Therapeutic Antiviral Effect of the Nucleic Acid Polymer REP 2055 against Persistent Duck Hepatitis B Virus Infection

Faseeha Noordeen, Catherine A Scougall, Arend Grosse, Qiao Qiao, Behzad B Ajilian, Georget Reaiche-Miller, John Finnie, Melanie Werner, Ruth Broering, Joerg F Schlaak, Andrew Vaillant, Allison R Jilbert, Faseeha Noordeen, Catherine A Scougall, Arend Grosse, Qiao Qiao, Behzad B Ajilian, Georget Reaiche-Miller, John Finnie, Melanie Werner, Ruth Broering, Joerg F Schlaak, Andrew Vaillant, Allison R Jilbert

Abstract

Previous studies have demonstrated that nucleic acid polymers (NAPs) have both entry and post-entry inhibitory activity against duck hepatitis B virus (DHBV) infection. The inhibitory activity exhibited by NAPs prevented DHBV infection of primary duck hepatocytes in vitro and protected ducks from DHBV infection in vivo and did not result from direct activation of the immune response. In the current study treatment of primary human hepatocytes with NAP REP 2055 did not induce expression of the TNF, IL6, IL10, IFNA4 or IFNB1 genes, confirming the lack of direct immunostimulation by REP 2055. Ducks with persistent DHBV infection were treated with NAP 2055 to determine if the post-entry inhibitory activity exhibited by NAPs could provide a therapeutic effect against established DHBV infection in vivo. In all REP 2055-treated ducks, 28 days of treatment lead to initial rapid reductions in serum DHBsAg and DHBV DNA and increases in anti-DHBs antibodies. After treatment, 6/11 ducks experienced a sustained virologic response: DHBsAg and DHBV DNA remained at low or undetectable levels in the serum and no DHBsAg or DHBV core antigen positive hepatocytes and only trace amounts of DHBV total and covalently closed circular DNA (cccDNA) were detected in the liver at 9 or 16 weeks of follow-up. In the remaining 5/11 REP 2055-treated ducks, all markers of DHBV infection rapidly rebounded after treatment withdrawal: At 9 and 16 weeks of follow-up, levels of DHBsAg and DHBcAg and DHBV total and cccDNA in the liver had rebounded and matched levels observed in the control ducks treated with normal saline which remained persistently infected with DHBV. These data demonstrate that treatment with the NAP REP 2055 can lead to sustained control of persistent DHBV infection. These effects may be related to the unique ability of REP 2055 to block release of DHBsAg from infected hepatocytes.

Conflict of interest statement

Competing Interests: This study was funded by Replicor Inc. Dr. Andrew Vaillant is employed by Replicor Inc. NAPs are protected by patents wholly owned by Replicor Inc and are currently in development for the treatment of hepatitis B / hepatitis D infection. Relevant patents are: US 8,067,385 Antiviral oligonucleotides targeting HBV US 8,008,270 Antiviral oligonucleotides targeting viral families US 7,358,068 Antiviral oligonucleotides, US 8,008,269 Antiviral oligonucleotides. This does not alter the authors' adherence to all the PLOS ONE policies on sharing data and materials, as detailed online in the guide for authors.

