Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2022 Aug 1.
Published in final edited form as: Curr Opin Virol. 2021 May 13;49:13–20. doi: 10.1016/j.coviro.2021.04.003

Innate immunity and HBV persistence

Carolina Chiale 1,#, Anthony M Marchese 1, Michael D Robek 1,*
PMCID: PMC8319108  NIHMSID: NIHMS1694248  PMID: 33992859

Abstract

Hepatitis B virus (HBV) causes chronic infections that are associated with immune dysfunction. Though T cell impairment is perhaps the most prominent immune change contributing to viral persistence, HBV interaction with the innate immune system is also likely key, as the lack of effective innate immunity has functional consequences that promote chronic infection. In addition to an intrinsic ability to fight viral infections, the innate immune system also impacts T cell responses and other adaptive immune mechanisms critical for HBV control. Therefore, it is essential to understand the relationships between HBV and innate immunity, as these interactions may be useful immunotherapeutic targets to manage the infection.

Keywords: Interferon, NK cell, dendritic cell, MDSC, macrophage, MAIT cell, Toll-like receptor

Introduction

The interactions between HBV and the immune system are important for the virus’ ability to persist, but the specific host and viral factors that impact whether an acute HBV infection is eliminated or progresses to chronic disease are poorly understood. Innate immunity plays crucial roles in the early response to viral infection and promotes adaptive immunity. Most viruses, including HBV, have evolved strategies to evade this response; however, the relationships between HBV and the cytokine and cellular components of innate immunity remain incompletely characterized. The pathways that drive innate immune activation represent promising therapeutic targets to overcome immune dysfunction and promote HBV cure. Here, we review the interactions between HBV and the interferon (IFN)- and cell-mediated mechanisms of innate immunity and discuss the therapeutic approaches that seek to engineer an effective innate immune response to the virus.

Interferon activation

HBV is often referred to as a “stealth” virus as it establishes infection without being detected by the innate immune system and thus avoids the activation of antiviral pathways [1]. Unlike other viruses that actively block innate responses, the lack of IFN induction by HBV in hepatocytes is thought to be a passive process [2-4]. This characteristic of HBV is debated, and multiple studies have aimed to investigate whether the virus stimulates innate immunity. In vivo analysis identified a lack of early antiviral gene expression in the chimpanzee liver after experimental HBV infection [5], and recent work using the woodchuck hepatitis virus (WHV) model of hepadnavirus infection also yielded findings consistent with the idea of HBV stealth [6]. Several mechanistic in vitro cell culture studies demonstrated that HBV passively avoids IFN activation [2,3], and recent findings further supporting this concept were made utilizing a novel scalable hepatic coculture system [7], which found no significant activation of the innate immune response by HBV [8]. Despite this evidence, some studies suggest that HBV stimulates or suppresses innate immunity [9-11], including recent work showing that activation of glycolysis by HBV inhibits RIG-I signaling [12]. Another recent study found that HBV infection induces TLR2-mediated expression of IL-6, IL-1β, and TNF-α in primary human hepatocytes without activating IFN or IFN-stimulated genes (ISG) [13]. Although these results seem to challenge the stealth virus concept, the high multiplicities of infection often required to reach this activation in vitro may not recapitulate a natural infection. Amongst the various experimental approaches used to evaluate HBV innate immune activation, the most physiologically relevant evidence to support HBV stealth perhaps comes from experiments performed using ex vivo human liver biopsy samples from infected patients [4]. These results showed a lack of ISG expression in the biopsies and no block of the IFN response, as hepatocytes from HBV-infected patients had a similar capacity to produce IFN as those from uninfected patients when exogenously stimulated with polyIC or Sendai virus [4].

Interferon antiviral activity

Although HBV appears to avoid activating innate immunity, IFN can nevertheless reduce viral replication [14]. There are three main types of IFN (α/β, γ, and λ) that signal through unique receptors with cell-type dependent expression patterns, and non-cytopathically inhibit HBV by downregulating HBV mRNA expression and viral pre-genomic RNA encapsidation [15-17].

IFN-α

IFN-α is the only immunomodulator currently approved for HBV therapy. In some cases, patients display prolonged viral suppression and HBsAg seroconversion following pegylated (PEG)-IFN-α treatment, but the reasons for why only certain patients respond to therapy remain unknown. While the precise therapeutic actions are unclear, recent studies have revealed mechanisms of ISG-mediated HBV suppression [18,19]. Interestingly, a recent study also found that activation of IFN-α signaling in the liver of HBV transgenic mice promotes the functional differentiation of intrahepatically primed CD8+ T cells [20]. After PEG-IFN-α treatment in humans, patients exhibit few changes in CD8+ T cells but increased proliferation and activation of CD56bright NK cells, which would suggest a role for these cells in HBV control [21]. However, additional studies characterizing treatment-induced immunological changes are needed to better understand IFN-α-mediated HBV inhibition mechanisms.

IFN-γ

Cytotoxic CD8+ T cells restrict HBV by killing infected hepatocytes and suppressing viral replication via cytokine-mediated noncytopathic mechanisms. Analysis of chimpanzees and transgenic mice demonstrated that IFN-γ produced by CD8+ T cells activates ISGs in the liver and suppresses HBV replication [22,23]. Despite its crucial role in HBV control, clinical trials that tested therapeutic IFN-γ intervention failed to demonstrate efficacy against chronic HBV infection [24]. Although not effective as a standalone therapy, IFN-γ remains an important marker of T cell functionality for other HBV immunotherapies due to its essential role in viral clearance.

IFN-λ

Unlike the more universally expressed IFN-α/β receptor, the IFN-λs utilize a unique receptor that is restricted to barrier surfaces, including epithelial cells of the intestinal, respiratory and reproductive tracts, and hepatocytes [25]. Notwithstanding differential receptor expression and signaling kinetics, the ISG expression profile of IFN-λ-driven antiviral responses is similar to that activated by IFN-α/β [26]. Due to preferential induction in hepatocytes, IFN-λ may be more relevant than IFN-α/β for HBV control [27], and its powerful and localized antiviral role suggested that this cytokine might be effective therapeutically. However, although PEG-IFN-λ1 therapy reduced viral loads in chronic HBV patients, the reduction achieved during the treatment duration was inferior post-treatment compared to PEG-IFN-α [28].

