A phase 2b randomised trial of the candidate malaria vaccines FP9 ME-TRAP and MVA ME-TRAP among children in Kenya

Philip Bejon, Jedidah Mwacharo, Oscar Kai, Tabitha Mwangi, Paul Milligan, Stephen Todryk, Sheila Keating, Trudie Lang, Brett Lowe, Caroline Gikonyo, Catherine Molyneux, Greg Fegan, Sarah C Gilbert, Norbert Peshu, Kevin Marsh, Adrian V S Hill, Philip Bejon, Jedidah Mwacharo, Oscar Kai, Tabitha Mwangi, Paul Milligan, Stephen Todryk, Sheila Keating, Trudie Lang, Brett Lowe, Caroline Gikonyo, Catherine Molyneux, Greg Fegan, Sarah C Gilbert, Norbert Peshu, Kevin Marsh, Adrian V S Hill

Abstract

Objective: The objective was to measure the efficacy of the vaccination regimen FFM ME-TRAP in preventing episodes of clinical malaria among children in a malaria endemic area. FFM ME-TRAP is sequential immunisation with two attenuated poxvirus vectors (FP9 and modified vaccinia virus Ankara), which both deliver the pre-erythrocytic malaria antigen construct multiple epitope-thrombospondin-related adhesion protein (ME-TRAP).

Design: The trial was randomised and double-blinded.

Setting: The setting was a rural, malaria-endemic area of coastal Kenya.

Participants: We vaccinated 405 healthy 1- to 6-year-old children.

Interventions: Participants were randomised to vaccination with either FFM ME-TRAP or control (rabies vaccine).

Outcome measures: Following antimalarial drug treatment children were seen weekly and whenever they were unwell during nine months of monitoring. The axillary temperature was measured, and blood films taken when febrile. The primary analysis was time to a parasitaemia of over 2,500 parasites/mul.

Results: The regime was moderately immunogenic, but the magnitude of T cell responses was lower than in previous studies. In intention to treat (ITT) analysis, time to first episode was shorter in the FFM ME-TRAP group. The cumulative incidence of febrile malaria was 52/190 (27%) for FFM ME-TRAP and 40/197 (20%) among controls (hazard ratio = 1.52). This was not statistically significant (95% confidence interval [CI] 1.0-2.3; p = 0.14 by log-rank). A group of 346 children were vaccinated according to protocol (ATP). Among these children, the hazard ratio was 1.3 (95% CI 0.8-2.1; p = 0.55 by log-rank). When multiple malaria episodes were included in the analyses, the incidence rate ratios were 1.6 (95% CI 1.1-2.3); p = 0.017 for ITT, and 1.4 (95% CI 0.9-2.1); p = 0.16 for ATP. Haemoglobin and parasitaemia in cross-sectional surveys at 3 and 9 mo did not differ by treatment group. Among children vaccinated with FFM ME-TRAP, there was no correlation between immunogenicity and malaria incidence.

Conclusions: No protection was induced against febrile malaria by this vaccine regimen. Future field studies will require vaccinations with stronger immunogenicity in children living in malarious areas.

Conflict of interest statement

Competing Interests: AVSH is cofounder of and an equity holder in Oxxon Therapeutics, a company developing prime-boost therapeutic vaccines. No other authors have any conflicts of interest.

Figures

Figure 1. Trial Profile
Figure 1. Trial Profile
After screening for eligibility, parents were invited to bring their children back to the dispensary for immunisation. Children were randomised on attending for vaccination. Of the 17 children who attended for the first, but not the final, vaccination, two had moved out of the area, and parents of the remaining 15 chose not to reattend. No severe adverse events were identified in these children. Before 9 mo monitoring was complete, eight children had moved out of the area.
Figure 2. Primary Analysis of Efficacy
Figure 2. Primary Analysis of Efficacy
The probability of remaining free of clinical malaria is plotted over the 9 mo of monitoring (the primary analysis). Numbers of children at risk are given below the Kaplan-Meier plots for ITT (top; p = 0.55) and ATP (bottom; p = 0.14). Both plots use an endpoint of over 2,500 parasites/μl and fever.
Figure 3. Secondary Analyses for Efficacy
Figure 3. Secondary Analyses for Efficacy
The results from secondary analyses of malaria episodes are shown. These multiple analyses were generated by varying the case definition and the statistical methodology, and are shown for both ATP and ITT. The more rigorous case definition (over 2,500 parasites/μl and fever) is the first in each group of comparisons. Cox regression was used to estimate hazard ratios for time to first episode (A) and Poisson regression was used to estimate incidence rate ratios for the frequency of episodes (B). Models were adjusted for age, village, and ITN use. The two groups of three points to the right of each panel show the hazard ratios and incidence rate ratios for subgroups FFM ME-TRAP-vaccinated children. Participants were divided into tertiles based on either ex vivo or cultured ELISPOT responses. Hazard ratios and incidence rate ratios for each tertile relative to the control group are shown, using a case definition of parasitaemia over 2,500/μl and fever.
Figure 4. Immunogenicity
Figure 4. Immunogenicity
T cell responses to vaccination identified by both ex vivo and cultured ELISPOT are displayed over time. Median, 25th, and 75th quartile, 5th and 95th quartile, and outlying results are given by box and whisker plots. Data were available for 400 children at screening (i.e., prevaccination), for 379 children 7 d after the last vaccination, for 345 at 3 mo, and for 304 at 9 mo. T cell numbers were similar at baseline for ex vivo (p = 0.4) and cultured (p = 0.91) responses. Ex vivo responses were higher among FFM vaccinees at 1 wk (p < 0.001) and 9 mo (p = 0.015), as were cultured responses. Ex vivo responses did not significantly differ at 3 mo (p = 0.27), but cultured responses did (p < 0.001).

