Targeting inside-out phosphatidylserine as a therapeutic strategy for viral diseases

M Melina Soares, Steven W King, Philip E Thorpe, M Melina Soares, Steven W King, Philip E Thorpe

Abstract

There is a pressing need for antiviral agents that are effective against multiple classes of viruses. Broad specificity might be achieved by targeting phospholipids that are widely expressed on infected host cells or viral envelopes. We reasoned that events occurring during virus replication (for example, cell activation or preapoptotic changes) would trigger the exposure of normally intracellular anionic phospholipids on the outer surface of virus-infected cells. A chimeric antibody, bavituximab, was used to identify and target the exposed anionic phospholipids. Infection of cells with Pichinde virus (a model for Lassa fever virus, a potential bioterrorism agent) led to the exposure of anionic phospholipids. Bavituximab treatment cured overt disease in guinea pigs lethally infected with Pichinde virus. Direct clearance of infectious virus from the blood and antibody-dependent cellular cytotoxicity of virus-infected cells seemed to be the major antiviral mechanisms. Combination therapy with bavituximab and ribavirin was more effective than either drug alone. Bavituximab also bound to cells infected with multiple other viruses and rescued mice with lethal mouse cytomegalovirus infections. Targeting exposed anionic phospholipids with bavituximab seems to be safe and effective. Our study demonstrates that anionic phospholipids on infected host cells and virions may provide a new target for the generation of antiviral agents.