Innate immune cells

HBV interactions with the various subsets of innate immune cells may impact the establishment of chronic infection. Similar to the capacity of HBV to initiate IFN responses, the relationship between HBV and several innate cell types is debated. The diversity of results observed may be explained by these cells’ unique roles in different stages of chronic HBV disease (HBeAg-positive chronic infection, HBeAg-positive chronic hepatitis, HBeAg-negative chronic infection, HBeAg-negative chronic hepatitis) and the tissue (liver or blood) analyzed.

Dendritic cells (DC)

The exact role of DCs in chronic HBV infection remains unclear. Several studies reported distinct findings regarding altered DC number or function [29-34], which is likely due to diverse patient study populations and the use of different experimental approaches [35]. Recent analyses of plasmacytoid DC (pDC) function showed impaired maturation and function in chronic HBV patients [33,36]. pDCs efficiently produce IFN upon recognizing viral proteins or nucleic acids, so their dysfunction might play a vital role in HBV persistence. Additionally, HBV-exposed DCs and BDCA1+ DCs from infected patients were recently found to be poor stimulators of IFN-γ production by natural killer (NK) cells, indicating a link between DC impairment and NK cell function in chronic HBV [37].

NK cells

NK cells constitute up to 40-50% of human liver lymphocytes and can have two opposing functions. On the one hand, they have cytotoxic capacity independent of antigen recognition and can produce IFN-γ that down-regulates HBV replication. However, these cells also negatively regulate adaptive immune responses by inhibiting or deleting T cells [38]. This negative regulation occurs through inhibitory cytokines, cytolytic enzymes, and signaling via death receptor ligands such as TRAIL [38]. Liver-resident NK cells show impaired IFN-γ production in chronic HBV patients, implying dysfunctional antiviral activity [39]. Dysregulated expression or function of NK cell-activating (NKG2D) or inhibitory (NKG2A) receptors may further contribute to HBV persistence [40-43]. Additionally, HBV-specific CD8+ T cells from chronic HBV patients display an abnormal expression of the apoptosis-inducing receptor TRAIL-R2, rendering them sensitive to depletion by TRAIL-expressing NK cells [44]. Blocking this receptor on hepatic T cells increases HBV-specific T cell responses ex vivo [44], which further indicates that the immune suppressive function of NK cells might predominate over their antiviral activities in chronic hepatitis B.

Macrophages

Liver macrophages can be divided into monocyte-derived infiltrating macrophages and Kupffer cells, which are tissue-resident macrophages in the liver sinusoids. Kupffer cells have crucial phagocytic and scavenger functions and represent the first line of immune defense in the liver [45]. These cells are also important regulators of inflammation and liver damage, as Kupffer cell scavenger function reduces inflammation and immunopathology in an HBV mouse model [46]. Several studies have shown that HBV can alter macrophage function and cytokine expression in a manner that may impact viral infection or persistence [2,47-49]. Recent work also demonstrated that liver [50] and lymphatic [51] macrophages promote experimental HBV therapeutic vaccine efficacy, further supporting a role for these cells in linking innate and adaptive immunity to HBV.

Myeloid-derived suppressor cells (MDSCs)

MDSCs are immature myeloid cells that consist of two principal subsets, monocytic (mMDSCs) and granulocytic (gMDSCs) [52]. Increased MDSC number in the peripheral blood of patients with chronic HBV was initially described in two reports [53,54], and in both, the increased MDSC frequency was associated with lower liver immunopathology. CD14+HLA-DR−/low mMDSCs produce IL-10, which is responsible for their suppression of T cell function [53]. The CD14 gMDSC population inhibits both HBV-specific and bystander T cells in a partially arginase-1-dependent manner [54]. Importantly, it was recently found that HBeAg induces mMDSC expansion, which reduces T cell function through the indoleamine 2,3-dioxygenase pathway [55]. Further investigation of these findings could lead to a combination therapy that blocks MDSC-mediated immunosuppression and therefore favors T cell antiviral immune functions.

Mucosal-associated invariant T (MAIT) cells

MAIT cells are T cells with an invariant TCR alpha chain and restricted beta chains [56]. These cells can be activated independently of their TCR and are considered innate-like cells [57]. MAIT cells are highly abundant in the human liver and represent up to 40% of T cells [58], but their interaction with HBV has only recently been investigated and is not fully understood. While one study reported normal MAIT cell numbers in the blood that had increased activation levels [59], others found decreased MAIT cells in the blood [60-62], which were functionally impaired and had elevated PD-1 levels [61,62]. More recently, Liu et al. examined MAIT cells in liver tissue and blood and found reduced numbers in both compartments, and the cells displayed MR1-dependent anti-HBV activity but altered activation and TCR-mediated expansion [63].

Innate immunity therapeutics

The failure of HBV to activate innate immunity and evidence of HBV IFN sensitivity has led to therapeutic interventions targeting IFN system activation. Two main treatment strategies to overcome the lack of innate immune stimulation have emerged: exogenous IFN and pattern recognition receptor (PRR) agonists. Various clinical trials studying IFN-stimulating PRR agonists are ongoing or were recently completed (Table 1). PRRs such as RIG-I-like receptors (RLR) and Toll-like receptors (TLR) are critical for activating innate immunity as they recognize and respond to infections by binding pathogen-associated molecular patterns and propagating antiviral signal transduction pathways. In theory, agonist-mediated PRR stimulation could overcome the lack of an innate response to HBV infection by activating IFN production, and has the potential to be more targeted, effective, and safe compared to exogenous IFN administration. Therapies that aim to activate IFN responses could be particularly beneficial as the passive failure of HBV-mediated IFN induction suggests that viral interference with innate immune-stimulating therapeutic interventions will be unlikely.

Table I.

