S. Phenylpropenamide derivatives AT-61 and AT-130 inhibit replication of wild-type and lamivudine-resistant strains of hepatitis B virus in vitro. Antimicrob Agents Chemother. 2002; 46:3057-3060. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82. Pérez-Segura C, Goh BC, Hadden-Perilla JA. All-atom MD simulations of the HBV capsid complexed with AT130 reveal secondary and tertiary structural changes and mechanisms of allostery. Viruses. 2021; 13:564. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83. Katen SP, Tan Z, Chirapu SR, Finn MG, Zlotnick A. Assembly-directed antivirals differentially bind quasiequivalent pockets to modify hepatitis B virus capsid tertiary and quaternary structure. Structure. 2013; 21:1406-1416. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84. Kondylis P, Schlicksup CJ, Katen SP, Lee LS, Zlotnick A, Jacobson SC. Evolution of intermediates during capsid assembly of hepatitis B virus with phenylpropenamide-based antivirals. ACS Infect Dis. 2019; 5:769-777. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85. Feld JJ, Colledge D, Sozzi V, Edwards R, Littlejohn M, Locarnini SA. The phenylpropenamide derivative AT- 130 blocks HBV replication at the level of viral RNA packaging. Antiviral Res. 2007; 76:168-177. [DOI] [PubMed] [Google Scholar]
  • 86. Vandyck K, Kesteleyn BRR, Pieters SMA, Rombouts G, Verschueren WG, Raboisson PJ-MB. Glyoxamide substituted pyrrolamide derivatives and the use thereof as medicaments for the treatment of hepatitis B.U.S. Patent 10,450,270[P]. 2019-10-22. [Google Scholar]
  • 87. Amblard F, Boucle S, Bassit L, et al. Novel hepatitis B virus capsid assembly modulator induces potent antiviral responses in vitro and in humanized mice. Antimicrob Agents Chemother. 2020; 64:e01701-e01719. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88. Hurwitz SJ, McBrearty N, Arzumanyan A, Bichenkov E, Tao S, Bassit L, Chen Z, Kohler JJ, Amblard F, Feitelson MA, Schinazi RF. Studies on the efficacy, potential cardiotoxicity and monkey pharmacokinetics of GLP-26 as a potent hepatitis B virus capsid assembly modulator. Viruses. 2021; 13:114. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89. Gane EJ, Schwabe C, Walker K, Flores L, Hartman GD, Klumpp K, Liaw S, Brown N. Phase 1a safety and pharmacokinetics of NVR 3-778, a potential first-in-class HBV core inhibitor: LB-19. Hepatology. 2014; 60:1279A. [Google Scholar]
  • 90. Gane EJ, Schwabe C, Berliba E, Tangkijvanich P, Jucov A, Ghicavii N, Verbinnen T, Lenz O, Talloen W, Kakuda TN, Westland C, Patel M, Yogaratnam JZ, Dragone L, Van Remoortere P. Safety, antiviral activity and pharmacokinetics of JNJ-64530440, a novel capsid assembly modulator, as 4 week monotherapy in treatment-naive patients with chronic hepatitis B virus infection. J Antimicrob Chemother. 2022; 77:1102-1110. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91. Janssen HLA, Hou J, Asselah T, et al. Randomised phase 2 study (JADE) of the HBV capsid assembly modulator JNJ-56136379 with or without a nucleos(t)ide analogue in patients with chronic hepatitis B infection. Gut. 2023; 72:1385-1398. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92. Yuen RMF, Agarwal K, Gane EJ, et al. Safety, pharmacokinetics, and antiviral activity of the class II capsid assembly modulator ALG-000184 in subjects with chronic hepatitis B. Poster Presentation (SAT835) The International Liver Congress 2022, 22-26 June 2022. Journal of Hepatology, v77 nS1, pS835-836 [Google Scholar]
  • 93. Jia H, Mai J, Wu M, et al. Safety, tolerability, pharmacokinetics, and antiviral activity of the novel core protein allosteric modulator ZM-H1505R (Canocapavir) in chronic hepatitis B patients: a randomized multiple-dose escalation trial. BMC Medicine. 2023; 21:98. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94. Li C, Wu M, Zhang H, Mai J, Yang L, Ding Y, Niu J, Mao J, Wu W, Zhang D, Tang Y, Yan W. Safety, tolerability, and pharmacokinetics of the novel hepatitis B virus capsid assembly modulator GST-HG141 in healthy Chinese subjects: a first-in-human single- and multiple-dose escalation trial. Antimicrob Agents Chemother. 2021; 65:e01220-e01221. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95. Wang L, Zhu Q, Zhang JD, et al. Discovery of a first-in-class orally available HBV cccDNA inhibitor. J Hepatol. 2023; 78:742-753. [DOI] [PubMed] [Google Scholar]
  • 96. Böttcher B, Nassal M. Structure of mutant hepatitis B core protein capsids with premature secretion phenotype. J Mol Biol. 2018; 430:4941-4954. [DOI] [PubMed] [Google Scholar]
  • 97. Zhao Z, Wang JC, Gonzalez-Gutierrez G, Venkatakrishnan B, Asor R, Khaykelson D, Raviv U, Zlotnick A. Structural differences between the woodchuck hepatitis virus core protein in the dimer and capsid states are consistent with entropic and conformational regulation of assembly. J Virol. 2019; 93:e00141-19. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98. Liu H, Hong X, Xi J, Menne S, Hu J, Wang JC. Cryo-EM structures of human hepatitis B and woodchuck hepatitis virus small spherical subviral particles. Sci Adv. 2022; 8:eabo4184. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99. Umeda R, Satouh Y, Takemoto M, Nakada-Nakura Y, Liu K, Yokoyama T, Shirouzu M, Iwata S, Nomura N, Sato K, Ikawa M, Nishizawa T, Nureki O. Structural insights into tetraspanin CD9 function. Nat Commun. 2020; 11:1606. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100. Gardner A, Autin L, Barbaro B, Olson AJ, Goodsell DS. CellPAINT: Interactive Illustration of Dynamic Mesoscale Cellular Environments. IEEE Comput Graph Appl. 2018; 38:51-66. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101. Pettersen EF, Goddard TD, Huang CC, Meng EC, Couch GS, Croll TI, Morris JH, Ferrin TE. UCSF chimeraX: structure visualization for researchers, educators, and developers. Protein Science. 2021; 30:70-82. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102. Mészáros B, Erdős G, Dosztányi Z. IUPred2A: context-dependent prediction of protein disorder as a function of redox state and protein binding. Nucleic Acids Res. 2018; 46:W329-W337. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Global Health & Medicine are provided here courtesy of National Center for Global Health and Medicine, Japan

RESOURCES