Figures

Figure 1. Bavituximab binds to Pichinde virus-infected…
Figure 1. Bavituximab binds to Pichinde virus-infected cells and to infectious virions
(a) Flow cytometric analysis of P388D1 cells after infection at an moi of 5. Cells were stained at 48 h with bavituximab (open histograms) or control Ig (filled histograms). Live cells were gated based on their exclusion of 7-AAD. (b) Flow cytometric analysis of expression of PS and Pichinde virus antigens over time. P388D1 cells were infected with Pichinde virus at an moi of 5 and stained at the indicated time points after infection. PS expression is detected with bavituximab (closed squares) and Pichinde virus antigen expression is detected with guinea pig Pichinde-specific antiserum (closed triangles). Points, ratio of mean fluorescence intensity (MFI) for cells stained with test antibodies to MFI of cells stained with appropriate negative control antibodies. The error on the points is ± 15%. (c) Immunofluorescence staining of uninfected Vero cells (left panels) and Pichinde virus-infected cells (right panels) with bavituximab 48 h after infection. Upper panels show cells stained with bavituximab (green). Lower panels show images merged with cytoskeleton (red). Nuclei are in blue (all panels). Control Ig did not stain and is not shown. Bar, 50 μm. (d) ELISA detection of virus binding to immobilized bavituximab or control Ig. Columns, average absorbance (n = 3); bars, s.e.m. P<0.0001 (unpaired t test) (e) Depletion of infectious Pichinde by bavituximab-coated magnetic beads. Beads coated with antibodies to Pichinde virus (PV) were used as a positive control. Columns, % depletion (n = 3); bars, s.e.m. P = 0.002 (unpaired t-test).
Figure 2. Therapeutic effect against Pichinde virus…
Figure 2. Therapeutic effect against Pichinde virus in lethally infected guinea pigs displaying overt signs of disease
(a) Kaplan-Meier survival curves of guinea pigs afterlethal infection with Pichinde virus and treatment with bavituximab or control Ig. Bavituximab or control Ig (6 mg kg−1) was administered i.p. to groups of guinea pigs (n = 8), beginning after they had developed disease signs (around day 7) and three times a week thereafter. The results are representative of those in five separate experiments. Survival in the bavituximab group was significantly superior to the control Ig group (P = 0.0036, Log-rank Mantel Cox test). (b) Virus load in tissues of treated guinea pigs 14 d after infection. Columns, average PFU per gram of tissue (n = 3); bars, s.e.m. The results are representative of two separate experiments. *P = 0.0164, **P = 0.0361, ***P = 0.0436, ****P = 0.0139, *****P = 0.0992, ******P = 0.038. (c) Additive effects of bavituximab and ribavirin treatment. Kaplan-Meier survival curves are shown for Pichinde virus-infected guinea pigs treated with bavituximab and ribavirin (n = 19), bavituximab (n = 20), ribavirin (n = 18) or control Ig (n = 20). Bavituximab or control Ig (6 mg kg−1) was administered i.p. three times per week and ribavirin (8 mg kg−1) was administered i.p. daily, beginning after the guinea pigs developed disease signs. The combination was significantly more effective than bavituximab alone (P = 0.011). All treatments were significantly different from control Ig (P<0.0001). The results are representative of two separate experiments.
Figure 3. Mechanism of anti-viral effects of…
Figure 3. Mechanism of anti-viral effects of bavituximab
(a) Lack of Pichinde virus-specific humoral response in bavituximab-treated guinea pigs. Plasma from Pichinde virus-infected animals (n = 3) was collected 7 d after onset of treatment (14 d after infection). Antibodies (IgG and IgM) to Pichinde virus were quantified by ELISA. The titer of serum from bavituximab-treated guinea pigs was not significantly different from that of control Ig-treated guinea pigs. Points, average absorbance (n = 3); bars, s.e.m. (b) Lack of Pichinde virus antigen-specific proliferative response in splenocytes from bavituximab-treated guinea pigs. Spleens from bavituximab- or control-treated Pichinde virus-infected animals (n = 3) were removed 7 d after onset of treatment (14 d after infection). Splenocytes were stimulated with Pichinde virus antigen or mock antigen and their ability to incorporate [3H]-thymidine was determined. Bavituximab treatment did not significantly increase the stimulation index (SI). Columns, average SI (n = 3); bars, s.e.m. (c) Clearance of Pichinde virus from blood of guinea pigs treated with bavituximab. Blood samples from groups of 4 guinea pigs were harvested 1 d after treatment with bavituximab or control Ig. P = 0.0145 (unpaired t-test). Columns, average PFU per ml (n = 3); bars, s.e.m. (d) Bavituximab mediates ADCC of Pichinde virus-infected guinea pig kidney fibroblasts 48 h after infection. Specific lysis was determined by quantifying 51Cr release. Bavituximab induced specific lysis of virus infected cells, P < 0.001 (unpaired t-test). Columns, average percentages (n = 3); bars, s.e.m.
Figure 4. Broad spectrum recognition of virus…
Figure 4. Broad spectrum recognition of virus infected cells and protection against cytomegalovirus infection in mice
(a) Flow cytometric analysis of virus-infected cells (right panels) and control uninfected cells (left panels). Cells were infected at an moi of 5. Cells were harvested (at 24 h for influenza A virus and VSV and at 48 h for Vaccinia virus) and stained with bavituximab (open histograms) or control Ig (filled histograms). Live intact cells were gated based on their exclusion of 7-AAD. (b) Immunofluorescence staining of mCMV-infected cells (right panels) and uninfected cells (left panels) with bavituximab. M2-10B4 cells on chamber slides were infected with mCMV at an moi of 5 and stained with bavituximab 48 h later. Upper panels show cells stained with bavituximab (green). Bavituximab is binding to externalized PS on infected cells. Lower panels show images merged with cytoskeleton (red). Nuclei are in blue (all panels). Control Ig did not stain and is not shown. Bar, 50 μm. (c) Kaplan-Meier survival curves of BALB/c mice after infection with an LD80 dose of mCMV and treatment with murine 3G4 (n = 10) or control Ig (n = 8). Treatment with 3G4 (4 mg kg−1) or control Ig was initiated 18 h after infection and administered three times a week thereafter. Survival in the 3G4 group was significantly superior to the control Ig group (P < 0.0001). The results are representative of two separate experiments.