Recent ongoing and completed PRR agonist clinical trials

Drug Target Research
Phase
NCT Number* Status Company/Sponsor
inarigivir RIG-I Phase II
Phase II
NCT02751996
NCT03932513
completed (02/10/2020)
terminated (12/21/2019)
F-star Therapeutics, Inc.
vesatolimod TLR7 Phase II
Phase II
NCT02166047
NCT02579382
completed (10/20/2016)
completed (05/03/2019)
Gilead Sciences
AL-034 TLR7 Phase I NCT03285620 completed (11/14/2018) Alios Biopharma Inc.
RO7020531 TLR7 Phase I
Phase I
NCT03530917
NCT02956850
completed (05/15/2019)
recruiting (expected 05/22/2021)
Hoffmann-La Roche
TQ-A3334 TLR7 Phase II NCT04180150 recruiting (expected 12/31/2021) Chia Tai Tianqing Pharmaceutical Group Co., Ltd.
selgantolimod TLR8 Phase II
Phase II
NCT03491553
NCT03615066
completed (8/10/2020)
completed (05/28/2020)
Gilead Sciences
HepTcell/IC31 TLR9 Phase II NCT04684914 recruiting (expected 02/2023) Altimmune, Inc.
RO7020531 + RO7049389 TLR7 + CpAM Phase II NCT04225715 recruiting (expected 08/19/2023) Hoffmann-La Roche
TQ-A3334 + TQ-B2450 TLR7 + PD-L1 Phase II NCT04202653 not yet recruiting (expected 06/30/2021) Tongji Hospital

RIG-I

RLR agonists have been considered for chronic HBV therapies due to the cytoplasmic expression of their receptors in hepatocytes. Interestingly, a recent study found that RIG-I activation inhibits the formation and accelerates the decay of HBV covalently closed circular DNA (cccDNA) in hepatocytes [64]. The oral RIG-I/NOD2 agonist inarigivir (SB9200) reduced serum HBV DNA and antigens in the WHV woodchuck model [65], and was evaluated in combination with tenofovir in a completed Phase II clinical trial (Table 1). In 2020 however, inarigivir studies were discontinued following unexpected adverse events in a Phase IIb trial.

TLR7

Multiple TLR7 agonists have been investigated as the receptor is highly expressed by pDCs, and these cells potently produce IFN-α upon TLR7 stimulation [66]. Vesatolimod (GS-9620) yielded promising anti-HBV activity in chimpanzees and woodchucks [67,68]. Despite these results in animal models, vesatolimod treatment in humans showed no significant HBsAg decline either alone or in patients virally suppressed with nucleo(s/t)ide analogues [69-71]. AL-034 completed Phase I trials in 2018 and awaits further study (Table 1). RO7020531 completed a Phase I trial in chronic HBV patients in 2019 and is currently being further evaluated for safety and pharmacodynamics in another Phase I study, which is expected to finish in 2021. Another TLR7 agonist, TQ-A3334, is recruiting for a Phase II trial scheduled to be completed in 2021.

TLR8

Compared to TLR7 stimulation, TLR8 agonists preferentially induce the expression of IL-12 and other inflammatory cytokines from monocytes and myeloid DCs [66]. These cytokines may further stimulate IFN-γ production by other immune cells, which as discussed earlier, has important anti-HBV activity. Selgantolimod (GS-9688) is an oral TLR8 agonist that recently showed promising results in the WHV woodchuck model [72], and has been evaluated in two completed Phase II clinical trials alone and in combination with other antiviral therapies (Table 1). Recently published results have shown selgantolimod to be well-tolerated and to induce dose-dependent increases in serum cytokines [73,74]. Interestingly, human PBMCs from healthy and chronic HBV patients treated with selgantolimod in vitro exhibit enhanced DC and mononuclear phagocyte IL-12 production and increased frequencies of activated NK cells, MAIT cells, CD4+ follicular helper T cells, and HBV-specific CD8+ T cells expressing IFN-γ [75].

TLR9

CpG oligodeoxynucleotides (CpGs) have TLR9 agonist activity and were recently evaluated as adjuvants for HBV therapeutic vaccines [76]. The immunostimulatory properties of CpGs in the liver may in part derive from the fact that they promote the formation of structures known as iMATEs (intrahepatic myeloid cell aggregates associated with T cell expansion), which stimulate the population expansion of cytotoxic T lymphocytes in liver infection [77]. In the WHV woodchuck model, combined treatment with CpG and entecavir efficiently suppressed viral load and synergistically reduced serum HBsAg [78]. In late 2020, Altimmune, Inc. began a Phase II trial investigating the treatment of chronic HBV patients with an HBV peptide-based immunotherapeutic adjuvanted by the TLR9 agonist IC31 (Table 1).

Combination therapies

Despite mixed results in completed clinical trials, innate immunity therapeutic research has continued to advance (Table 1). Combination therapy with the TLR7 agonist RO7020531 and an HBV capsid allosteric modulator RO7049389 (CpAM) was effective in a preclinical AAV-based mouse model of HBV infection [79], and a phase II trial studying RO7020531 combined with RO7049389 began in July 2020 and is expected to be completed in March 2023. In a study not yet recruiting, the TLR7 agonist TQ-A3334 will be tested in combination with TQ-B2450, a humanized monoclonal antibody targeting programmed cell death ligand-1 (PD-L1). Checkpoint inhibitors targeting PD-1 or PD-L1 may restore impaired T cell function in chronically infected patients, and although their potential efficacy is currently unclear, a pilot study has provided promising results [80]. Combination approaches targeting innate and adaptive immunity may improve TLR agonist function; for example, treatment of human PBMCs with the TLR8 agonist selgantolimod led to a selection of MDSCs expressing higher levels of PD-L1 despite a beneficial reduction in MDSC frequency in vitro [75]. In January 2021, Gilead Sciences announced intentions to initiate Phase II trials investigating a combination of selgantolimod, antiviral siRNA VIR-2218 (Vir Biotechnology), and a commercially sourced PD-1 inhibitor. Preclinical studies of dual-acting agonists, such as the TLR2/7 agonist CL413 and the TLR7/8 agonist R848, have shown broader proinflammatory cytokine induction and enhanced efficacy compared to the single-acting TLR7 agonist CL264 or TLR9 agonist CpG-B in HBV-infected primary human hepatocytes [81]. It may be that the simultaneous activation of multiple PRRs yields a more robust cytokine spectrum capable of controlling HBV [81].

