Chimeric severe acute respiratory syndrome coronavirus (SARS-CoV) S glycoprotein and influenza matrix 1 efficiently form virus-like particles (VLPs) that protect mice against challenge with SARS-CoV

Ye V Liu, Michael J Massare, Dale L Barnard, Thomas Kort, Margret Nathan, Lei Wang, Gale Smith, Ye V Liu, Michael J Massare, Dale L Barnard, Thomas Kort, Margret Nathan, Lei Wang, Gale Smith

Abstract

SARS-CoV was the cause of the global pandemic in 2003 that infected over 8000 people in 8 months. Vaccines against SARS are still not available. We developed a novel method to produce high levels of a recombinant SARS virus-like particles (VLPs) vaccine containing the SARS spike (S) protein and the influenza M1 protein using the baculovirus insect cell expression system. These chimeric SARS VLPs have a similar size and morphology to the wild type SARS-CoV. We tested the immunogenicity and protective efficacy of purified chimeric SARS VLPs and full length SARS S protein vaccines in a mouse lethal challenge model. The SARS VLP vaccine, containing 0.8 μg of SARS S protein, completely protected mice from death when administered intramuscular (IM) or intranasal (IN) routes in the absence of an adjuvant. Likewise, the SARS VLP vaccine, containing 4 μg of S protein without adjuvant, reduced lung virus titer to below detectable level, protected mice from weight loss, and elicited a high level of neutralizing antibodies against SARS-CoV. Sf9 cell-produced full length purified SARS S protein was also an effective vaccine against SARS-CoV but only when co-administered IM with aluminum hydroxide. SARS-CoV VLPs are highly immunogenic and induce neutralizing antibodies and provide protection against lethal challenge. Sf9 cell-based VLP vaccines are a potential tool to provide protection against novel pandemic agents.

Copyright © 2011 Elsevier Ltd. All rights reserved.

Figures

Fig. 1
Fig. 1
Expression and purification of SARS S and S/M1 chimeric VLP. (A) pFastBac1 baculovirus transfer vector for SARS S/Flu M1 chimeric VLP vaccine: SARS S protein with the influenza A/Indonesia/5/2005 hemagglutinin (HA) transmembrane and carboxyl terminus (TM/CT) and the M1 influenza matrix protein 1. Each gene is under the control of its own polyhedrin promoter; (B) pFastBac1 baculovirus transfer vector for wild type full length SARS S protein with its native TM/CT; (C) purified SARS VLP vaccine, left panel: coomassie blue stain, right panel: Western blot using anti-S and anti-M1 antibodies. Positions of the 160 kDa SARS S protein and the 25 kDa M1 protein are labeled. Bands labeled # and * were identified as baculovirus gp64 protein and insect cell β tubulin protein, respectively; (D) purified native SARS S protein, left panel: coomassie blue stain, right panel: Western blot using anti-SARS S antibody. Lane M contains precision plus protein molecular weight marker (Bio-Rad, Hercules, CA). (For interpretation of the references to color in this figure legend, the reader is referred to the web version of this article.)
Fig. 2
Fig. 2
Transmission electron microscopy (EM) analysis of SARS VLPs. (A) Phosphotungstic acid negative stain of EM image of SARS VLPs; (B) immuno EM image of SARS VLPs using anti-SARS S protein primary antibody and 6 nm colloidal gold labeled secondary antibody. Black dots indicated gold labeled antibody binding to the SARS S protein. Bar: 100 nm.
Fig. 3
Fig. 3
Survival rate and lung virus titer of mice intramuscularly (IM) immunized with SARS S protein vaccine or SARS VLP vaccine after SARS-CoV lethal challenge. Mice were immunized (IM) with vehicle, 0.8 μg or 4 μg of SARS S protein or SARS VLPs, with or without aluminum hydroxide, on days 0 and 21 (n = 14 or 15). Mice were challenged with 2 LD50 of SARS-CoV strain v2163 on day 42. Five mice per group were sacrificed on day 45. Lung tissues were collected, homogenized, and assayed for virus titer. The remaining mice were monitored for survival and body weight from day 42 to day 63. (A) Survival percentage curve; (B) survival table with the mean day of death; (C) animal weight change for 21 days post challenge; (D) lung SARS-CoV titer at day 45, 3 days post challenge. Virus titer was calculated as log10 CCID50/gram lung tissue with limit of detection as 0.75 (dashed line). Results were expressed as mean titer ± SEM. Statistical significance between the vehicle and vaccine groups was determined by the student t test. **p < 0.01, ***p < 0.001.
Fig. 4
Fig. 4
Survival rate and lung virus titer of mice intranasally (IN) immunized with SARS S protein vaccine or SARS VLP vaccine after SARS-CoV lethal challenge. Mice were immunized (IN) with vehicle, 0.8 μg or 4 μg of SARS S protein or SARS VLPs, without adjuvant, on day 0 and 21 (n = 14 or 15). Mice were challenged with 2 LD50 of SARS-CoV strain v2163 on day 42. Five mice per group were sacrificed on day 45. Lung tissues were collected and assayed for virus titer. The remaining mice were monitored for survival and body weight from day 42 to day 63. (A) Survival percentage curve; (B) survival table with the mean day of death; (C) animal weight change for 21 days after challenge; (D) lung SARS-CoV titer at day 45, 3 days post challenge. Virus titer was calculated as log10 CCID50/gram lung tissue. Results were expressed as mean titer ± SEM. Statistical significance between the vehicle and vaccine groups was determined by the student t test. ***p < 0.001.
Fig. 5
Fig. 5
Neutralizing antibody titer of mice intramuscularly (IM) immunized with SARS S protein vaccine or SARS VLP vaccine. Mice were immunized (IM) with vehicle, 0.8 μg or 4 μg of SARS S protein or SARS VLPs, with or without aluminum hydroxide, on days 0 and 21. Mice were bled on day 21 (grey bar) and 42 (black bar). Sera samples were 2 fold serially diluted and assayed for neutralizing antibodies. SARS-CoV neutralizing antibody titer was defined as the inverse of the greatest dilution of serum that neutralized 100% and had no visible virus CPE. Results are expressed as the geometric mean titer ± SEM. A 1:20 dilution was the lowest dilution of serum tested. Statistical significance between vehicle and vaccine groups for day 42 sera samples was determined by the student t test. *p < 0.05.