References

    1. Hay SI, Guerra CA, Tatem AJ, Noor AM, Snow RW. The global distribution and population at risk of malaria: Past, present, and future. Lancet Infect Dis. 2004;4:327–336.
    1. Doolan DL, Hoffman SL. The complexity of protective immunity against liver-stage malaria. J Immunol. 2000;165:1453–1462.
    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. Nature. 1991;352:595–600.
    1. Herrington D, Davis J, Nardin E, Beier M, Cortese J, et al. Successful immunization of humans with irradiated malaria sporozoites: Humoral and cellular responses of the protected individuals. Am J Trop Med Hyg. 1991;45:539–547.
    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. Moore AC, Hill AV. Progress in DNA-based heterologous prime-boost immunization strategies for malaria. Immunol Rev. 2004;199:126–143.
    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. Webster DP, Dunachie S, Vuola JM, Berthoud T, Keating S, et al. Enhanced T cell-mediated protection against malaria in human challenges by using the recombinant poxviruses FP9 and modified vaccinia virus Ankara. Proc Natl Acad Sci U S A. 2005;102:4836–4841.
    1. Bejon P, Andrews L, Andersen RF, Dunachie S, Webster D, et al. Calculation of liver-to-blood inocula, parasite growth rates, and preerythrocytic vaccine efficacy, from serial quantitative polymerase chain reaction studies of volunteers challenged with malaria sporozoites. J Infect Dis. 2005;191:619–626.
    1. Moorthy VS, Imoukhuede EB, Milligan P, Bojang K, Keating S, et al. A randomised, double-blind, controlled vaccine efficacy trial of DNA/MVA ME-TRAP against malaria infection in Gambian adults. Plos Med. 2004;1:e33.
    1. Sagara I, Sangare D, Dolo G, Guindo A, Sissoko M, et al. A high malaria reinfection rate in children and young adults living under a low entomological inoculation rate in a periurban area of Bamako, Mali. Am J Trop Med Hyg. 2002;66:310–313.
    1. Owusu-Agyei S, Koram KA, Baird JK, Utz GC, Binka FN, et al. Incidence of symptomatic and asymptomatic Plasmodium falciparum infection following curative therapy in adult residents of northern Ghana. Am J Trop Med Hyg. 2001;65:197–203.
    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. Anderson RJ, Hannan CM, Gilbert SC, Laidlaw SM, Sheu EG, et al. Enhanced CD8+ T cell immune responses and protection elicited against Plasmodium berghei malaria by prime boost immunization regimens using a novel attenuated fowlpox virus. J Immunol. 2004;172:3094–3100.
    1. Godkin AJ, Thomas HC, Openshaw PJ. Evolution of epitope-specific memory CD4+ T cells after clearance of hepatitis C virus. J Immunol. 2002;169:2210–2214.
    1. Flanagan KL, Lee EA, Gravenor MB, Reece WH, Urban BC, et al. Unique T cell effector functions elicited by Plasmodium falciparum epitopes in malaria-exposed Africans tested by three T cell assays. J Immunol. 2001;167:4729–4737.
    1. Keating SM, Bejon P, Berthoud T, Vuola JM, Todryk S, et al. Durable human memory T cells quantifiable by cultured enzyme-linked immunospot assays are induced by heterologous prime boost immunization and correlate with protection against malaria. J Immunol. 2005;175:5675–5680.
    1. Sun P, Schwenk R, White K, Stoute JA, Cohen J, et al. Protective immunity induced with malaria vaccine, RTS,S, is linked to Plasmodium falciparum circumsporozoite protein-specific CD4+ and CD8+ T cells producing IFN-gamma. J Immunol. 2003;171:6961–6967.
    1. Bejon P, Kai OK, Mwacharo J, Keating S, Lang T, et al. Alternating vector immunizations encoding pre-erythrocytic malaria antigens enhance memory responses in a malaria endemic area. Eur J Immunol. 2006;36:2264–2272.