Conclusions

The passive evasion of IFN activation by HBV implies that therapeutic approaches aiming to stimulate these pathways will not be restrained by mechanisms of viral evasion or resistance, yet, the success of these approaches clinically has been thus far limited. Though PEG-IFN-α treatment can lead to significant sustained reductions in viremia and serum HBsAg in some cases, further work is needed to understand the mechanisms by which these positive clinical outcomes are achieved. This phenomenon nevertheless motivates future therapeutic advancements utilizing innate immune stimulation. To continue to improve upon previous work, it may be necessary to focus more specifically on the interplay of the innate and adaptive immune systems (Figure 1). It is possible that cell-specific targeting of DCs, NK cells, or MDSCs might reactivate the virus-specific T cells that these cells regulate [39,44,54]. Altering macrophage function to enhance T cell activation or to limit immunopathology is another tactic to consider [48]. Further, it is clear that cytokines such as IFN-α/β, -γ, and -λ can reduce HBV replication or promote adaptive immunity [82], so strategies that aim to activate innate immune cell production of these and other cytokines warrant continued evaluation. Combining these approaches with other methods to activate adaptive immunity, such as therapeutic vaccines, may also increase clinical efficacy. While an HBV cure has remained elusive, recent advancements nevertheless call for optimism and the need for continued basic and translational efforts focused on innate immune stimulation.

Figure 1. Therapeutic impacts of innate immune cells and cytokines on HBV replication.

Figure 1.

Stimulating IFN-α/β or IFN-λ production by hepatocytes or DCs with RLR or TLR agonists, or activating IFN-γ production by T cells, NK cells, or MAIT cells may directly inhibit HBV replication in hepatocytes. In addition to activating innate antiviral mechanisms, IFN-α/β production may also stimulate virus-specific intrahepatic T cell responses. These T cell responses may be further enhanced by inhibiting the negative regulatory activities of MDSCs and NK cells or by altering macrophage function, which may also reduce immunopathology.

Acknowledgments

This publication was supported in part by the National Institute of Allergy and Infectious Diseases of the National Institutes of Health under award number R01AI124006 and R01AI148354. The content is solely the responsibility of the authors and does not necessarily represent the official views of the National Institutes of Health. Figure 1 was created using Servier Medical Art, a publicly available service provided by Les Laboratoires Servier. Images have been adapted for the purposes of the Figure and their use does not imply endorsement by Les Laboratoires Servier.

Footnotes

Competing interests statement

M.D.R. reports grants from NIH/NIAID during the conduct of the work, financial relationships with CaroGen Corporation, and research funding from Gilead Sciences outside of this work. In addition, M.D.R. has received royalties from a patent issued to Yale University. The other authors declare no competing interests.