References

    1. Enjuanes L., Dediego M.L., Alvarez E., Deming D., Sheahan T., Baric R. Vaccines to prevent severe acute respiratory syndrome coronavirus-induced disease. Virus Res. 2008;133(1):45–62.
    1. World Health Organization. Summary of probable SARS cases with onset of illness from 1 November 2002 to 31 July 2003. .
    1. Ahmad A., Krumkamp R., Reintjes R. Controlling SARS: a review on China's response compared with other SARS-affected countries. Trop Med Int Health. 2009;14(Suppl. 1):36–45.
    1. Beutels P., Jia N., Zhou Q.Y., Smith R., Cao W.C., de Vlas S.J. The economic impact of SARS in Beijing, China. Trop Med Int Health. 2009;14(Suppl. 1):85–91.
    1. Chan-Yeung M., Ooi G.C., Hui D.S., Ho P.L., Tsang K.W. Severe acute respiratory syndrome. Int J Tuberc Lung Dis. 2003;7(12):1117–1130.
    1. Drosten C., Günther S., Preiser W., van der Werf S., Brodt H.R., Becker S. Identification of a novel coronavirus in patients with severe acute respiratory syndrome. N Engl J Med. 2003;348(20):1967–1976.
    1. Masters P.S. The molecular biology of coronaviruses. Adv Virus Res. 2006;66:193–292.
    1. Li W., Moore M.J., Vasilieva N., Sui J., Wong S.K., Berne M.A. Angiotensin-converting enzyme 2 is a functional receptor for the SARS coronavirus. Nature. 2003;426(6965):450–454.
    1. Qiu M., Shi Y., Guo Z., Chen Z., He R., Chen R. Antibody responses to individual proteins of SARS coronavirus and their neutralization activities. Microbes Infect. 2005;7(5–6):882–889.
    1. Bisht H., Roberts A., Vogel L., Bukreyev A., Collins P.L., Murphy B.R. Severe acute respiratory syndrome coronavirus spike protein expressed by attenuated vaccinia virus protectively immunizes mice. Proc Natl Acad Sci USA. 2004;101(17):6641–6646.
    1. Yang Z.Y., Kong W.P., Huang Y., Roberts A., Murphy B.R., Subbarao K. A DNA vaccine induces SARS coronavirus neutralization and protective immunity in mice. Nature. 2004;428(6982):561–564.
    1. Vogel L.N., Roberts A., Paddock C.D., Genrich G.L., Lamirande E.W., Kapadia S.U. Utility of the aged BALB/c mouse model to demonstrate prevention and control strategies for severe acute respiratory syndrome coronavirus (SARS-CoV) Vaccine. 2007;25(12):2173–2179.
    1. Bai B., Lu X., Meng J., Hu Q., Mao P., Lu B. Vaccination of mice with recombinant baculovirus expressing spike or nucleocapsid protein of SARS-like coronavirus generates humoral and cellular immune responses. Mol Immunol. 2007;45:868–875.
    1. Rihtaric D., Hostnik P., Steyer A., Grom J., Toplak I. Identification of SARS-like coronaviruses in horseshoe bats (Rhinolophus hipposideros) in Slovenia. Arch Virol. 2010;155(4):507–514.
    1. Anderson L.J., Tong S. Update on SARS research and other possibly zoonotic coronaviruses. Int J Antimicrob Agents. 2010;36(November (Suppl. 1)):S21–S25. Epub 2010 (August 30)
    1. Guan Y., Zheng B.J., He Y.Q., Liu X.L., Zhuang Z.X., Cheung C.L. Isolation and characterization of viruses related to the SARS coronavirus from animals in southern China. Science. 2003;302(5643):276–278.
    1. World Health Organization. China's latest SARS outbreak has been contained, but biosafety concerns remain-Update 7, 18 May 2004. .
    1. NIAID Category A, B, and C Priority Pathogens. .
    1. Spruth M., Kistner O., Savidis-Dacho H., Hitter E., Crowe B., Gerencer M. A double-inactivated whole virus candidate SARS coronavirus vaccine stimulates neutralising and protective antibody responses. Vaccine. 2006;24(5):652–661.