    1. Bejon P, Mwacharo J, Kai OK, Todryk S, Keating S, et al. Immunogenicity of the candidate malaria vaccines FP9 and modified vaccinia virus Ankara encoding the pre-erythrocytic antigen ME-TRAP in 1–6 year old children in a malaria endemic area. Vaccine. 2006;24:4709–4715.
    1. Bejon P, Peshu N, Gilbert SC, Lowe BS, Molyneux CS, et al. Safety profile of the viral vectors of attenuated fowlpox strain FP9 and modified vaccinia virus Ankara recombinant for either of 2 preerythrocytic malaria antigens, ME-TRAP or the circumsporozoite protein, in children and adults in Kenya. Clin Infect Dis. 2006;42:1102–1110.
    1. Mwangi TW, Ross A, Snow RW, Marsh K. Case definitions of clinical malaria under different transmission conditions in Kilifi District, Kenya. J Infect Dis. 2005;191:1932–1939.
    1. Mbogo CM, Mwangangi JM, Nzovu J, Gu W, Yan G, et al. Spatial and temporal heterogeneity of Anopheles mosquitoes and Plasmodium falciparum transmission along the Kenyan coast. Am J Trop Med Hyg. 2003;68:734–742.
    1. Armstrong Schellenberg JR, Smith T, Alonso P, Hayes R. What is clinical malaria? Finding case definitions for field research in highly endemic areas. Parasitology Today. 1994;10:439–441.
    1. Scott JA, Mlacha Z, Nyiro J, Njenga S, Lewa P, et al. Diagnosis of invasive pneumococcal disease among children in Kenya with enzyme-linked immunosorbent assay for immunoglobulin G antibodies to pneumococcal surface adhesin A. Clin Diagn Lab Immunol. 2005;12:1195–1201.
    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, Gupta S, Morris J, Cox M, et al. Association of malaria parasite population structure, HLA, and immunological antagonism. Science. 1998;279:1173–1177.
    1. Walther M, Tongren JE, Andrews L, Korbel D, King E, et al. Upregulation of TGF-beta, FOXP3, and CD4+CD25+ regulatory T cells correlates with more rapid parasite growth in human malaria infection. Immunity. 2005;23:287–296.
    1. Flanagan KL, Plebanski M, Odhiambo K, Sheu E, Mwangi T, et al. Cellular reactivity to the P. falciparum protein trap in adult Kenyans: Novel epitopes, complex cytokine patterns, and the impact of natural antigenic variation. Am J Trop Med Hyg. 2006;74:367–375.
    1. Flanagan KL, Mwangi T, Plebanski M, Odhiambo K, Ross A, et al. Ex vivo interferon-gamma immune response to thrombospondin-related adhesive protein in coastal Kenyans: Longevity and risk of Plasmodium falciparum infection. Am J Trop Med Hyg. 2003;68:421–430.
    1. Wilson NS, Behrens GM, Lundie RJ, Smith CM, Waithman J, et al. Systemic activation of dendritic cells by Toll-like receptor ligands or malaria infection impairs cross-presentation and antiviral immunity. Nat Immunol. 2006;7:165–172.
    1. Brustoski K, Moller U, Kramer M, Hartgers FC, Kremsner PG, et al. Reduced cord blood immune effector-cell responsiveness mediated by CD4+ cells induced in utero as a consequence of placental Plasmodium falciparum infection. J Infect Dis. 2006;193:146–154.
    1. Smith T, Hurt N, Teuscher T, Tanner M. Is fever a good sign for clinical malaria in surveys of endemic communities? Am J Trop Med Hyg. 1995;52:306–310.
    1. Einterz EM, Bates ME. Fever in Africa: Do patients know when they are hot? Lancet. 1997;350:781.
    1. Alonso PL, Sacarlal J, Aponte JJ, Leach A, Macete E, et al. Efficacy of the RTS,S/AS02A vaccine against Plasmodium falciparum infection and disease in young African children: Randomised controlled trial. Lancet. 2004;364:1411–1420.
    1. Stoute JA, Slaoui M, Heppner DG, Momin P, Kester KE, et al. A preliminary evaluation of a recombinant circumsporozoite protein vaccine against Plasmodium falciparum malaria. RTS,S Malaria Vaccine Evaluation Group. N Engl J Med. 1997;336:86–91.

Source: PubMed

3
Iratkozz fel