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

References

  • 1.Wieland SF, Chisari FV: Stealth and cunning: hepatitis B and hepatitis C viruses. J Virol 2005, 79:9369–9380. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Cheng X, Xia Y, Serti E, Block PD, Chung M, Chayama K, Rehermann B, Liang TJ: Hepatitis B virus evades innate immunity of hepatocytes but activates cytokine production by macrophages. Hepatology 2017, 66:1779–1793. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Mutz P, Metz P, Lempp FA, Bender S, Qu B, Schoneweis K, Seitz S, Tu T, Restuccia A, Frankish J, et al. : HBV Bypasses the Innate Immune Response and Does Not Protect HCV From Antiviral Activity of Interferon. Gastroenterology 2018, 154:1791–1804. [DOI] [PubMed] [Google Scholar]
  • 4.Suslov A, Boldanova T, Wang X, Wieland S, Heim MH: Hepatitis B Virus Does Not Interfere With Innate Immune Responses in the Human Liver. Gastroenterology 2018, 154:1778–1790.**This study demonstrated a lack of IFN activation in ex vivo HBV patient liver biopsy samples, and also found no inhibition of the IFN response by HBV after exogenous stimulation.
  • 5.Wieland S, Thimme R, Purcell RH, Chisari FV: Genomic analysis of the host response to hepatitis B virus infection. Proc Natl Acad Sci U S A 2004, 101:6669–6674. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Suresh M, Czerwinski S, Murreddu MG, Kallakury BV, Ramesh A, Gudima SO, Menne S: Innate and adaptive immunity associated with resolution of acute woodchuck hepatitis virus infection in adult woodchucks. PLoS Pathog 2019, 15:e1008248.*These results showed no activation of IFN or ISG expression in the woodchuck liver early after WHV infection.
  • 7.Winer BY, Huang TS, Pludwinski E, Heller B, Wojcik F, Lipkowitz GE, Parekh A, Cho C, Shrirao A, Muir TW, et al. : Long-term hepatitis B infection in a scalable hepatic co-culture system. Nat Commun 2017, 8:125. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Winer BY, Gaska JM, Lipkowitz G, Bram Y, Parekh A, Parsons L, Leach R, Jindal R, Cho Ch, Shrirao A, et al. : Analysis of Host Responses to Hepatitis B and Delta Viral Infections in a Micro-scalable Hepatic Co-culture System. Hepatology 2020, 71:14–30.**This work showed minimal IFN activation by HBV in a novel hepatic cell culture system.
  • 9.Lucifora J, Durantel D, Testoni B, Hantz O, Levrero M, Zoulim F: Control of hepatitis B virus replication by innate response of HepaRG cells. Hepatology 2010, 51:63–72. [DOI] [PubMed] [Google Scholar]
  • 10.Wang H, Ryu WS: Hepatitis B virus polymerase blocks pattern recognition receptor signaling via interaction with DDX3: implications for immune evasion. PLoS Pathog 2010, 6:e1000986. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Wang X, Li Y, Mao A, Li C, Li Y, Tien P: Hepatitis B virus X protein suppresses virus-triggered IRF3 activation and IFN-beta induction by disrupting the VISA-associated complex. Cell Mol Immunol 2010, 7:341–348. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Zhou L, He R, Fang P, Li M, Yu H, Wang Q, Yu Y, Wang F, Zhang Y, Chen A, et al. : Hepatitis B virus rigs the cellular metabolome to avoid innate immune recognition. Nat Commun 2021, 12:98. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Zhang Z, Trippler M, Real CI, Werner M, Luo X, Schefczyk S, Kemper T, Anastasiou OE, Ladiges Y, Treckmann J, et al. : Hepatitis B Virus Particles Activate Toll-Like Receptor 2 Signaling Initially Upon Infection of Primary Human Hepatocytes. Hepatology 2020, 72:829–844.*The authors found activation of some inflammatory gene expression, but not IFN or ISGs, in HBV-infected primary human hepatocytes.
  • 14.Guidotti LG, Chisari FV: Cytokine-induced viral purging--role in viral pathogenesis. Curr Opin Microbiol 1999, 2:388–391. [DOI] [PubMed] [Google Scholar]
  • 15.Pagliaccetti NE, Chu EN, Bolen CR, Kleinstein SH, Robek MD: Lambda and alpha interferons inhibit hepatitis B virus replication through a common molecular mechanism but with different in vivo activities. Virology 2010, 401:197–206. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Uprichard SL, Wieland SF, Althage A, Chisari FV: Transcriptional and posttranscriptional control of hepatitis B virus gene expression. Proc Natl Acad Sci U S A 2003, 100:1310–1315. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Wieland SF, Eustaquio A, Whitten-Bauer C, Boyd B, Chisari FV: Interferon prevents formation of replication-competent hepatitis B virus RNA-containing nucleocapsids. Proc Natl Acad Sci U S A 2005, 102:9913–9917. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Imam H, Kim GW, Mir SA, Khan M, Siddiqui A: Interferon-stimulated gene 20 (ISG20) selectively degrades N6-methyladenosine modified Hepatitis B Virus transcripts. PLoS Pathog 2020, 16:e1008338. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Wang YX, Niklasch M, Liu T, Wang Y, Shi B, Yuan W, Baumert TF, Yuan Z, Tong S, Nassal M, et al. : Interferon-inducible MX2 is a host restriction factor of hepatitis B virus replication. J Hepatol 2020, 72:865–876. [DOI] [PubMed] [Google Scholar]
  • 20.Kawashima K, Isogawa M, Onishi M, Baudi I, Saito S, Nakajima A, Fujita T, Tanaka Y: Restoration of type I interferon signaling in intrahepatically primed CD8+ T cells promotes functional differentiation. JCI Insight 2021, 6. doi: 10.1172/jci.insight.145761. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Gill US, Peppa D, Micco L, Singh HD, Carey I, Foster GR, Maini MK, Kennedy PT: Interferon Alpha Induces Sustained Changes in NK Cell Responsiveness to Hepatitis B Viral Load Suppression In Vivo. PLoS Pathog 2016, 12:e1005788. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Guidotti LG, Ishikawa T, Hobbs MV, Matzke B, Schreiber R, Chisari FV: Intracellular inactivation of the hepatitis B virus by cytotoxic T lymphocytes. Immunity 1996, 4:25–36. [DOI] [PubMed] [Google Scholar]