Figures

Fig 1. Experimental design of REP 2055…
Fig 1. Experimental design of REP 2055 and NS treatment in in vivo Experiment 1 and 2.
Fourteen-day-old ducks were infected with 5x108 DHBV genome equivalents via the jugular vein. In Experiment 1 (a) all ducks were treated by IP injection with REP 2055. Group 1 received 10 mg/kg/day from 1 day prior to DHBV infection to 14 dpi; Group 2 received 10 mg/kg/day from 12–19 dpi and 10 mg/kg once weekly for 49 days; Group 3 received 10 mg/kg/day from 4–18 dpi and; Group 4 received 2 mg/kg/day from 4–18 dpi. After treatment, ducks in Groups 1, 3 and 4 were followed for an additional 49 days, from 19–68 dpi. Liver biopsies were performed prior to treatment in Group 2 and at the end of treatment in Groups 1, 3 and 4. Autopsies were performed at 68 dpi at the end of treatment in Group 2 and the end of follow-up in Groups 1, 3 and 4. In Experiment 2 (b), 14 DHBV-infected ducks were treated by IP injection with 10 mg/kg/day of REP 2055 from 12–40 dpi. A control group of 14 DHBV-infected ducks received daily IP injections of NS. Blood samples were collected during treatment and from 41–103 dpi during the first 9 weeks of follow-up. Based on interim analysis of serum DHBV DNA, 7 REP 2055-treated ducks that maintained control of their infection and 7 randomly selected NS-treated ducks, were followed from 103–155 dpi (total 16 weeks of follow-up). Liver biopsies were performed in all animals at 12 dpi prior to treatment. Biopsies or autopsies were performed at 103 dpi (9 weeks follow-up) and additional autopsies were performed in 7 animals per group at 155 dpi (16 weeks follow-up).
Fig 2. Lack of cytokine gene upregulation…
Fig 2. Lack of cytokine gene upregulation in PHH treated with REP 2055.
The expression levels of TNF (a), IL6 (b), IL10 (c), IFNA4 (d) and IFNB1 (e) genes were assessed by quantitative RT-PCR 6 hr after treatment with the NAP REP 2055 (0.01–10 μM) or without treatment (w/o), ODN 2216 (a CpG oligonucleotide TLR-9 agonist; 2 μM), poly I:C (a double stranded RNA TLR-3 agonist; 25 μg/ml) or Pam3CK4 (a TLR-1/2 agonist; 1 μM). Values represent mean ± SEM (normalized to 100,000 copies of beta actin mRNA). Statistically significant changes compared to untreated controls are reported for p< 0.05 (*) and p< 0.01 (**).
Fig 3. In vivo Experiment 1.
Fig 3. In vivo Experiment 1.
Response to various REP 2055 treatment regimens in Groups 1–4. Individual duck data for serum DHBsAg (top row) and serum DHBV DNA (bottom row). Days of REP 2055 treatment are indicated at the top of each graph by inverted triangles (see also Fig 1A). LLOQ for DHBsAg (0.88 μg/ml) and DHBV DNA (24000 copies/ml) are shown as dashed lines.
Fig 4. In vivo Experiment 2.
Fig 4. In vivo Experiment 2.
Response to treatment with NS or REP 2055. Levels of DHBsAg (a), anti-DHBs antibodies (b), DHBV DNA (c and e) and anti-DHBc antibodies (d) in the serum and DHBV total and cccDNA in the liver (f) during and after treatment are shown. For NS-treated ducks in all panels, n = 13 except for the last 7 measurements, where n = 7 (see *). Horizontal lines in (a-e) represent the lower limit of quantification. For REP 2055duck n = 6 but SVR duck 188 is plotted separately in (a) and (e). For REP 2055 NO SVR ducks, n = 5 but NO SVR duck 194 is plotted separately in (a) and (e). Duck 194 and duck 188 are plotted separately in (a) and (e) as the response of these ducks was different from the other SVR or NO SVR ducks. Treatment interval in (a-e) is indicated on the x-axis by the shaded bar. Differences in mean responses of REP 2055 SVR and REP 2055 NO SVR were significantly different from NS in a, b and c (p

