Safety and Immunogenicity of a Tetravalent Dengue DNA Vaccine Administered with a Cationic Lipid-Based Adjuvant in a Phase 1 Clinical Trial

Janine R Danko, Tadeusz Kochel, Nimfa Teneza-Mora, Thomas C Luke, Kanakatte Raviprakash, Peifang Sun, Monika Simmons, James E Moon, Rafael De La Barrera, Luis Javier Martinez, Stephen J Thomas, Richard T Kenney, Larry Smith, Kevin R Porter, Janine R Danko, Tadeusz Kochel, Nimfa Teneza-Mora, Thomas C Luke, Kanakatte Raviprakash, Peifang Sun, Monika Simmons, James E Moon, Rafael De La Barrera, Luis Javier Martinez, Stephen J Thomas, Richard T Kenney, Larry Smith, Kevin R Porter

Abstract

We conducted an open label, dose escalation Phase 1 clinical trial of a tetravalent dengue DNA vaccine (TVDV) formulated in Vaxfectin® to assess safety and immunogenicity. A total of 40 dengue- and flavivirus-naive volunteers received either low-dose (1 mg) TVDV alone (N = 10, group 1), low-dose TVDV (1 mg) formulated in Vaxfectin (N = 10, group 2), or high-dose TVDV (2 mg, group 3) formulated in Vaxfectin® (N = 20). Subjects were immunized intramuscularly with three doses on a 0-, 30-, 90-day schedule and monitored. Blood samples were obtained after each immunization and various time points thereafter to assess anti-dengue antibody and interferon gamma (IFNγ) T-cell immune responses. The most common adverse events (AEs) across all groups included mild to moderate pain and tenderness at the injection site, which typically resolved within 7 days. Common solicited signs and symptoms included fatigue (42.5%), headache (45%), and myalgias (47.5%). There were no serious AEs related to the vaccine or study procedures. No anti-dengue antibody responses were detected in group 1 subjects who received all three immunizations. There were minimal enzyme-linked immunosorbent assay and neutralizing antibody responses among groups 2 and 3 subjects who completed the immunization schedule. By contrast, IFNγ T-cell responses, regardless of serotype specificity, occurred in 70%, 50%, and 79% of subjects in groups 1, 2, and 3, respectively. The largest IFNγ T-cell responses were among group 3 subjects. We conclude that TVDV was safe and well-tolerated and elicited predominately anti-dengue T-cell IFNγ responses in a dose-related fashion.

Trial registration: ClinicalTrials.gov NCT00290147.

Figures

Figure 1.
Figure 1.
Study subject disposition. Blocks indicate the total number of subjects in each group and the disposition of such subjects during the study. * Lost to follow-up because of relocation, $ Consent withdrawn (N = 1) after first dose but safety data available.
Figure 2.
Figure 2.
Interferon gamma T-cell enzyme-linked immunospot (ELISPOT) results. Each graph represents ELISPOT results for subjects who received either 1 mg total of TVDV alone (A, group 1), 1 mg TVDV with Vaxfectin (B, group 2) or 2 mg with Vaxfectin (C, group 3). The individual volunteer numbers are listed on the x axis, group by day. The y axis shows ELISPOT result in number of spots/105 total cells. The z axis shows the serotype-specific peptide pool used to stimulate the cells. Each bar represents the average ELISPOT result for the indicated volunteer for the indicated peptide pool.

