A SARS DNA vaccine induces neutralizing antibody and cellular immune responses in healthy adults in a Phase I clinical trial

Julie E Martin, Mark K Louder, LaSonji A Holman, Ingelise J Gordon, Mary E Enama, Brenda D Larkin, Charla A Andrews, Leatrice Vogel, Richard A Koup, Mario Roederer, Robert T Bailer, Phillip L Gomez, Martha Nason, John R Mascola, Gary J Nabel, Barney S Graham, VRC 301 Study Team, Margaret McCluskey, Sarah Hubka, Steve Rucker, Laura Novik, Pamela Edmonds, LaChonne Stanford, Woody Dubois, Tiffany Alley, Erica Eaton, Sandra Sitar, Ericka Thompson, Andrew Catanzaro, Joseph Casazza, Laurence Lemiale, Rebecca Sheets, Julie E Martin, Mark K Louder, LaSonji A Holman, Ingelise J Gordon, Mary E Enama, Brenda D Larkin, Charla A Andrews, Leatrice Vogel, Richard A Koup, Mario Roederer, Robert T Bailer, Phillip L Gomez, Martha Nason, John R Mascola, Gary J Nabel, Barney S Graham, VRC 301 Study Team, Margaret McCluskey, Sarah Hubka, Steve Rucker, Laura Novik, Pamela Edmonds, LaChonne Stanford, Woody Dubois, Tiffany Alley, Erica Eaton, Sandra Sitar, Ericka Thompson, Andrew Catanzaro, Joseph Casazza, Laurence Lemiale, Rebecca Sheets

Abstract

Background: The severe acute respiratory syndrome (SARS) virus is a member of the Coronaviridae (CoV) family that first appeared in the Guangdong Province of China in 2002 and was recognized as an emerging infectious disease in March 2003. Over 8000 cases and 900 deaths occurred during the epidemic. We report the safety and immunogenicity of a SARS DNA vaccine in a Phase I human study.

Methods: A single-plasmid DNA vaccine encoding the Spike (S) glycoprotein was evaluated in 10 healthy adults. Nine subjects completed the 3 dose vaccination schedule and were evaluated for vaccine safety and immune responses. Immune response was assessed by intracellular cytokine staining (ICS), ELISpot, ELISA, and neutralization assays.

Results: The vaccine was well tolerated. SARS-CoV-specific antibody was detected by ELISA in 8 of 10 subjects and neutralizing antibody was detected in all subjects who received 3 doses of vaccine. SARS-CoV-specific CD4+ T-cell responses were detected in all vaccinees, and CD8+ T-cell responses in approximately 20% of individuals.

Conclusions: The VRC SARS DNA vaccine was well tolerated and produced cellular immune responses and neutralizing antibody in healthy adults.

Figures

Fig. 1
Fig. 1
Magnitude and frequency of neutralizing antibody response. Individual subjects are designated by letters A–J, sorted by ascending age on the x-axis. The IC80 (inhibitory concentration 80%) reciprocal titer is represented on the y-axis. The time course of the study is shown for each subject: week 0 (yellow bars), week 8 (green bars), week 12 (blue bars), and week 32 (orange bars). Vaccinations were administered at weeks 0, 4, and 8. Subject “G” received 2 of 3 vaccinations.
Fig. 2
Fig. 2
Magnitude and frequency of CD4 and CD8 T-cell responses by ICS and ELISpot analysis at specific timepoints throughout the study. Magnitude of response is represented on the upper graph, percent positive CD4 (red bars) or CD8 cells (green bars) for ICS or spot-forming colonies (SFC) for ELISpot (blue bars). The horizontal black bars represent the mean. A sample was considered positive if it was above the thresholds indicated by the dashed lines. Separate positivity criteria for CD4 and CD8 ICS and ELISpot were developed and validated for overlapping peptide-based stimulations. The frequency of response is represented by percent responders on the lower graph. Weeks after enrollment is shown on the x-axis, applicable for upper and lower graphs. Vaccinations were administered at weeks 0, 4, and 8.