  • 23.Guidotti LG, Rochford R, Chung J, Shapiro M, Purcell R, Chisari FV: Viral clearance without destruction of infected cells during acute HBV infection. Science 1999, 284:825–829. [DOI] [PubMed] [Google Scholar]
  • 24.Lau JY, Lai CL, Wu PC, Chung HT, Lok AS, Lin HJ: A randomised controlled trial of recombinant interferon-gamma in Chinese patients with chronic hepatitis B virus infection. J Med Virol 1991, 34:184–187. [DOI] [PubMed] [Google Scholar]
  • 25.Ye L, Schnepf D, Staeheli P: Interferon-lambda orchestrates innate and adaptive mucosal immune responses. Nat Rev Immunol 2019, 19:614–625. [DOI] [PubMed] [Google Scholar]
  • 26.Bolen CR, Ding S, Robek MD, Kleinstein SH: Dynamic expression profiling of type I and type III interferon-stimulated hepatocytes reveals a stable hierarchy of gene expression. Hepatology 2014, 59:1262–1272. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Sato S, Li K, Kameyama T, Hayashi T, Ishida Y, Murakami S, Watanabe T, Iijima S, Sakurai Y, Watashi K, et al. : The RNA sensor RIG-I dually functions as an innate sensor and direct antiviral factor for hepatitis B virus. Immunity 2015, 42:123–132. [DOI] [PubMed] [Google Scholar]
  • 28.Chan HLY, Ahn SH, Chang TT, Peng CY, Wong D, Coffin CS, Lim SG, Chen PJ, Janssen HLA, Marcellin P, et al. : Peginterferon lambda for the treatment of HBeAg-positive chronic hepatitis B: A randomized phase 2b study (LIRA-B). J Hepatol 2016, 64:1011–1019. [DOI] [PubMed] [Google Scholar]
  • 29.Beckebaum S, Cicinnati VR, Dworacki G, Muller-Berghaus J, Stolz D, Harnaha J, Whiteside TL, Thomson AW, Lu L, Fung JJ, et al. : Reduction in the circulating pDC1/pDC2 ratio and impaired function of ex vivo-generated DC1 in chronic hepatitis B infection. Clin Immunol 2002, 104:138–150. [DOI] [PubMed] [Google Scholar]
  • 30.Gehring AJ, Haniffa M, Kennedy PT, Ho ZZ, Boni C, Shin A, Banu N, Chia A, Lim SG, Ferrari C, et al. : Mobilizing monocytes to cross-present circulating viral antigen in chronic infection. J Clin Invest 2013, 123:3766–3776. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Vincent IE, Zannetti C, Lucifora J, Norder H, Protzer U, Hainaut P, Zoulim F, Tommasino M, Trepo C, Hasan U, et al. : Hepatitis B virus impairs TLR9 expression and function in plasmacytoid dendritic cells. PLoS One 2011, 6:e26315. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.van der Molen RG, Sprengers D, Binda RS, de Jong EC, Niesters HG, Kusters JG, Kwekkeboom J, Janssen HL: Functional impairment of myeloid and plasmacytoid dendritic cells of patients with chronic hepatitis B. Hepatology 2004, 40:738–746. [DOI] [PubMed] [Google Scholar]
  • 33.Ouaguia L, Leroy V, Dufeu-Duchesne T, Durantel D, Decaens T, Hubert M, Valladeau-Guilemond J, Bendriss-Vermare N, Chaperot L, Aspord C: Circulating and Hepatic BDCA1+, BDCA2+, and BDCA3+ Dendritic Cells Are Differentially Subverted in Patients With Chronic HBV Infection. Front Immunol 2019, 10:112. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Martinet J, Dufeu-Duchesne T, Bruder Costa J, Larrat S, Marlu A, Leroy V, Plumas J, Aspord C: Altered functions of plasmacytoid dendritic cells and reduced cytolytic activity of natural killer cells in patients with chronic HBV infection. Gastroenterology 2012, 143:1586–1596. [DOI] [PubMed] [Google Scholar]
  • 35.Gehring AJ, Ann D'Angelo J: Dissecting the dendritic cell controversy in chronic hepatitis B virus infection. Cell Mol Immunol 2015, 12:283–291. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Yonejima A, Mizukoshi E, Tamai T, Nakagawa H, Kitahara M, Yamashita T, Arai K, Terashima T, Iida N, Fushimi K, et al. : Characteristics of Impaired Dendritic Cell Function in Patients With Hepatitis B Virus Infection. Hepatology 2019, 70:25–39.*This study demonstrated altered DC function in patients with chronic HBV infection that correlated with IL-6 signal transducer (IL6ST) expression.
  • 37.De Pasquale C, Campana S, Barberi C, Sidoti Migliore G, Oliveri D, Lanza M, Musolino C, Raimondo G, Ferrone S, Pollicino T, et al. : Human hepatitis B virus negatively impacts the protective immune cross-talk between natural killer and dendritic cells. Hepatology 2021. doi: 10.1002/hep.31725.**This study identified HBV-mediated changes in DC function that affect NK cell proliferation and IFN-γ expression.
  • 38.Crouse J, Xu HC, Lang PA, Oxenius A: NK cells regulating T cell responses: mechanisms and outcome. Trends Immunol 2015, 36:49–58. [DOI] [PubMed] [Google Scholar]
  • 39.Peppa D, Micco L, Javaid A, Kennedy PT, Schurich A, Dunn C, Pallant C, Ellis G, Khanna P, Dusheiko G, et al. : Blockade of immunosuppressive cytokines restores NK cell antiviral function in chronic hepatitis B virus infection. PLoS Pathog 2010, 6:e1001227. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Huang WC, Easom NJ, Tang XZ, Gill US, Singh H, Robertson F, Chang C, Trowsdale J, Davidson BR, Rosenberg WM, et al. : T Cells Infiltrating Diseased Liver Express Ligands for the NKG2D Stress Surveillance System. J Immunol 2017, 198:1172–1182. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Li F, Wei H, Wei H, Gao Y, Xu L, Yin W, Sun R, Tian Z: Blocking the natural killer cell inhibitory receptor NKG2A increases activity of human natural killer cells and clears hepatitis B virus infection in mice. Gastroenterology 2013, 144:392–401. [DOI] [PubMed] [Google Scholar]
  • 42.Ma Q, Dong X, Liu S, Zhong T, Sun D, Zong L, Zhao C, Lu Q, Zhang M, Gao Y, et al. : Hepatitis B e Antigen Induces NKG2A(+) Natural Killer Cell Dysfunction via Regulatory T Cell-Derived Interleukin 10 in Chronic Hepatitis B Virus Infection. Front Cell Dev Biol 2020, 8:421. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Wang Y, Wang W, Shen C, Wang Y, Jiao M, Yu W, Yin H, Shang X, Liang Q, Zhao C: NKG2D modulates aggravation of liver inflammation by activating NK cells in HBV infection. Sci Rep 2017, 7:88. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Peppa D, Gill US, Reynolds G, Easom NJ, Pallett LJ, Schurich A, Micco L, Nebbia G, Singh HD, Adams DH, et al. : Up-regulation of a death receptor renders antiviral T cells susceptible to NK cell-mediated deletion. J Exp Med 2013, 210:99–114. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Faure-Dupuy S, Durantel D, Lucifora J: Liver macrophages: Friend or foe during hepatitis B infection? Liver Int 2018, 38:1718–1729. [DOI] [PubMed] [Google Scholar]