References

    1. Williamson P, Schlegel RA. Back and forth: the regulation and function of transbilayer phospholipid movement in eukaryotic cells. Mol Membr Biol. 1994;11:199–216.
    1. Zwaal RF, Schroit AJ. Pathophysiologic implications of membrane phospholipid asymmetry in blood cells. Blood. 1997;89:1121–1132.
    1. Seigneuret M, Devaux PF. ATP-dependent asymmetric distribution of spin-labeled phospholipids in the erythrocyte membrane: relation to shape changes. Proc Natl Acad Sci U S A. 1984;81:3751–3755.
    1. Balasubramanian K, Schroit AJ. Aminophospholipid asymmetry: A matter of life and death. Annu Rev Physiol. 2003;65:701–734.
    1. Daleke DL. Regulation of transbilayer plasma membrane phospholipid asymmetry. J Lipid Res. 2003;44:233–242.
    1. Holthuis JC, Levine TP. Lipid traffic: floppy drives and a superhighway. Nat Rev Mol Cell Biol. 2005;6:209–220.
    1. Simmons A, Aluvihare V, McMichael A. Nef triggers a transcriptional program in T cells imitating single-signal T cell activation and inducing HIV virulence mediators. Immunity. 2001;14:763–777.
    1. Rawls WE, Banerjee SN, McMillan CA, Buchmeier MJ. Inhibition of Pichinde virus replication by actinomycin D. J Gen Virol. 1976;33:421–434.
    1. Lukashevich IS, Lemeshko NN, Shkolina TV. Effect of actinomycin D on the reproduction of the Machupo virus. Vopr Virusol. 1984;29:569–572.
    1. Choe W, Volsky DJ, Potash MJ. Activation of NF-kappaB by R5 and X4 human immunodeficiency virus type 1 induces macrophage inflammatory protein 1alpha and tumor necrosis factor alpha in macrophages. J Virol. 2002;76:5274–5277.
    1. Takizawa T, et al. J Gen Virol. 1993;74(Pt 11):2347–2355.
    1. Banki K, Hutter E, Gonchoroff NJ, Perl A. Molecular ordering in HIV-induced apoptosis. Oxidative stress, activation of caspases, and cell survival are regulated by transaldolase. J Biol Chem. 1998;273:11944–11953.
    1. Gautier I, Coppey J, Durieux C. Early apoptosis-related changes triggered by HSV-1 in individual neuronlike cells. Exp Cell Res. 2003;289:174–183.
    1. Mercer J, Helenius A. Vaccinia virus uses macropinocytosis and apoptotic mimicry to enter host cells. Science. 2008;320:531–535.
    1. Ran S, et al. Clin Cancer Res. 2005;11:1551–1562.
    1. Luster TA, et al. Plasma protein beta-2-glycoprotein 1 mediates interaction between the anti-tumor monoclonal antibody 3G4 and anionic phospholipids on endothelial cells. J Biol Chem. 2006;281:29863–29871.
    1. Jahrling PB, Hesse RA, Rhoderick JB, Elwell MA, Moe JB. Pathogenesis of a pichinde virus strain adapted to produce lethal infections in guinea pigs. Infect Immun. 1981;32:872–880.
    1. Huang RT, Lichtenberg B, Rick O. Involvement of annexin V in the entry of influenza viruses and role of phospholipids in infection. FEBS Lett. 1996;392:59–62.
    1. Callahan MK, et al. J Immunol. 2003;170:4840–4845.
    1. Briggs JAG, Wilk T, Fuller SD. Do lipid rafts mediate virus assembly and pseudotyping? J Gen Virol. 2003;84:757–768.
    1. Ono A, Freed EO. Plasma membrane rafts play a critical role in HIV-1 assembly and release. Proc Natl Acad Sci U S A. 2001;98:13925–13930.
    1. Scheiffele P, Rietveld A, Wilk T, Simons K. Influenza viruses select ordered lipid domains during budding from the plasma membrane. J Biol Chem. 1999;274:2038–2044.