References

    1. Peiris J.S., Guan Y., Yuen K.Y. Severe acute respiratory syndrome. Nat Med. 2004;10(12 Suppl.):S88–S97.
    1. Cumulative number of reported probable cases of SARS. 2003.
    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. Holmes K.V. SARS-associated coronavirus. N Engl J Med. 2003;348(20):1948–1951.
    1. Navas-Martin S.R., Weiss S. Coronavirus replication and pathogenesis: implications for the recent outbreak of severe acute respiratory syndrome (SARS), and the challenge for vaccine development. J Neurovirol. 2004;10(2):75–85.
    1. Wong S.K., Li W., Moore M.J., Choe H., Farzan M. A 193-amino acid fragment of the SARS coronavirus S protein efficiently binds angiotensin-converting enzyme 2. J Biol Chem. 2004;279(5):3197–3201.
    1. Gallagher T.M. Murine coronavirus spike glycoprotein. Receptor binding and membrane fusion activities. Adv Exp Med Biol. 2001;494:183–192.
    1. Yang Z.Y., Huang Y., Ganesh L., Leung K., Kong W.P., Schwartz O. pH-dependent entry of severe acute respiratory syndrome coronavirus is mediated by the spike glycoprotein and enhanced by dendritic cell transfer through DC-SIGN. J Virol. 2004;78(11):5642–5650.
    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. Nie Y., Wang G., Shi X., Zhang H., Qiu Y., He Z. Neutralizing antibodies in patients with severe acute respiratory syndrome-associated coronavirus infection. J Infect Dis. 2004;190(6):1119–1126.
    1. Marra M.A., Jones S.J., Astell C.R., Holt R.A., Brooks-Wilson A., Butterfield Y.S. The genome sequence of the SARS-associated coronavirus. Science. 2003;300(5624):1399–1404.
    1. Rota P.A., Oberste M.S., Monroe S.S., Nix W.A., Campagnoli R., Icenogle J.P. Characterization of a novel coronavirus associated with severe acute respiratory syndrome. Science. 2003;300(5624):1394–1399.
    1. Lin J.T., Zhang J.S., Su N., Xu J.G., Wang N., Chen J.T. Safety and immunogenicity from a phase I trial of inactivated severe acute respiratory syndrome coronavirus vaccine. Antivir Ther. 2007;12(7):1107–1113.
    1. Catanzaro A.T., Roederer M., Koup R.A., Bailer R.T., Enama M.E., Nason M.C. Phase I clinical evaluation of a six-plasmid multiclade HIV-1 DNA candidate vaccine. Vaccine. 2007;25(20):4085–4092.
    1. Graham B.S., Koup R.A., Roederer M., Bailer R.T., Enama M.E., Moodie Z. Phase 1 safety and immunogenicity evaluation of a multiclade HIV-1 DNA candidate vaccine. J Infect Dis. 2006;194(12):1650–1660.
    1. Martin J.E., Sullivan N.J., Enama M.E., Gordon I.J., Roederer M., Koup R.A. A DNA vaccine for Ebola virus is safe and immunogenic in a phase I clinical trial. Clin Vaccine Immunol. 2006;13(11):1267–1277.
    1. Barouch D.H., Yang Z.Y., Kong W.P., Korioth-Schmitz B., Sumida S.M., Truitt D.M. A human T-cell leukemia virus type 1 regulatory element enhances the immunogenicity of human immunodeficiency virus type 1 DNA vaccines in mice and nonhuman primates. J Virol. 2005;79(14):8828–8834.
    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. Shu Y., Winfrey S., Yang Z.Y., Xu L., Rao S.S., Srivastava I. Efficient protein boosting after plasmid DNA or recombinant adenovirus immunization with HIV-1 vaccine constructs. Vaccine. 2007;25(8):1398–1408.
    1. Subbarao K., McAuliffe J., Vogel L., Fahle G., Fischer S., Tatti K. Prior infection and passive transfer of neutralizing antibody prevent replication of severe acute respiratory syndrome coronavirus in the respiratory tract of mice. J Virol. 2004;78(7):3572–3577.
    1. Martin J.E., Pierson T.C., Hubka S., Rucker S., Gordon I.J., Enama M.E. A West Nile virus DNA vaccine induces neutralizing antibody in healthy adults during a phase 1 clinical trial. J Infect Dis. 2007;196(12):1732–1740.
    1. Buchholz U.J., Bukreyev A., Yang L., Lamirande E.W., Murphy B.R., Subbarao K. Contributions of the structural proteins of severe acute respiratory syndrome coronavirus to protective immunity. Proc Natl Acad Sci USA. 2004;101(26):9804–9809.
    1. Lu L., Manopo I., Leung B.P., Chng H.H., Ling A.E., Chee L.L. Immunological characterization of the spike protein of the severe acute respiratory syndrome coronavirus. J Clin Microbiol. 2004;42(4):1570–1576.
    1. Chen J., Subbarao K. The immunobiology of SARS*. Annu Rev Immunol. 2007;25:443–472.
    1. Yang L.T., Peng H., Zhu Z.L., Li G., Huang Z.T., Zhao Z.X. Long-lived effector/central memory T-cell responses to severe acute respiratory syndrome coronavirus (SARS-CoV) S antigen in recovered SARS patients. Clin Immunol. 2006;120(2):171–178.
    1. Peng H., Yang L.T., Li J., Lu Z.Q., Wang L.Y., Koup R.A. Human memory T cell responses to SARS-CoV E protein. Microbes Infect. 2006;8(9–10):2424–2431.
    1. Peng H., Yang L.T., Wang L.Y., Li J., Huang J., Lu Z.Q. Long-lived memory T lymphocyte responses against SARS coronavirus nucleocapsid protein in SARS-recovered patients. Virology. 2006;351(2):466–475.
    1. Yang L., Peng H., Zhu Z., Li G., Huang Z., Zhao Z. Persistent memory CD4+ and CD8+ T-cell responses in recovered severe acute respiratory syndrome (SARS) patients to SARS coronavirus M antigen. J Gen Virol. 2007;88(Pt 10):2740–2748.

Source: PubMed

3
구독하다