  • 46.Sitia G, Iannacone M, Aiolfi R, Isogawa M, van Rooijen N, Scozzesi C, Bianchi ME, von Andrian UH, Chisari FV, Guidotti LG: Kupffer cells hasten resolution of liver immunopathology in mouse models of viral hepatitis. PLoS Pathog 2011, 7:e1002061. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Boltjes A, van Montfoort N, Biesta PJ, Op den Brouw ML, Kwekkeboom J, van der Laan LJ, Janssen HL, Boonstra A, Woltman AM: Kupffer cells interact with hepatitis B surface antigen in vivo and in vitro, leading to proinflammatory cytokine production and natural killer cell function. J Infect Dis 2015, 211:1268–1278. [DOI] [PubMed] [Google Scholar]
  • 48.Faure-Dupuy S, Delphin M, Aillot L, Dimier L, Lebosse F, Fresquet J, Parent R, Matter MS, Rivoire M, Bendriss-Vermare N, et al. : Hepatitis B virus-induced modulation of liver macrophage function promotes hepatocyte infection. J Hepatol 2019, 71:1086–1098. [DOI] [PubMed] [Google Scholar]
  • 49.Tian Y, Kuo CF, Akbari O, Ou JH: Maternal-Derived Hepatitis B Virus e Antigen Alters Macrophage Function in Offspring to Drive Viral Persistence after Vertical Transmission. Immunity 2016, 44:1204–1214. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Horng JH, Lin WH, Wu CR, Lin YY, Wu LL, Chen DS, Chen PJ: HBV X protein-based therapeutic vaccine accelerates viral antigen clearance by mobilizing monocyte infiltration into the liver in HBV carrier mice. J Biomed Sci 2020, 27:70. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Wang W, Zhou X, Bian Y, Wang S, Chai Q, Guo Z, Wang Z, Zhu P, Peng H, Yan X, et al. : Dual-targeting nanoparticle vaccine elicits a therapeutic antibody response against chronic hepatitis B. Nat Nanotechnol 2020, 15:406–416. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Bergenfelz C, Leandersson K: The Generation and Identity of Human Myeloid-Derived Suppressor Cells. Front Oncol 2020, 10:109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Huang A, Zhang B, Yan W, Wang B, Wei H, Zhang F, Wu L, Fan K, Guo Y: Myeloid-derived suppressor cells regulate immune response in patients with chronic hepatitis B virus infection through PD-1-induced IL-10. J Immunol 2014, 193:5461–5469. [DOI] [PubMed] [Google Scholar]
  • 54.Pallett LJ, Gill US, Quaglia A, Sinclair LV, Jover-Cobos M, Schurich A, Singh KP, Thomas N, Das A, Chen A, et al. : Metabolic regulation of hepatitis B immunopathology by myeloid-derived suppressor cells. Nat Med 2015, 21:591–600. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Yang F, Yu X, Zhou C, Mao R, Zhu M, Zhu H, Ma Z, Mitra B, Zhao G, Huang Y, et al. : Hepatitis B e antigen induces the expansion of monocytic myeloid-derived suppressor cells to dampen T-cell function in chronic hepatitis B virus infection. PLoS Pathog 2019, 15:e1007690.*This work demonstrated that HBeAg induces mMDSC expansion, which impairs T cell proliferation and IFN-γ production in an IDO-dependent manner.
  • 56.Toubal A, Nel I, Lotersztajn S, Lehuen A: Mucosal-associated invariant T cells and disease. Nat Rev Immunol 2019, 19:643–657. [DOI] [PubMed] [Google Scholar]
  • 57.Godfrey DI, Koay HF, McCluskey J, Gherardin NA: The biology and functional importance of MAIT cells. Nat Immunol 2019, 20:1110–1128. [DOI] [PubMed] [Google Scholar]
  • 58.Bolte FJ, Rehermann B: Mucosal-Associated Invariant T Cells in Chronic Inflammatory Liver Disease. Semin Liver Dis 2018, 38:60–65. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Boeijen LL, Montanari NR, de Groen RA, van Oord GW, van der Heide-Mulder M, de Knegt RJ, Boonstra A: Mucosal-Associated Invariant T Cells Are More Activated in Chronic Hepatitis B, but Not Depleted in Blood: Reversal by Antiviral Therapy. J Infect Dis 2017, 216:969–976. [DOI] [PubMed] [Google Scholar]
  • 60.Dias J, Hengst J, Parrot T, Leeansyah E, Lunemann S, Malone DFG, Hardtke S, Strauss O, Zmmer CL, Berglin L, et al. : Chronic hepatitis delta virus infection leads to functional impairment and severe loss of MAIT cells. J Hepatol 2019, 71:301–312. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Yong YK, Saeidi A, Tan HY, Rosmawati M, Enstrom PF, Batran RA, Vasuki V, Chattopadhyay I, Murugesan A, Vignesh R, et al. : Hyper-Expression of PD-1 Is Associated with the Levels of Exhausted and Dysfunctional Phenotypes of Circulating CD161(++)TCR iValpha7.2(+) Mucosal-Associated Invariant T Cells in Chronic Hepatitis B Virus Infection. Front Immunol 2018, 9:472. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Yong YK, Tan HY, Saeidi A, Rosmawati M, Atiya N, Ansari AW, Rajarajeswaran J, Vadivelu J, Velu V, Larsson M, et al. : Decrease of CD69 levels on TCR Valpha7.2(+)CD4(+) innate-like lymphocytes is associated with impaired cytotoxic functions in chronic hepatitis B virus-infected patients. Innate Immun 2017, 23:459–467. [DOI] [PubMed] [Google Scholar]
  • 63.Liu Y, Zhu P, Wang W, Tan X, Liu C, Chen Y, Pei R, Cheng X, Wu M, Guo Q, et al. : Mucosal-Associated Invariant T Cell Dysregulation Correlates With Conjugated Bilirubin Level in Chronic HBV Infection. Hepatology 2020. doi: 10.1002/hep.31602.*The authors found MR1-dependent MAIT cell anti-HBV activity, and showed that conjugated bilirubin can impact MAIT cell frequency and function.