References

    1. Bhatt S, et al. 2013. The global distribution and burden of dengue. Nature 496: 504–507.
    1. Beckett CG, et al. 2011. Evaluation of a prototype dengue-1 DNA vaccine in a phase 1 clinical trial. Vaccine 29: 960–968.
    1. Hartikka J, et al. 2001. Vaxfectin enhances the humoral immune response to plasmid DNA-encoded antigens. Vaccine 19: 1911–1923.
    1. Hartikka J, Bozoukova V, Yang CK, Ye M, Rusalov D, Shlapobersky M, Vilalta A, Wei Q, Rolland A, Smith LR, 2009. Vaxfectin, a cationic lipid-based adjuvant for protein-based influenza vaccines. Vaccine 27: 6399–6403.
    1. Margalith M, Vilalta A, 2006. Sustained protective rabies neutralizing antibody titers after administration of cationic lipid-formulated pDNA vaccine. Genet Vaccines Ther 4: 2.
    1. Sullivan SM, Doukas J, Hartikka J, Smith L, Rolland A, 2010. Vaxfectin: a versatile adjuvant for plasmid DNA- and protein-based vaccines. Expert Opin Drug Deliv 7: 1433–1446.
    1. Porter KR, Ewing D, Chen L, Wu SJ, Hayes CG, Ferrari M, Teneza-Mora N, Raviprakash K, 2012. Immunogenicity and protective efficacy of a vaxfectin-adjuvanted tetravalent dengue DNA vaccine. Vaccine 30: 336–341.
    1. Smith LR, et al. 2010. Phase 1 clinical trials of the safety and immunogenicity of adjuvanted plasmid DNA vaccines encoding influenza A virus H5 hemagglutinin. Vaccine 28: 2565–2572.
    1. Simmons M, Porter KR, Hayes CG, Vaughn DW, Putnak R, 2006. Characterization of antibody responses to combinations of a dengue virus type 2 DNA vaccine and two dengue virus type 2 protein vaccines in rhesus macaques. J Virol 80: 9577–9585.
    1. Putnak JR, et al. 2008. Comparative evaluation of three assays for measurement of dengue virus neutralizing antibodies. Am J Trop Med Hyg 79: 115–122.
    1. Thomas SJ, 2014. Developing a dengue vaccine: progress and future challenges. Ann N Y Acad Sci 1323: 140–159.
    1. Sabchareon A, et al. 2012. Protective efficacy of the recombinant, live-attenuated, CYD tetravalent dengue vaccine in Thai schoolchildren: a randomised, controlled phase 2b trial. Lancet 380: 1559–1567.
    1. Villar L, et al. 2015. Efficacy of a tetravalent dengue vaccine in children in Latin America. N Engl J Med 372: 113–123.
    1. Capeding MR, et al. 2014. Clinical efficacy and safety of a novel tetravalent dengue vaccine in healthy children in Asia: a phase 3, randomised, observer-masked, placebo-controlled trial. Lancet 384: 1358–1365.
    1. Sirivichayakul C, Sabchareon A, Limkittikul K, Yoksan S, 2014. Plaque reduction neutralization antibody test does not accurately predict protection against dengue infection in Ratchaburi cohort, Thailand. Virol J 11: 48.
    1. Rothman AL, Kanesa-thasan N, West K, Janus J, Saluzzo JF, Ennis FA, 2001. Induction of T lymphocyte responses to dengue virus by a candidate tetravalent live attenuated dengue virus vaccine. Vaccine 19: 4694–4699.
    1. Balas C, Kennel A, Deauvieau F, Sodoyer R, Arnaud-Barbe N, Lang J, Guy B, 2011. Different innate signatures induced in human monocyte-derived dendritic cells by wild-type dengue 3 virus, attenuated but reactogenic dengue 3 vaccine virus, or attenuated nonreactogenic dengue 1–4 vaccine virus strains. J Infect Dis 203: 103–108.
    1. Guy B, et al. 2008. Cell-mediated immunity induced by chimeric tetravalent dengue vaccine in naive or flavivirus-primed subjects. Vaccine 26: 5712–5721.
    1. Thakur A, Pedersen LE, Jungersen G, 2012. Immune markers and correlates of protection for vaccine induced immune responses. Vaccine 30: 4907–4920.
    1. Halstead SB, 2013. Identifying protective dengue vaccines: guide to mastering an empirical process. Vaccine 31: 4501–4507.
    1. Li J, Valentin A, Beach RK, Alicea C, Felber BK, Pavlakis GN, 2015. DNA is an efficient booster of dendritic cell-based vaccine. Hum Vaccin Immunother 11: 1927–1935.
    1. Lillie PJ, et al. 2012. Preliminary assessment of the efficacy of a T-cell-based influenza vaccine, MVA-NP+M1, in humans. Clin Infect Dis 55: 19–25.
    1. Vandermeulen G, Vanvarenberg K, De Beuckelaer A, De Koker S, Lambricht L, Uyttenhove C, Reschner A, Vanderplasschen A, Grooten J, Preat V, 2015. The site of administration influences both the type and the magnitude of the immune response induced by DNA vaccine electroporation. Vaccine 33: 3179–3185.

Source: PubMed

3
Iratkozz fel