    1. Pickl WF, Pimentel-Muinos FX, Seed B. Lipid rafts and pseudotyping. J Virol. 2001;75:7175–7183.
    1. Bavari S, et al. Lipid raft microdomains: a gateway for compartmentalized trafficking of Ebola and Marburg viruses. J Exp Med. 2002;195:593–602.
    1. Brown G, Rixon HW, Sugrue RJ. Respiratory syncytial virus assembly occurs in GM1-rich regions of the host-cell membrane and alters the cellular distribution of tyrosine phosphorylated caveolin-1. J Gen Virol. 2002;83:1841–1850.
    1. Pike LJ, Han X, Chung KN, Gross RW. Lipid rafts are enriched in arachidonic acid and plasmenylethanolamine and their composition is independent of caveolin-1 expression: a quantitative electrospray ionization/mass spectrometric analysis. Biochemistry. 2002;41:2075–2088.
    1. Bakht O, Pathak P, London E. Effect of the structure of lipids favoring disordered domain formation on the stability of cholesterol-containing ordered domains (lipid rafts): identification of multiple raft-stabilization mechanisms. Biophys J. 2007;93:4307–4318.
    1. Dillon SR, Mancini M, Rosen A, Schlissel MS. Annexin V binds to viable B cells and colocalizes with a marker of lipid rafts upon B cell receptor activation. J Immunol. 2000;164:1322–1332.
    1. Frasch SC, et al. Phospholipid flip-flop and phospholipid scramblase 1 (PLSCR1) co-localize to uropod rafts in formylated Met-Leu-Phe-stimulated neutrophils. J Biol Chem. 2004;279:17625–17633.
    1. Sutherland MR, Raynor CM, Leenknegt H, Wright JF, Pryzdial EL. Coagulation initiated on herpesviruses. Proc Natl Acad Sci U S A. 1997;94:13510–13514.
    1. Pryzdial EL, Wright JF. Prothrombinase assembly on an enveloped virus: evidence that the cytomegalovirus surface contains procoagulant phospholipid. Blood. 1994;84:3749–3757.
    1. Sanchez V, Spector DH. Virology. CMV makes a timely exit. Science. 2002;297:778–779.
    1. de Laat B, Derksen RH, van Lummel M, Pennings MT, de Groot PG. Pathogenic anti-beta2-glycoprotein I antibodies recognize domain I of beta2-glycoprotein I only after a conformational change. Blood. 2006;107:1916–1924.
    1. Godofsky EW, Shan J. Phase 1 single dose study of bavituximab, a chimeric anti-phosphatidylserine monoclonal antibody, in subjects with chronic hepatitis C. Hepatology. 2006;44:236A.
    1. Lawitz E, Godofsky E, Shan J. Multiple dose safety and pharmacokinetic study of bavituximab, a chimeric anti-phosphatidylserine monoclonal antibody, in subjects with chronic hepatitis C virus (HCV) infection. Hepatology. 2007;46:257A.
    1. Chen X, Doffek K, Sugg SL, Shilyansky J. Phosphatidylserine regulates the maturation of human dendritic cells. J Immunol. 2004;173:2985–2994.
    1. Fadok VA, et al. PAF. J Clin Invest. 1998;101:890–898.
    1. Hoffman PR, et al. Interaction between Phosphatidylserine and the Phosphatidylserine Receptor Inhibits Immune Responses In Vivo. Journal of Immunology. 2005;174:1393–1404.
    1. Ferguson TA, et al. Uptake of apoptotic antigen-coupled cells by lymphoid dendritic cells and cross-priming of CD8(+) T cells produce active immune unresponsiveness. J Immunol. 2002;168:5589–5595.
    1. Gallucci S, Lolkema M, Matzinger P. Natural adjuvants: endogenous activators of dendritic cells. Nat Med. 1999;5:1249–1255.

Source: PubMed

3
Suscribir