References

    1. Kowdley KV. The cost of managing chronic hepatitis B infection: a global perspective. J. Clin. Gastroenterol. 2004; 38:S132–S133.
    1. Chang TT, Gish RG, de Man R, Gadano A, Sollano J, Chao YC et al. A comparison of entecavir and lamivudine for HBeAg-positive chronic hepatitis B. N. Engl. J. Med. 2006; 354:1001–1010.
    1. Lai CL, Shouval D, Lok AS, Chang TT, Cheinquer H, Goodman Z, et al. Entecavir versus lamivudine for patients with HBeAg-negative chronic hepatitis B. N. Engl. J. Med. 2006; 354:1011–1020
    1. Marcellin P, Heathcote EJ, Buti M, Gane E, de Man RA, Krastev Z, et al. Tenofovir disoproxil fumarate versus adefovir dipivoxil for chronic hepatitis B. N. Engl. J. Med. 2008; 359:2442–2455. 10.1056/NEJMoa0802878
    1. Lau GK, Piratvisuth T, Luo KX, Marcellin P, Thongsawat S, Cooksley G, et al. Peginterferon Alfa-2a, lamivudine, and the combination for HBeAg-positive chronic hepatitis B. N. Engl. J. Med. 2005; 352:2682–2695
    1. Marcellin P, Lau GK, Bonino F, Farci P, Hadziyannis S, Jin R, et al. Peginterferon alfa-2a alone, lamivudine alone, and the two in combination in patients with HBeAg-negative chronic hepatitis B. N. Engl. J. Med. 2004; 351:1206–1217.
    1. Vaillant A, Juteau JM, Lu H, Liu S, Lackman-Smith C, Ptak R et al. Phosphorothioate oligonucleotides inhibit human immunodeficiency virus type 1 fusion by blocking gp41 core formation. Antimicrob. Agents Chemother. 2006; 50:1393–1401.
    1. Lamb RA, Jardetzky TS. Structural basis of viral invasion lesion from paramyxovirus F. Curr. Opin. Struct. Biol. 2007; 17: 427–436.
    1. Kielian M., Rey FM. Virus membrane-fusion proteins: more than one way to make a hairpin. Nature Reviews 2006; 4: 67–76.
    1. Guzman EM, Cheshenko N, Shende V, Keller MJ, Goyette N, Juteau JM et al. Amphipathic DNA polymers are candidate vaginal microbicides and block herpes simplex virus binding, entry and viral gene expression. Antiviral Ther. 2007; 12:1147–1156.
    1. Bernstein DI, Goyette N, Cardin R, Kern ER, Boivin G, Ireland J et al. Amphipathic DNA polymers exhibit antiherpetic activity in vitro and in vivo. Antimicrob. Agents Chemother. 2008; 52:2727–2733. 10.1128/AAC.00279-08
    1. Lee AM, Rojek JM, Gundersen A, Ströher U, Juteau JM, Vaillant A et al. Inhibition of cellular entry of lymphocytic choriomeningitis virus by amphipathic DNA polymers. Virology 2008; 372:107–117.
    1. Cardin RD, Bravo FJ, Sewell AP, Cummins J, Flamand L, Juteau JM et al. Amphipathic DNA polymers exhibit antiviral activity against systemic murine Cytomegalovirus infection. Virol. J. 2007; 6:214.
    1. Agrawal S, Tang JY, Brown DM. Analytical study of phosphorothioate analogues of oligodeoxynucleotides using high performance liquid chromatography. J. Chromatogr. 1990; 509: 396–399.
    1. Matsumura T, Hu Z, Kato T, Dreux M, Zhang YY, Imamura M et al. Amphipathic DNA polymers inhibit hepatitis C virus infection by blocking entry. Gastroenterology 2009; 137:673–681. 10.1053/j.gastro.2009.04.048
    1. Noordeen F, Vaillant A, Jilbert AR. Nucleic acid polymers inhibit duck hepatitis B virus infection in vitro . Antimicrob. Agents Chemother. 2013; 57:5291–5298. 10.1128/AAC.01003-13
    1. Noordeen F, Vaillant A, Jilbert AR. Nucleic acid polymers prevent the establishment of duck hepatitis B virus infection in vivo . Antimicrob. Agents Chemother. 2013; 57:5299–5306. 10.1128/AAC.01005-13
    1. Broering R, Lutterbeck M, Trippler M, Kleinher K, Poggenpohl L, Paul A et al. Long-term stimulation of Toll-like receptor 3 in primary human hepatocytes leads to sensitization for antiviral responses induced by poly I:C treatment. J. Viral Hep. 2013; 21: 480–490.
    1. Foster WK, Miller DS, Scougall CA, Kotlarski I, Colonno RJ, Jilbert AR. The effect of antiviral treatment with entecavir on age- and dose-related outcomes of duck hepatitis B virus infection. J. Virol. 2005; 79:5819–5832.
    1. Jilbert AR, Botten JA, Miller DS, Bertram EM, Hall PM, Kotlarski I et al. Characterisation of age- and dose-related outcomes of duck hepatitis B virus infection. Virology 1998; 244:273–282.
