Evidence of blood stage efficacy with a virosomal malaria vaccine in a phase IIa clinical trial

Fiona M Thompson, David W Porter, Shinji L Okitsu, Nicole Westerfeld, Denise Vogel, Stephen Todryk, Ian Poulton, Simon Correa, Claire Hutchings, Tamara Berthoud, Susanna Dunachie, Laura Andrews, Jack L Williams, Robert Sinden, Sarah C Gilbert, Gerd Pluschke, Rinaldo Zurbriggen, Adrian V S Hill, Fiona M Thompson, David W Porter, Shinji L Okitsu, Nicole Westerfeld, Denise Vogel, Stephen Todryk, Ian Poulton, Simon Correa, Claire Hutchings, Tamara Berthoud, Susanna Dunachie, Laura Andrews, Jack L Williams, Robert Sinden, Sarah C Gilbert, Gerd Pluschke, Rinaldo Zurbriggen, Adrian V S Hill

Abstract

Background: Previous research indicates that a combination vaccine targeting different stages of the malaria life cycle is likely to provide the most effective malaria vaccine. This trial was the first to combine two existing vaccination strategies to produce a vaccine that induces immune responses to both the pre-erythrocytic and blood stages of the P. falciparum life cycle.

Methods: This was a Phase I/IIa study of a new combination malaria vaccine FFM ME-TRAP+PEV3A. PEV3A includes peptides from both the pre-erythrocytic circumsporozoite protein and the blood-stage antigen AMA-1. This study was conducted at the Centre for Clinical Vaccinology and Tropical Medicine, University of Oxford, Oxford, UK. The participants were healthy, malaria naïve volunteers, from Oxford. The interventions were vaccination with PEV3A alone, or PEV3A+FFM ME-TRAP. The main outcome measure was protection from malaria in a sporozoite challenge model. Other outcomes included measures of parasite specific immune responses induced by either vaccine; and safety, assessed by collection of adverse event data.

Results: We observed evidence of blood stage immunity in PEV3A vaccinated volunteers, but no volunteers were completely protected from malaria. PEV3A induced high antibody titres, and antibodies bound parasites in immunofluorescence assays. Moreover, we observed boosting of the vaccine-induced immune response by sporozoite challenge. Immune responses induced by FFM ME-TRAP were unexpectedly low. The vaccines were safe, with comparable side effect profiles to previous trials. Although there was no sterile protection two major observations support an effect of the vaccine-induced response on blood stage parasites: (i) Lower rates of parasite growth were observed in volunteers vaccinated with PEV3A compared to unvaccinated controls (p = 0.012), and this was reflected in the PCR results from PEV3A vaccinated volunteers. These showed early control of parasitaemia by some volunteers in this group. One volunteer, who received PEV3A alone, was diagnosed very late, on day 20 compared to an average of 11.8 days in unvaccinated controls. (ii). Morphologically abnormal parasites were present in the blood of all (n = 24) PEV3A vaccinated volunteers, and in only 2/6 controls (p = 0.001). We describe evidence of vaccine-induced blood stage efficacy for the first time in a sporozoite challenge study.

Trial registration: ClinicalTrials.gov NCT00408668.

Conflict of interest statement

Competing Interests: Rinaldo Zurbriggen is Chief Scientific Officer and a member of the Executive Board of Pevion Biotech Ltd., which provided the vaccine.

Figures

Figure 1. Study Design.
Figure 1. Study Design.
P = Vaccination with PEV3A, F = Vaccination with FP9 ME-TRAP, M = Vaccination with MVA ME-TRAP, C = Sporozoite challenge
Figure 2. CONSORT Flow Chart
Figure 2. CONSORT Flow Chart
Figure 3. Anti-peptide responses.
Figure 3. Anti-peptide responses.
A. (Upper panel) Geometric mean anti-AMA49-C1 (PEV301) and anti UK-39 (PEV302) endpoint-titres (log10) with 95% confidence intervals for group 1 (PEV3A), group 2 (PEV3A+ME-TRAP) and unvaccinated controls. B. (Lower panel) Mean avidity increase (relative to avidity index after first immunisation) with 95% confidence intervals. Arrows along the x axis represent the timing of each vaccination, and the asterisk denotes the challenge.
Figure 4. Antibody reactivity with P. falciparum.
Figure 4. Antibody reactivity with P. falciparum.
A. (Upper panel) IgG endpoint titres measured in IFA with P. falciparum (K1) blood stage parasites. B. (Lower panel) IgG endpoint titres measured in IFA with P. falciparum (NF54) sporozoites. Sera were tested before vaccination, after every vaccination and after sporozoite challenge. Individual titres and the geometric mean for every time point are shown on a log scale.
Figure 5. Number of infected hepatocytes.
Figure 5. Number of infected hepatocytes.
Estimated numbers of infected hepatocytes for individual volunteers are shown. Lines represent the geometric mean for each group with IQ ranges.
Figure 6. Kaplan Meier Survival curve.
Figure 6. Kaplan Meier Survival curve.
A Kaplan Meier plot showing time to diagnosis of malaria for each group of volunteers after the challenge.
Figure 7. PCR data.
Figure 7. PCR data.
Number of parasites per mL, estimated using a calibration curve, plotted by day post sporozoite challenge. A. Three volunteers selected to demonstrate expected pattern of PCR results: an exponential increase in the numbers of parasites over time, with some cycling seen as parasites are sequestered and released. B. This figure shows data from all 4 volunteers who had unusual PCR results. Up to 150 parasites/mL are detected, and are subsequently undetectable for one or more time points, before they are measured again. This pattern occurs several times, up to a maximum of 5 times in volunteer N525.
Figure 8. Growth rates, mean and IQ…
Figure 8. Growth rates, mean and IQ ranges, by group.
Growth rate (parasites per mL per cycle) for each individual is shown; the lines represent the group means and inter quartile ranges.
Figure 9. Normal parasites and crisis forms.
Figure 9. Normal parasites and crisis forms.
Crisis forms differ from normal parasites in their slightly more ‘ragged’ morphology and altered staining of the nuclear material-live parasites have nuclei that stain red/blue, these abnormal parasites lose all red chromatin staining. Magnified high power images are shown in A&B. Normal parasites are seen in Figure 9A, whilst 9B shows a film containing a crisis form. The remaining films are unmagnified and white blood cell nuclei are clearly visible in blue, with smaller headphone shaped parasites. Figure 9C & D show single crisis forms, whilst 9E & F show multiple normal parasites, with the characteristic red/blue nuclear staining.

