Postexposure protection of guinea pigs against a lethal ebola virus challenge is conferred by RNA interference

Thomas W Geisbert, Lisa E Hensley, Elliott Kagan, Erik Zhaoying Yu, Joan B Geisbert, Kathleen Daddario-DiCaprio, Elizabeth A Fritz, Peter B Jahrling, Kevin McClintock, Janet R Phelps, Amy C H Lee, Adam Judge, Lloyd B Jeffs, Ian MacLachlan, Thomas W Geisbert, Lisa E Hensley, Elliott Kagan, Erik Zhaoying Yu, Joan B Geisbert, Kathleen Daddario-DiCaprio, Elizabeth A Fritz, Peter B Jahrling, Kevin McClintock, Janet R Phelps, Amy C H Lee, Adam Judge, Lloyd B Jeffs, Ian MacLachlan

Abstract

Background: Ebola virus (EBOV) infection causes a frequently fatal hemorrhagic fever (HF) that is refractory to treatment with currently available antiviral therapeutics. RNA interference represents a powerful, naturally occurring biological strategy for the inhibition of gene expression and has demonstrated utility in the inhibition of viral replication. Here, we describe the development of a potential therapy for EBOV infection that is based on small interfering RNAs (siRNAs).

Methods: Four siRNAs targeting the polymerase (L) gene of the Zaire species of EBOV (ZEBOV) were either complexed with polyethylenimine (PEI) or formulated in stable nucleic acid-lipid particles (SNALPs). Guinea pigs were treated with these siRNAs either before or after lethal ZEBOV challenge.

Results: Treatment of guinea pigs with a pool of the L gene-specific siRNAs delivered by PEI polyplexes reduced plasma viremia levels and partially protected the animals from death when administered shortly before the ZEBOV challenge. Evaluation of the same pool of siRNAs delivered using SNALPs proved that this system was more efficacious, as it completely protected guinea pigs against viremia and death when administered shortly after the ZEBOV challenge. Additional experiments showed that 1 of the 4 siRNAs alone could completely protect guinea pigs from a lethal ZEBOV challenge.

Conclusions: Further development of this technology has the potential to yield effective treatments for EBOV HF as well as for diseases caused by other agents that are considered to be biological threats.