    1. Miller DS, Halpern M, Kotlarski I, Jilbert AR. Vaccination of ducks with a whole-cell vaccine expressing duck hepatitis B virus core antigen elicits antiviral immune responses that enable rapid resolution of de novo infection. Virology 2006; 348:297–308.
    1. Miller DS, Kotlarski I, Jilbert AR. DNA vaccines expressing the duck hepatitis B virus surface proteins lead to reduced numbers of infected cells and protect ducks against the development of chronic infection in a virus dose dependent manner. Virology 2006; 351:159–169.
    1. Miller DS, Bertram EM, Scougall CA, Kotlarski I, Jilbert AR. Studying host immune responses against duck hepatitis B virus infection. 2004; Methods Mol. Med. 96: 3–25.
    1. Reaiche GY, Le Mire MF, Mason WS, Jilbert AR. The persistence in the liver of residual duck hepatitis B virus covalently closed circular DNA is not dependent upon new viral DNA synthesis. Virology 2010; 406:286–292. 10.1016/j.virol.2010.07.013
    1. Pugh JC, Di Q, Mason WS, Simmons H. Susceptibility to duck hepatitis B virus infection is associated with the presence of cell surface receptor sites that efficiently bind viral particles. J. Virol. 1995; 69:4814–4822.
    1. Jilbert AR, Miller DS, Scougall CA, Turnbull H, Burrell CJ. Kinetics of duck hepatitis B virus infection following low dose virus inoculation: One virus DNA genome is infectious in neonatal ducks. Virology 1996; 226:338–345.
    1. Jilbert AR, Wu TT, England JM, Hall PM, Carp NZ, O'Connell AP et al. Rapid resolution of duck hepatitis B virus infections occurs after massive hepatocellular involvement. J Virol. 1992; 66:1377–1388.
    1. Reaiche-Miller GY, Thorpe M, Low HC, Qiao Q, Scougall CA, Mason WS et al. Duck hepatitis B virus covalently closed circular DNA appears to survive hepatocyte mitosis in the growing liver. Virology. 2013; 446:357–364. 10.1016/j.virol.2013.08.014
    1. Le Mire MF, Miller DS, Foster WK, Burrell CJ, Jilbert AR. Covalently closed circular DNA is the predominant form of duck hepatitis B virus DNA that persists following transient infection. J. Virol. 2005; 79:12242–12252.
    1. Wu JW, Lu M., Meng Z, Trippler M, Broering R, Szczeponek A et al. Toll-like receptor-mediated control of HBV infection by non-parenchymal liver cells in mice. Hepatology 2007; 46: 1769–1778.
    1. Raal FJ, Santos RD, Blom DJ, Marais AD, Charng MJ, Cromwell WC et al. Mipomersen, an apolipoprotein B synthesis inhibitor, for lowering of LDL cholesterol concentrations in patients with homozygous familial hypercholesterolaemia: a randomised, double-blind, placebo-controlled trial. Lancet. 2010; 375:998–1006. 10.1016/S0140-6736(10)60284-X
    1. Galbraith W.M., Hobson W.C., Giclas P.C., Schechter P.J. and Agrawal S. Complement activation and hemodynamic changes following intravenous administration of phosphorothioate oligonucleotides in the monkey. Antisense Res. Dev. 1994; 4:201–206.
    1. Ruan P, Yang C, Su J, Cao J, Ou C, Luo C et al. Histopathological changes in the liver of tree shrew (Tupaia belangeri chinensis) persistently infected with hepatitis B virus. Virol. J. 2013; 10:333 10.1186/1743-422X-10-333
    1. Kajino K, Jilbert AR, Saputelli J, Aldrich CE, Cullen J, Mason WS. Woodchuck hepatitis virus infections: very rapid recovery after a prolonged viremia and infection of virtually every hepatocyte. J. Virol. 1994; 68: 5792–5803.
    1. 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
    1. Wieland SF, Spangenberg HC, Thimme R, Purcell RH, Chisari FV, Expansion and contraction of the hepatitis B virus transcriptional template in infected chimpanzees. Proc. Natl. Acad. Sci. U.S.A. 2004; 101: 2129–2134.
    1. Belloni L, Allweiss L, Guerrieri F, Pediconi N, Volz T, Pollicino T et al. IFN-a inhibits HBV transcription and replication in cell culture and in humanized mice by targeting the epigenetic regulation of the nuclear cccDNA minichromosome. J. Clin. Invest. 2012; 122: 529–537. 10.1172/JCI58847
    1. 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: 825–9.
