A randomised, double-blind, controlled vaccine efficacy trial of DNA/MVA ME-TRAP against malaria infection in Gambian adults

Vasee S Moorthy, Egeruan B Imoukhuede, Paul Milligan, Kalifa Bojang, Sheila Keating, Pauline Kaye, Margaret Pinder, Sarah C Gilbert, Gijs Walraven, Brian M Greenwood, Adrian S V Hill, Vasee S Moorthy, Egeruan B Imoukhuede, Paul Milligan, Kalifa Bojang, Sheila Keating, Pauline Kaye, Margaret Pinder, Sarah C Gilbert, Gijs Walraven, Brian M Greenwood, Adrian S V Hill

Abstract

Background: Many malaria vaccines are currently in development, although very few have been evaluated for efficacy in the field. Plasmodium falciparum multiple epitope (ME)- thrombospondin-related adhesion protein (TRAP) candidate vaccines are designed to potently induce effector T cells and so are a departure from earlier malaria vaccines evaluated in the field in terms of their mechanism of action. ME-TRAP vaccines encode a polyepitope string and the TRAP sporozoite antigen. Two vaccine vectors encoding ME-TRAP, plasmid DNA and modified vaccinia virus Ankara (MVA), when used sequentially in a prime-boost immunisation regime, induce high frequencies of effector T cells and partial protection, manifest as delay in time to parasitaemia, in a clinical challenge model.

Methods and findings: A total of 372 Gambian men aged 15-45 y were randomised to receive either DNA ME-TRAP followed by MVA ME-TRAP or rabies vaccine (control). Of these men, 296 received three doses of vaccine timed to coincide with the beginning of the transmission season (141 in the DNA/MVA group and 155 in the rabies group) and were followed up. Volunteers were given sulphadoxine/pyrimethamine 2 wk before the final vaccination. Blood smears were collected weekly for 11 wk and whenever a volunteer developed symptoms compatible with malaria during the transmission season. The primary endpoint was time to first infection with asexual P. falciparum. Analysis was per protocol. DNA ME-TRAP and MVA ME-TRAP were safe and well-tolerated. Effector T cell responses to a non-vaccine strain of TRAP were 50-fold higher postvaccination in the malaria vaccine group than in the rabies vaccine group. Vaccine efficacy, adjusted for confounding factors, was 10.3% (95% confidence interval, -22% to +34%; p = 0.49). Incidence of malaria infection decreased with increasing age and was associated with ethnicity.

Conclusions: DNA/MVA heterologous prime-boost vaccination is safe and highly immunogenic for effector T cell induction in a malaria-endemic area. But despite having produced a substantial reduction in liver-stage parasites in challenge studies of non-immune volunteers, this first generation T cell-inducing vaccine was ineffective at reducing the natural infection rate in semi-immune African adults.

Conflict of interest statement

Competing Interests: VSM, EBI, PM, KB, SK, PK, MP, SCG, GW, and BMG have declared that no competing interests exist. AVSH is a co-founder and equity holder in Oxxon Therapeutics, a company developing prime-boost therapeutic vaccines.

Figures

Figure 1. Trial Profile
Figure 1. Trial Profile
Figure 2. Kaplan-Meier Survival Curves Showing the…
Figure 2. Kaplan-Meier Survival Curves Showing the Probability of Remaining Free of P. falciparum Infection during the 11 wk of Surveillance
Week 0 of surveillance began in October 2002, 14 d after the third dose of vaccine was administered.