Figures

Figure 1
Figure 1
Inhibition of the replication of the Zaire species of Ebola virus (ZEBOV) in Vero cells by a pool of 4 different small interfering RNAs (siRNAs) targeting individual regions of the ZEBOV polymerase (L) gene. Cells were transfected with either the siRNA pool or an equivalent dose of EbL-Scram1, an irrelevant scrambled sequence (SCR), as a control. At various time points after transfection (0, 24, and 48 h), the transfected cells were infected with ZEBOV, and cells and culture fluids were harvested 24 h later, for determination of the level of infectious virus by plaque assay
Figure 2
Figure 2
Inhibition of the replication of the Zaire species of Ebola virus (ZEBOV) in Vero cells, as demonstrated by immunofluorescence staining. Cells were transfected with either a pool of 4 different small interfering RNAs (siRNAs) targeting individual regions of the ZEBOV polymerase (L) gene or an equivalent dose of EbL-Scram1, an irrelevant scrambled sequence (SCR), as a control. Cells were counterstained with 4′,6′-diamidino-2-phenylindole and Evans blue, to aid visualization. ZEBOV-positive cells are identified by green fluorescence
Figure 3
Figure 3
Plasma viremia levels of inbred strain 13 guinea pigs 4 days after challenge with the Zaire species of Ebola virus (ZEBOV). Guinea pigs were treated 3 h before the ZEBOV challenge and 1, 2, and 4 days afterward with either a pool of 4 different small interfering RNAs (siRNAs) targeting individual regions of the ZEBOV polymerase (L) gene (n=5) or an equivalent dose of EbL-Scram1, an irrelevant scrambled sequence (SCR) (n=5). Data are means±SDs
Figure 4
Figure 4
In vivo clearance and biodistribution of stable nucleic acid–lipid particles (SNALPs). Shown are plasma clearance (A) and biodistribution (B and C) of [3H]cholesteryl oleyl ether–labeled SNALPs. Each guinea pig received a single intravenous injection of 0.75 mg/kg small interfering RNAs (siRNA) formulated as SNALPs. Biodistribution data were collected 24 h after injection. Data are means±SDs (for all tissues, n=5 guinea pigs, except for testes [n=3] and ovaries [n=2])
Figure 5
Figure 5
Antiviral efficacy of a pool of 4 different small interfering RNAs (siRNAs) targeting individual regions of the Zaire species of Ebola virus (ZEBOV) polymerase (L) gene and encapsulated in stable nucleic acid–lipid particles (SNALPs). Shown are plasma viremia levels (A) and survival rates (B) for inbred strain 13 guinea pigs after ZEBOV challenge. One hour after challenge and daily on days 1–6 thereafter, guinea pigs were treated, via the SNALP delivery system, with the siRNA pool (1.0 mg/kg) or an equivalent dose of EbL-Scram1, an irrelevant scrambled sequence (SCR). Plasma viremia levels were determined on day 7. Viremia data are means±SDs (n=5)
Figure 6
Figure 6
Antiviral efficacy of small interfering RNAs (siRNAs) targeting individual regions of the Zaire species of Ebola virus (ZEBOV) polymerase (L) gene and encapsulated in stable nucleic acid–lipid particles (SNALPs). Shown are plasma viremia levels (A) and survival rates (B) for inbred strain 13 guinea pigs after ZEBOV challenge. One hour after the challenge and daily on days 1–6 thereafter, guinea pigs were treated, via the SNALP delivery system, with a pool of 4 siRNA (0.75 mg/kg); an equivalent dose of EbL-Scram1, an irrelevant scrambled sequence (SCR); or 1 of the 4 siRNAs alone (EK1–EK4). Plasma viremia levels were determined on day 7. Viremia data are means±SDs (n=5). The P value is for the comparison between EK2 and SCR
Figure 7
Figure 7
Small interfering RNA (siRNA)–mediated cytokine induction in mice. Shown are serum interferon (IFN)–α (A) and IFN-β (B) levels 6 h after intravenous administration of 100 μg (∼5 mg/kg) of stable nucleic acid–lipid particles encapsulating either a pool of 4 different siRNAs that targeted individual regions of the Zaire species of Ebola virus polymerase (L) gene; an equivalent dose of EbL-Scram1, an irrelevant scrambled sequence (SCR); or 1 of the 4 siRNAs alone (EK1–EK4). Injection of PBS alone induced no detectable IFN-α or IFN-β. Note that injection of empty liposomes or naked siRNA alone also failed to induce detectable IFN-α or IFN-β (data not shown). Data are means±SDs (n=4)