    1. Foster WK, Miller DS, Marion PL, Colonno RJ, Kotlarski I, Jilbert AR. Entecavir therapy combined with DNA vaccination for persistent duck hepatitis B virus infection. Antimicrob. Agents Chemother. 2003; 47:2624–2635.
    1. Lin E, Luscombe C, Colledge D, Wang YY, Locarnini S. Long-term therapy with the guanine nucleoside analog penciclovir controls chronic duck hepatitis B virus infection in vivo. Antimicrob. Agents Chemother. 1998; 42:2132–2137.
    1. Nicoll AJ, Colledge DL, Toole JJ, Angus PW, Smallwood RA, Locarnini SA. Inhibition of duck hepatitis B virus replication by 9-(2-phosphonylmethoxyethyl) adenine, an acyclic phosphonate nucleoside analogue. Antimicrob. Agents Chemother. 1998; 42:3130–3135.
    1. Ganem D, Prince AM. Hepatitis B virus infection—natural history and clinical consequences. N. Engl. J. Med. 2004; 350:1118–1129.
    1. Franke C, Matschl U, Bruns M. Enzymatic treatment of duck hepatitis B virus: topology of the surface proteins for virions and non-infectious subviral particles. Virology 2007; 359: 26–136.
    1. Cheng J, Imanishi H, Morisaki H, Liu W, Nakamura H, Morisaki T et al. Recombinant HBsAg inhibits LPS-induced COX-2 expression and IL-18 production by interfering with the NFκB pathway in a human monocytic cell line, THP-1. J. Hepatol. 2005; 43:465–471.
    1. Op den Brouw ML, Binda RS, van Roosmalen MH, Protzer U, Janssen HL, van der Molen RG et al. Hepatitis B virus surface antigen impairs myeloid dendritic cell function: a possible immune escape mechanism of hepatitis B virus. Immunology 2008; 126:280–289. 10.1111/j.1365-2567.2008.02896.x
    1. Shi B, Ren G, Hu Y, Wang S, Zhang Z, Yuan Z. HBsAg inhibits INF-α production in plasmacytoid dendritic cells through TNFα and IL-10 induction in monocytes. PLOS One 2012; 7:e44900
    1. Vanlandschoot P, Van Houtte F, Roobrouck A, Farhoudi A, Lerous-Roels G. Hepatitis B virus surface antigen suppresses the activation of monocytes through interaction with a serum protein and a monocyte-specific receptor. J. Gen. Virol. 2002; 82:1281–1289.
    1. Wu J, Meng Z, Jiang M, Pei R, Trippler M, Broering R et al. Hepatitis B virus suppresses Toll-like receptor-mediated innate immune responses in murine parenchymal and nonparenchymal liver cells. Hepatology 2009; 49:1132–1140. 10.1002/hep.22751
    1. Xu Y, Hu Y, Shi B, Zhang X, Wang J, Zhang Z et al. HBsAg inhibits TLR-9-mediated activation and INF-α production in plasmacytoid dendritic cells. Mol. Immunol. 2009; 46:2640–2646. 10.1016/j.molimm.2009.04.031
    1. Moucari R, Mackiewicz V, Lada O, Ripault MP, Castelnau C, Martinot-Peignoux M et al. Early serum HBsAg drop: a strong predictor of sustained virological response to pegylated interferon alfa-2a in HBeAg-negative patients. Hepatology 2009; 49:1151–1157. 10.1002/hep.22744
    1. Frenette CT, Gish RG. To “Be” or not to “Be”: That is the question. Am. J. Gastroenterol. 2009; 104:1948–1952. 10.1038/ajg.2009.204
    1. Sunyach C, Chassot S, Jamard C, Kay A, Trepo C, Cova L. In vivo Selection of Duck Hepatitis B Virus Pre-S Variants Which Escape from Neutralization. Virology 1997; 234: 291–299.
    1. Burda MR, Gunther S, Dandri M, Will H, Petersen J. Structural and functional heterogeneity of naturally occurring hepatitis B virus variants. Antiviral Res. 2001; 52: 125–138
    1. Cooreman MP, Leroux-Roels G, Paulij WP. Vaccine- and hepatitis B immune globulin-induced escape mutations of hepatitis B virus surface antigen. J Biomed Sci. 2001; 8:237–47.
    1. Hsu CW, Yeh C-T. Emergence of Hepatitis B Virus S gene Mutants in Patients Experiencing Hepatitis B Surface Antigen Seroconversion After Peginterferon Therapy. Hepatology 2011; 54: 101–108. 10.1002/hep.24363
    1. Protzer-Knolle U, Naumann U, Bartenschlager R, Berg T, Hopf U, Meyer zum Büschenfelde KH et al. Hepatitis B virus with antigenically altered hepatitis B surface antigen is selected by high-dose hepatitis B immune globulin after liver transplantation. Hepatology 1998; 27: 254–63.

Source: PubMed

3
Prenumerera