References

    1. Mutabingwa T, Nzila A, Mberu E, Nduati E, Winstanley P, et al. Chlorproguanil-dapsone for treatment of drug-resistant falciparum malaria in Tanzania. Lancet. 2001;358:1218–1223.
    1. Hemingway J, Field L, Vontas J. An overview of insecticide resistance. Science. 2002;298:96–97.
    1. Schofield L, Villaquiran J, Ferreira A, Schellekens H, Nussenzweig R, et al. Gamma interferon, CD8+ T cells and antibodies required for immunity to malaria sporozoites. Nature. 1987;330:664–666.
    1. Rodrigues MM, Cordey AS, Arreaza G, Corradin G, Romero P, et al. CD8+ cytolytic T cell clones derived against the Plasmodium yoelii circumsporozoite protein protect against malaria. Int Immunol. 1991;3:579–585.
    1. Weiss WR, Mellouk S, Houghten RA, Sedegah M, Kumar S, et al. Cytotoxic T cells recognize a peptide from the circumsporozoite protein on malaria-infected hepatocytes. J Exp Med. 1990;171:763–773.
    1. Good MF, Pombo D, Quakyi IA, Riley EM, Houghten RA, et al. Human T-cell recognition of the circumsporozoite protein of Plasmodium falciparum Immunodominant T-cell domains map to the polymorphic regions of the molecule. Proc Natl Acad Sci U S A. 1988;85:1199–1203.
    1. Hill AV, Allsopp CE, Kwiatkowski D, Anstey NM, Twumasi P, et al. Common west African HLA antigens are associated with protection from severe malaria [see comments] Nature. 1991;352:595–600.
    1. Hill AV, Elvin J, Willis AC, Aidoo M, Allsopp CE, et al. Molecular analysis of the association of HLA-B53 and resistance to severe malaria [see comments] Nature. 1992;360:434–439.
    1. Gilbert SC, Plebanski M, Gupta S, Morris J, Cox M, et al. Association of malaria parasite population structure, HLA, and immunological antagonism [see comments] Science. 1998;279:1173–1177.
    1. Schneider J, Gilbert SC, Blanchard TJ, Hanke T, Robson KJ, et al. Enhanced immunogenicity for CD8+ T cell induction and complete protective efficacy of malaria DNA vaccination by boosting with modified vaccinia virus Ankara. Nat Med. 1998;4:397–402.
    1. Sedegah M, Jones TR, Kaur M, Hedstrom R, Hobart P, et al. Boosting with recombinant vaccinia increases immunogenicity and protective efficacy of malaria DNA vaccine. Proc Natl Acad Sci U S A. 1998;95:7648–7653.
    1. Moorthy VS, McConkey S, Roberts M, Gothard P, Arulanantham N, et al. Safety of DNA and modified vaccinia virus Ankara vaccines against liver-stage P. falciparum malaria in non-immune volunteers. Vaccine. 2003;21:2004–2011.
    1. Ong'echa JM, Lal AA, Terlouw DJ, Ter Kuile FO, Kariuki SK, et al. Association of interferon-gamma responses to pre-erythrocytic stage vaccine candidate antigens of Plasmodium falciparum in young Kenyan children with improved hemoglobin levels: XV. Asembo Bay Cohort Project. Am J Trop Med Hyg. 2003;68:590–597.
    1. Plebanski M, Gilbert SC, Schneider J, Hannan CM, Layton G, et al. Protection from Plasmodium berghei infection by priming and boosting T cells to a single class I-restricted epitope with recombinant carriers suitable for human use. Eur J Immunol. 1998;28:4345–4355.
    1. Gilbert SC, Schneider J, Plebanski M, Hannan CM, Blanchard TJ, et al. Ty virus-like particles, DNA vaccines and Modified Vaccinia Virus Ankara; comparisons and combinations. Biol Chem. 1999;380:299–303.
    1. McShane H, Brookes R, Gilbert SC, Hill AV. Enhanced immunogenicity of CD4(+) t-cell responses and protective efficacy of a DNA-modified vaccinia virus Ankara prime-boost vaccination regimen for murine tuberculosis. Infect Immun. 2001;69:681–686.
    1. Hanke T, Samuel RV, Blanchard TJ, Neumann VC, Allen TM, et al. Effective induction of simian immunodeficiency virus-specific cytotoxic T lymphocytes in macaques by using a multiepitope gene and DNA prime-modified vaccinia virus Ankara boost vaccination regimen. J Virol. 1999;73:7524–7532.
    1. Sullivan NJ, Sanchez A, Rollin PE, Yang ZY, Nabel GJ. Development of a preventive vaccine for Ebola virus infection in primates. Nature. 2000;408:605–609.
    1. Shiver JW, Fu TM, Chen L, Casimiro DR, Davies ME, et al. Replication-incompetent adenoviral vaccine vector elicits effective anti-immunodeficiency-virus immunity. Nature. 2002;415:331–335.