References

    1. Walsh PD, Abernethy KA, Bermejo M, et al. Catastrophic ape decline in western equatorial Africa. Nature. 2003;422:611–4.
    1. International Society for Infectious Diseases Ebola, lab accident death—Russia (Siberia). Archive no: 20040522.1377. 22 May. 2004 Available at: . Accessed 1 May 2006.
    1. Sanchez A, Kiley MP, Holloway BP, Auperin DD. Sequence analysis of the Ebola virus genome: organization, genetic elements, and comparison with the genome of Marburg virus. Virus Res. 1993;29:215–40.
    1. Tan FL, Yin JQ. RNAi, a new therapeutic strategy against viral infection. Cell Res. 2004;14:460–6.
    1. Colbere-Garapin F, Blondel B, Saulnier A, Pelletier I, Labadie K. Silencing viruses by RNA interference. Microbes Infect. 2005;7:767–75.
    1. Fowler T, Bamberg S, Moller P, et al. Inhibition of Marburg virus protein expression and viral release by RNA interference. J Gen Virol. 2005;86:1181–8.
    1. Sanchez AB, Perez M, Cornu T, de la Torre JC. RNA interference-mediated virus clearance from cells both acutely and chronically infected with the prototypic arenavirus lymphocytic choriomeningitis virus. J Virol. 2005;79:11071–81.
    1. Wu CJ, Huang HW, Liu CY, Hong CF, Chan YL. Inhibition of SARS-CoV replication by siRNA. Antiviral Res. 2005;65:45–8.
    1. McCaffrey AP, Meuse L, Pham TT, Conklin DS, Hannon GJ. RNA interference in adult mice. Nature. 2002;418:38–9.
    1. Giladi H, Ketzinel-Gilad M, Rivkin L, Felig Y, Nussbaum O. Small interfering RNA inhibits hepatitis B virus replication in mice. Mol Ther. 2003;8:769–76.
    1. Song E, Lee SK, Wang J, et al. RNA interference targeting Fas protects mice from fulminant hepatitis. Nat Med. 2003;9:347–51.
    1. Ge Q, Filip L, Bai A, Nguyen T, Eisen HN. Inhibition of influenza virus production in virus-infected mice by RNA interference. Proc Natl Acad Sci USA. 2004;101:8676–81.
    1. Morrissey DV, Lockridge JA, Shaw L, et al. Potent and persistent in vivo anti-HBV activity of chemically modified siRNAs. Nat Biotechnol. 2005;23:1002–7.
    1. Connolly BM, Steele KE, Davis KJ, et al. Pathogenesis of experimental Ebola virus infection in guinea pigs. J Infect Dis. 1999;179(Suppl 1):S203–17.
    1. National Research Council . Guide for the care and use of laboratory animals. Washington, DC: National Academy Press; 1996.
    1. Jeffs LB, Palmer LR, Ambegia EG, Giesbrecht C, Ewanick S. A scalable, extrusion-free method for efficient liposomal encapsulation of plasmid DNA. Pharm Res. 2005;22:362–72.
    1. Heyes J, Palmer L, Bremner K, MacLachlan I. Cationic lipid saturation influences intracellular delivery of encapsulated nucleic acids. J Control Release. 2005;107:276–87.
    1. Stein Y, Halperin G, Stein O. Biological stability of [3H]cholesteryl oleyl ether in cultured fibroblasts and intact rat. FEBS Lett. 1980;111:104–6.
    1. Jahrling PB, Geisbert TW, Geisbert JB, et al. Evaluation of immune globulin and recombinant interferon-α2b for treatment of experimental Ebola virus infections. J Infect Dis. 1999;179(Suppl 1):S224–34.
    1. Mosca JD, Pitha PM. Transcriptional and posttranscriptional regulation of exogenous human beta interferon gene in simian cells defective in interferon synthesis. Mol Cell Biol. 1986;6:2279–83.
    1. Spann KM, Tran KC, Collins PL. Effects of nonstructural proteins NS1 and NS2 of human respiratory syncytial virus on interferon regulatory factor 3, NF-kappaB, and proinflammatory cytokines. J Virol. 2005;79:5353–62.
    1. Emeny JM, Morgan MJ. Regulation of the interferon system: evidence that Vero cells have a genetic defect in interferon production. J Gen Virol. 1979;43:247–52.
    1. Bray M, Davis K, Geisbert T, Schmaljohn C, Huggins J. A mouse model for evaluation of prophylaxis and therapy of Ebola hemorrhagic fever. J Infect Dis. 1998;178:651–61.
    1. Ryabchikova EI, Kolesnikova LV, Luchko SV. An analysis of features of pathogenesis in two animal models of Ebola virus infection. J Infect Dis. 1999;179(Suppl 1):S199–202.