  • 64.Lee S, Goyal A, Perelson AS, Ishida Y, Saito T, Gale M Jr.: Suppression of hepatitis B virus through therapeutic activation of RIG-I and IRF3 signaling in hepatocytes. iScience 2021, 24:101969. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Korolowicz KE, Iyer RP, Czerwinski S, Suresh M, Yang J, Padmanabhan S, Sheri A, Pandey RK, Skell J, Marquis JK, et al. : Antiviral Efficacy and Host Innate Immunity Associated with SB 9200 Treatment in the Woodchuck Model of Chronic Hepatitis B. PLoS One 2016, 11:e0161313. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Gorden KB, Gorski KS, Gibson SJ, Kedl RM, Kieper WC, Qiu X, Tomai MA, Alkan SS, Vasilakos JP: Synthetic TLR agonists reveal functional differences between human TLR7 and TLR8. J Immunol 2005, 174:1259–1268. [DOI] [PubMed] [Google Scholar]
  • 67.Lanford RE, Guerra B, Chavez D, Giavedoni L, Hodara VL, Brasky KM, Fosdick A, Frey CR, Zheng J, Wolfgang G, et al. : GS-9620, an oral agonist of Toll-like receptor-7, induces prolonged suppression of hepatitis B virus in chronically infected chimpanzees. Gastroenterology 2013, 144:1508–1517. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Menne S, Tumas DB, Liu KH, Thampi L, AlDeghaither D, Baldwin BH, Bellezza CA, Cote PJ, Zheng J, Halcomb R, et al. : Sustained efficacy and seroconversion with the Toll-like receptor 7 agonist GS-9620 in the Woodchuck model of chronic hepatitis B. J Hepatol 2015, 62:1237–1245. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Gane EJ, Lim YS, Gordon SC, Visvanathan K, Sicard E, Fedorak RN, Roberts S, Massetto B, Ye Z, Pflanz S, et al. : The oral toll-like receptor-7 agonist GS-9620 in patients with chronic hepatitis B virus infection. J Hepatol 2015, 63:320–328. [DOI] [PubMed] [Google Scholar]
  • 70.Janssen HLA, Brunetto MR, Kim YJ, Ferrari C, Massetto B, Nguyen AH, Joshi A, Woo J, Lau AH, Gaggar A, et al. : Safety, efficacy and pharmacodynamics of vesatolimod (GS-9620) in virally suppressed patients with chronic hepatitis B. J Hepatol 2018, 68:431–440. [DOI] [PubMed] [Google Scholar]
  • 71.Agarwal K, Ahn SH, Elkhashab M, Lau AH, Gaggar A, Bulusu A, Tian X, Cathcart AL, Woo J, Subramanian GM, 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. [DOI] [PubMed] [Google Scholar]
  • 72.Daffis S, Balsitis S, Chamberlain J, Zheng J, Santos R, Rowe W, Ramakrishnan D, Pattabiraman D, Spurlock S, Chu R, et al. : Toll-Like Receptor 8 Agonist GS-9688 Induces Sustained Efficacy in the Woodchuck Model of Chronic Hepatitis B. Hepatology 2020. doi.org 10.1002/hep.31255.*This study demonstrated that a TLR8 agonist is well-tolerated and activates a sustained antiviral response in the WHV woodchuck infection model.
  • 73.Gane EJ, Kim HJ, Visvanathan K, Kim YJ, Nguyen AH, Wallin JJ, Chen DY, McDonald C, Arora P, Tan SK, et al. : Safety, pharmacokinetics, and pharmacodynamics of the oral TLR8 agonist selgantolimod in chronic hepatitis B. Hepatology 2021. doi: 10.1002/hep.31795 [DOI] [PubMed] [Google Scholar]
  • 74.Reyes M, Lutz JD, Lau AH, Gaggar A, Grant EP, Joshi A, Mackman RL, Ling J, Tan SK, Ayithan N, et al. : Safety, pharmacokinetics and pharmacodynamics of selgantolimod, an oral Toll-like receptor 8 agonist: a Phase Ia study in healthy subjects. Antivir Ther 2020, 25:171–180. [DOI] [PubMed] [Google Scholar]
  • 75.Amin OE, Colbeck EJ, Daffis S, Khan S, Ramakrishnan D, Pattabiraman D, Chu R, Micolochick Steuer H, Lehar S, Peiser L, et al. : Therapeutic potential of TLR8 agonist GS-9688 (selgantolimod) in chronic hepatitis B: re-modelling of antiviral and regulatory mediators. Hepatology 2020. doi: 10.1002/hep.31695.**This work elucidated the impact of TLR8 agonism on immune cell frequency and function in PBMC from healthy controls and chronic HBV patients stimulated with GS-9688 in vitro.
  • 76.Kosinska AD, Moeed A, Kallin N, Festag J, Su J, Steiger K, Michel ML, Protzer U, Knolle PA: Synergy of therapeutic heterologous prime-boost hepatitis B vaccination with CpG-application to improve immune control of persistent HBV infection. Sci Rep 2019, 9:10808. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Huang LR, Wohlleber D, Reisinger F, Jenne CN, Cheng RL, Abdullah Z, Schildberg FA, Odenthal M, Dienes HP, van Rooijen N, et al. : Intrahepatic myeloid-cell aggregates enable local proliferation of CD8(+) T cells and successful immunotherapy against chronic viral liver infection. Nat Immunol 2013, 14:574–583. [DOI] [PubMed] [Google Scholar]
  • 78.Meng Z, Zhang X, Pei R, Zhang E, Kemper T, Vollmer J, Davis HL, Glebe D, Gerlich W, Roggendorf M, et al. : Combination therapy including CpG oligodeoxynucleotides and entecavir induces early viral response and enhanced inhibition of viral replication in a woodchuck model of chronic hepadnaviral infection. Antiviral Res 2016, 125:14–24. [DOI] [PubMed] [Google Scholar]
  • 79.Gao L, Dai L, Yu Y, Gu L, Zhao J, Ji Y, Yun H, Zhu W, Young JAT: Combination treatment of a TLR7 agonist RO7020531 and a capsid assembly modulator RO7049389 achieved sustainable viral load suppression and HBsAg loss in an AAV-HBV mouse model. Journal of Viral Hepatitis 2018, 25:12–12. [Google Scholar]
  • 80.Gane E, Verdon DJ, Brooks AE, Gaggar A, Nguyen AH, Subramanian GM, Schwabe C, Dunbar PR: Anti-PD-1 blockade with nivolumab with and without therapeutic vaccination for virally suppressed chronic hepatitis B: A pilot study. J Hepatol 2019, 71:900–907. [DOI] [PubMed] [Google Scholar]
  • 81.Janovec V, Hodek J, Clarova K, Hofman T, Dostalik P, Fronek J, Chlupac J, Chaperot L, Durand S, Baumert TF, et al. : Toll-like receptor dual-acting agonists are potent inducers of PBMC-produced cytokines that inhibit hepatitis B virus production in primary human hepatocytes. Sci Rep 2020, 10:12767.*The authors found that dual acting TLR agonists show superior antiviral activity against HBV compared to single-acting agonists in infected primary human hepatocytes.
  • 82.Isorce N, Lucifora J, Zoulim F, Durantel D: Immune-modulators to combat hepatitis B virus infection: From IFN-alpha to novel investigational immunotherapeutic strategies. Antiviral Res 2015, 122:69–81. [DOI] [PubMed] [Google Scholar]

RESOURCES