    1. Amara RR, Villinger F, Altman JD, Lydy SL, O'Neil SP, et al. Control of a mucosal challenge and prevention of AIDS by a multiprotein DNA/MVA vaccine. Science. 2001;292:69–74.
    1. McConkey SJ, Reece WH, Moorthy VS, Webster D, Dunachie S, et al. Enhanced T-cell immunogenicity of plasmid DNA vaccines boosted by recombinant modified vaccinia virus Ankara in humans. Nat Med. 2003;9:729–735.
    1. Moorthy VS, Imoukhuede EB, Keating S, Pinder M, Webster D, et al. Phase 1 evaluation of 3 highly immunogenic prime-boost regimens, including a 12-month reboosting vaccination, for malaria vaccination in Gambian men. J Infect Dis. 2004;189:2213–2219.
    1. Gilbert SC, Plebanski M, Harris SJ, Allsopp CE, Thomas R, et al. A protein particle vaccine containing multiple malaria epitopes. Nat Biotechnol. 1997;15:1280–1284.
    1. Robson KJ, Hall JR, Jennings MW, Harris TJ, Marsh K, et al. A highly conserved amino-acid sequence in thrombospondin, properdin and in proteins from sporozoites and blood stages of a human malaria parasite. Nature. 1988;335:79–82.
    1. Rogers WO, Malik A, Mellouk S, Nakamura K, Rogers MD, et al. Characterization of Plasmodium falciparum sporozoite surface protein 2. Proc Natl Acad Sci U S A. 1992;89:9176–9180.
    1. Hay SI, Rogers DJ, Toomer JF, Snow RW. Annual Plasmodium falciparum entomological inoculation rates (EIR) across Africa: Literature survey, Internet access and review. Trans R Soc Trop Med Hyg. 2000;94:113–127.
    1. Clarke SE, Bogh C, Brown RC, Walraven GE, Thomas CJ, et al. Risk of malaria attacks in Gambian children is greater away from malaria vector breeding sites. Trans R Soc Trop Med Hyg. 2002;96:499–506.
    1. Moorthy VS, Pinder M, Reece WH, Watkins K, Atabani S, et al. Safety and immunogenicity of DNA/modified vaccinia virus ankara malaria vaccination in African adults. J Infect Dis. 2003;188:1239–1244.
    1. Kester KE, McKinney DA, Tornieporth N, Ockenhouse CF, Heppner DG, et al. Efficacy of recombinant circumsporozoite protein vaccine regimens against experimental Plasmodium falciparum malaria. J Infect Dis. 2001;183:640–647.
    1. Bojang KA, Milligan PJ, Pinder M, Vigneron L, Alloueche A, et al. Efficacy of RTS,S/AS02 malaria vaccine against Plasmodium falciparum infection in semi-immune adult men in The Gambia: A randomised trial. Lancet. 2001;358:1927–1934.
    1. Alonso PL, Lindsay SW, Armstrong JR, Conteh M, Hill AG, et al. The effect of insecticide-treated bed nets on mortality of Gambian children. Lancet. 1991;337:1499–1502.
    1. Rodrigues M, Nussenzweig RS, Romero P, Zavala F. The in vivo cytotoxic activity of CD8+ T cell clones correlates with their levels of expression of adhesion molecules. J Exp Med. 1992;175:895–905.
    1. Bejon P, Andrews L, Andersen RF, Dunachie S, Webster D, et al. Calculation of liver-to-blood inocula, parasite growth rates and pre-erythrocytic vaccine efficacy from serial quantitative PCR studies of malaria sporozoite challengees. J Infect Dis. 2004 In press.
    1. Prieur E, Gilbert SC, Schneider J, Moore AC, Sheu EG, et al. A Plasmodium falciparum candidate vaccine based on a six-antigen polyprotein encoded by recombinant poxviruses. Proc Natl Acad Sci U S A. 2004;101:290–295.
    1. Genton B, Al-Yaman F, Anders R, Saul A, Brown G, et al. Safety and immunogenicity of a three-component blood-stage malaria vaccine in adults living in an endemic area of Papua New Guinea. Vaccine. 2000;18:2504–2511.
    1. Coulibaly D, Diallo DA, Thera MA, Dicko A, Guindo AB, et al. Impact of preseason treatment on incidence of falciparum malaria and parasite density at a site for testing malaria vaccines in Bandiagara, Mali. Am J Trop Med Hyg. 2002;67:604–610.
    1. Modiano D, Petrarca V, Sirima BS, Nebie I, Diallo D, et al. Different response to Plasmodium falciparum malaria in west African sympatric ethnic groups. Proc Natl Acad Sci U S A. 1996;93:13206–13211.
    1. Seder RA, Hill AV. Vaccines against intracellular infections requiring cellular immunity. Nature. 2000;406:793–798.

Source: PubMed

3
購読する