    1. Geisbert TW, Pushko P, Anderson K, Smith J, Davis KJ. Evaluation in nonhuman primates of vaccines against Ebola virus. Emerg Infect Dis. 2002;8:503–7.
    1. Bray M, Hatfill S, Hensley L, Huggins JW. Haematological, biochemical and coagulation changes in mice, guinea-pigs and monkeys infected with a mouse-adapted variant of Ebola Zaire virus. J Comp Pathol. 2001;125:243–53.
    1. Bowen ET, Platt GS, Lloyd G, Raymond RT, Simpson DI. A comparative study of strains of Ebola virus isolated from southern Sudan and northern Zaire in 1976. J Med Virol. 1980;6:129–38.
    1. Hevey M, Negley D, Pushko P, Smith J, Schmaljohn A. Marburg virus vaccines based upon alphavirus replicons protect guinea pigs and nonhuman primates. Virology. 1998;251:28–37.
    1. Ryabchikova E, Strelets L, Kolesnikova L, Pyankov O, Sergeev A. Respiratory Marburg virus infection in guinea pigs. Arch Virol. 1996;141:2177–90.
    1. Fenske DB, MacLachlan I, Cullis PR. Stabilized plasmid-lipid particles: a systemic gene therapy vector. Methods Enzymol. 2002;346:36–71.
    1. Geisbert TW, Hensley LE, Larsen T, et al. Pathogenesis of Ebola hemorrhagic fever in cynomolgus macaques: evidence that dendritic cells are early and sustained targets of infection. Am J Pathol. 2003;163:2347–70.
    1. Nishikawa M, Yamauchi M, Morimoto K, Ishida E, Takakura Y. Hepatocyte-targeted in vivo gene expression by intravenous injection of plasmid DNA complexed with synthetic multi-functional gene delivery system. Gene Ther. 2000;7:548–55.
    1. Collard WT, Yang Y, Kwok KY, Park Y, Rice KG. Biodistribution, metabolism, and in vivo gene expression of low molecular weight glycopeptide polyethylene glycol peptide DNA co-condensates. J Pharm Sci. 2000;89:499–512.
    1. Biessen EA, Vietsch H, Rump ET, et al. Targeted delivery of oligodeoxynucleotides to parenchymal liver cells in vivo. Biochem J. 1999;340(Pt 3):783–92.
    1. Soutschek J, Akinc A, Bramlage B, et al. Therapeutic silencing of an endogenous gene by systemic administration of modified siRNAs. Nature. 2004;432:173–8.
    1. Fang D, McClintock K. Sequence-dependent stimulation of the mammalian innate immune response by synthetic siRNA. Nat Biotechnol. 2005;23:457–62.
    1. Hornung V, Guenthner-Biller M, Bourquin C, et al. Sequence-specific potent induction of IFN-alpha by short interfering RNA in plasmacytoid dendritic cells through TLR7. Nat Med. 2005;11:263–70.
    1. Isaacson M, Sureau P, Courteille G, Pattyn SR. Clinical aspects of Ebola virus disease at the Ngaliema Hospital, Kinshasa, Zaire. In: Pattyn SR, editor. Ebola virus haemorrhagic fever. New York: Elsevier/North-Holland Biomedical Press; 1978. pp. 15–20.
    1. Huggins J, Zhang ZX, Bray M. Antiviral drug therapy of filovirus infections: S-adenosylhomocysteine hydrolase inhibitors inhibit Ebola virus in vitro and in a lethal mouse model. J Infect Dis. 1999;179(Suppl 1):S240–7.
    1. Bray M, Raymond JL, Geisbert T, Baker RO. 3-deazaneplanocin A induces massively increased interferon-alpha production in Ebola virus-infected mice. Antiviral Res. 2002;55:151–9.
    1. Parren PW, Geisbert TW, Maruyama T, Jahrling PB, Burton DR. Pre- and postexposure prophylaxis of Ebola virus infection in an animal model by passive transfer of a neutralizing human antibody. J Virol. 2002;76:6408–12.
    1. Geisbert TW, Hensley LE, Jahrling PB, et al. Treatment of Ebola virus infection with a recombinant inhibitor of factor VIIa/tissue factor: a study in rhesus monkeys. Lancet. 2003;362:1953–8.
    1. Huggins JW, Zhang ZX, Davis K, Coulombe RA. Inhibition of Ebola virus by S-adenosylhomocysteine hydrolase inhibitors. Antiviral Res. 1995;26:A301.
    1. Parren PW, Geisbert TW, Geisbert J, Sullivan NJ, Jahrling PB. Program and abstracts of the VRC Symposium on Viral Hemorrhagic Fevers. Bethesda, MD: National Institute of Allergy and Infectious Diseases; 2003. Antibody activity against Ebola virus in vitro and in vivo.

Source: PubMed

3
購読する