    1. Marshall E., Enserink M. Medicine. Caution urged on SARS vaccines. Science. 2004;303(5660):944–946.
    1. Qin E., Shi H., Tang L., Wang C., Chang G., Ding Z. Immunogenicity and protective efficacy in monkeys of purified inactivated Vero-cell SARS vaccine. Vaccine. 2006;24(7):1028–1034.
    1. Martin J.E., Louder M.K., Holman L.A., Gordon I.J., Enama M.E., Larkin B.D. A SARS DNA vaccine induces neutralizing antibody and cellular immune responses in healthy adults in a phase I clinical trial. Vaccine. 2008;26(50):6338–6343.
    1. Bisht H., Roberts A., Vogel L., Subbarao K., Moss B. Neutralizing antibody and protective immunity to SARS coronavirus infection of mice induced by a soluble recombinant polypeptide containing an N-terminal segment of the spike glycoprotein. Virology. 2005;334(2):160–165.
    1. Zhou Z., Post P., Chubet R., Holtz K., McPherson C., Petric M. A recombinant baculovirus-expressed S glycoprotein vaccine elicits high titers of SARS-associated coronavirus (SARS-CoV) neutralizing antibodies in mice. Vaccine. 2006;24(17):3624–3631.
    1. Roy P., Noad R. Virus-like particles as a vaccine delivery system: myths and facts. Hum Vaccine. 2008;4(1):5–12.
    1. Hsieh P.K., Chang S.C., Huang C.C., Lee T.T., Hsiao C.W., Kou Y.H. Assembly of severe acute respiratory syndrome coronavirus RNA packaging signal into virus-like particles is nucleocapsid dependent. J Virol. 2005;79(22):13848–13855.
    1. Huang Y., Yang Z.Y., Kong W.P., Nabel G.J. Generation of synthetic severe acute respiratory syndrome coronavirus pseudoparticles: implications for assembly and vaccine production. J Virol. 2004;78(22):12557–12565.
    1. Mortola E., Roy P. Efficient assembly and release of SARS coronavirus-like particles by a heterologous expression system. FEBS Lett. 2004;576:174–178.
    1. Ho Y., Lin P.H., Liu C.Y., Lee S.P., Chao Y.C. Assembly of human severe acute respiratory syndrome coronavirus-like particles. Biochem Biophys Res Commun. 2004;318(4):833–838.
    1. Lu X., Chen Y., Bai B., Hu H., Tao L., Yang J. Immune responses against severe acute respiratory syndrome coronavirus induced by virus-like particles in mice. Immunology. 2007;122(4):496–502.
    1. Bai B., Hu Q., Hu H., Zhou P., Shi Z., Meng J. Virus-like particles of SARS-like coronavirus formed by membrane proteins from different origins demonstrate stimulating activity in human dendritic cells. PLoS One. 2008;3(7):e2685.
    1. Lokugamage K.G., Yoshikawa-Iwata N., Ito N., Watts D.M., Wyde P.R., Wang N. Chimeric coronavirus-like particles carrying severe acute respiratory syndrome coronavirus (SCoV) S protein protect mice against challenge with SCoV. Vaccine. 2008;26(6):797–808.
    1. Roper R.L., Rehm K.E. SARS vaccines: where are we? Expert Rev Vaccines. 2009;8(7):887–898.
    1. Bright R.A., Carter D.M., Crevar C.J., Toapanta F.R., Steckbeck J.D., Cole K.S. Cross-clade protective immune responses to influenza viruses with H5N1 HA and NA elicited by an influenza virus-like particle. PLoS One. 2008;3(1):e1501.
    1. Song H., Wittman V., Byers A., Tapia T., Zhou B., Warren W. In vitro stimulation of human influenza-specific CD8+ T cells by dendritic cells pulsed with an influenza virus-like particle (VLP) vaccine. Vaccine. 2010;28(34):5524–5532.
    1. Almeida J., Tyrell D. The morphology of three previously uncharacterized human respiratory viruses that grow in organ culture. J Gen Virol. 1967;2:175–178.
    1. Zhang Q.F., Cui J.M., Huang X.J., Lin W., Tan D.Y., Xu J.W. Morphology and morphogenesis of severe acute respiratory syndrome (SARS)-associated virus. Acta Biochim Biophys Sin. 2003;35(6):587–591.
    1. Drosten C., Lau A., Preiser W., So L., Yam L. In: SARS reference. 3rd ed. Kamps B.S., Hoffmann C., editors. Flying Publisher; 2003. p. 41. .
    1. Bright R.A., Carter D.M., Daniluk S., Toapanta F.R., Ahmad A., Gavrilov V. Influenza virus-like particles elicit broader immune responses than whole virion inactivated influenza virus or recombinant hemagglutinin. Vaccine. 2007;25(19):3871–3878.
    1. Pushko P., Kort T., Nathan M., Pearce M.B., Smith G., Tumpey T.M. Recombinant H1N1 virus-like particle vaccine elicits protective immunity in ferrets against the 2009 pandemic H1N1 influenza virus. Vaccine. 2010;28(30):4771–4776.
    1. Hall L.J., Clare S., Dougan G. Probing local innate immune responses after mucosal immunisation. J Immune Based Ther Vaccines. 2010;8(1):5.
    1. Hamming I., Timens W., Bulthuis M.L., Lely A.T., Navis G.J., van Goor H. Tissue distribution of ACE2 protein, the functional receptor for SARS coronavirus. A first step in understanding SARS pathogenesis. J Pathol. 2004;203(2):631–637.
    1. Ding Y., He L., Zhang Q., Huang Z., Che X., Hou J. Organ distribution of severe acute respiratory syndrome (SARS) associated coronavirus (SARS-CoV) in SARS patients: implications for pathogenesis and virus transmission pathways. J Pathol. 2004;203(2):622–630.
    1. Chen J., Subbarao K. The immunobiology of SARS. Annu Rev Immunol. 2007;25:443–472.
    1. Day C.W., Baric R., Cai S.X., Frieman M., Kumaki Y., Morrey J.D. A new mouse-adapted strain of SARS-CoV as a lethal model for evaluating antiviral agents in vitro and in vivo. Virology. 2009;395(2):210–222.
    1. Murti K.G., Portner A., Thoughton K., Deshpande K. Localization of proteins on viral nucleocapsids using immunoelectron microscopy. J Electron Microsc Tech. 1985;2:139–146.
    1. Feng Q., Liu Y., Qu X., Deng H., Ding M., Lau T.L. Baculovirus surface display of SARS coronavirus (SARS-CoV) spike protein and immunogenicity of the displayed protein in mice models. Immunology. 2007;122(4):496–502.
    1. Lu B., Huang Y., Huang L., Li B., Zheng Z., Chen Z. Effect of mucosal and systemic immunization with virus-like particles of severe acute respiratory syndrome coronavirus in mice. Immunology. 2010;130(2):254–261.
    1. Ludwig C., Wagner R. Virus-like particles–universal molecular toolboxes. Curr Opin Biotechnol. 2007;18(6):537–545.
    1. Grgacic E.V., Anderson D.A. Virus-like particles: passport to immune recognition. Methods. 2006;40(1):60–65.
    1. López-Macías C, Ferat-Osorio E, Tenorio-Calvo A, Isibasi A, Talavera J, Arteaga-Ruiz O, et al. Safety and immunogenicity of a virus-like particle pandemic influenza A (H1N1) 2009 vaccine in a blinded, randomized, placebo-controlled trial of adults in Mexico. Vaccine, under review.
    1. Hu M.C., Jones T., Kenney R.T., Barnard D.L., Burt D.S., Lowell G.H. Intranasal protollin-formulated recombinant SARS S-protein elicits respiratory and serum neutralizing antibodies and protection in mice. Vaccine. 2007;25(34):6334–6340.
    1. Ohkawa T., Volkman L.E., Welch M.D. Actin-based motility drives baculovirus transit to the nucleus and cell surface. J Cell Biol. 2010;190(2):187–195.

Source: PubMed

3
구독하다