References

    1. Snow RW, Guerra CA, Noor AM, Myint HY, Hay SI. The global distribution of clinical episodes of Plasmodium falciparum malaria. Nature. 2005;434:214–217.
    1. Richie TL, Saul A. Progress and challenges for malaria vaccines. Nature. 2002;415:694–701.
    1. Miller LH, Howard RJ, Carter R, Good MF, Nussenzweig V, et al. Research toward malaria vaccines. Science. 1986;234:1349–1356.
    1. Wang R, Charoenvit Y, Daly TM, Long CA, Corradin G, et al. Protective efficacy against malaria of a combination sporozoite and erythrocytic stage vaccine. Immunol Lett. 1996;53:83–93.
    1. Hutchings CL, Birkett AJ, Moore AC, Hill AV. Combination of protein and viral vaccines induces potent cellular and humoral immune responses and enhanced protection from murine malaria challenge. Infect Immun. 2007;75:5819–5826.
    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. 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. 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. 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. Bejon P, Mwacharo J, Kai O, Mwangi T, Milligan P, et al. A Phase 2b Randomised Trial of the Candidate Malaria Vaccines FP9 ME-TRAP and MVA ME-TRAP among Children in Kenya. PLoS Clin Trials. 2006;1:e29.
    1. Mueller MS, Renard A, Boato F, Vogel D, Naegeli M, et al. Induction of parasite growth-inhibitory antibodies by a virosomal formulation of a peptidomimetic of loop I from domain III of Plasmodium falciparum apical membrane antigen 1. Infect Immun. 2003;71:4749–4758.
    1. Okitsu SL, Kienzl U, Moehle K, Silvie O, Peduzzi E, et al. Structure-activity-based design of a synthetic malaria peptide eliciting sporozoite inhibitory antibodies in a virosomal formulation. Chem Biol. 2007;14:577–587.
    1. Gluck R, Mischler R, Brantschen S, Just M, Althaus B, et al. Immunopotentiating reconstituted influenza virus virosome vaccine delivery system for immunization against hepatitis A. J Clin Invest. 1992;90:2491–2495.
    1. Genton B, Pluschke G, Degen L, Kammer AR, Westerfeld N, et al. A randomized placebo-controlled phase Ia malaria vaccine trial of two virosome-formulated synthetic peptides in healthy adult volunteers. PLoS ONE. 2007;2:e1018.
    1. Webster DP, Dunachie S, McConkey S, Poulton I, Moore AC, et al. Safety of recombinant fowlpox strain FP9 and modified vaccinia virus Ankara vaccines against liver-stage P. falciparum malaria in non-immune volunteers. Vaccine. 2006;24:3026–3034.
    1. Chulay JD, Schneider I, Cosgriff TM, Hoffman SL, Ballou WR, et al. Malaria transmitted to humans by mosquitoes infected from cultured Plasmodium falciparum. Am J Trop Med Hyg. 1986;35:66–68.
    1. Andrews L, Andersen RF, Webster D, Dunachie S, Walther RM, et al. Quantitative real-time polymerase chain reaction for malaria diagnosis and its use in malaria vaccine clinical trials. Am J Trop Med Hyg. 2005;73:191–198.
    1. Hermsen CC, de Vlas SJ, van Gemert GJ, Telgt DS, Verhage DF, et al. Testing vaccines in human experimental malaria: statistical analysis of parasitemia measured by a quantitative real-time polymerase chain reaction. Am J Trop Med Hyg. 2004;71:196–201.
    1. Vuola JM, Keating S, Webster DP, Berthoud T, Dunachie S, et al. Differential immunogenicity of various heterologous prime-boost vaccine regimens using DNA and viral vectors in healthy volunteers. J Immunol. 2005;174:449–455.
    1. Ferreira MU, Katzin AM. The assessment of antibody affinity distribution by thiocyanate elution: a simple dose-response approach. J Immunol Methods. 1995;187:297–305.
    1. Malkin EM, Diemert DJ, McArthur JH, Perreault JR, Miles AP, et al. Phase 1 clinical trial of apical membrane antigen 1: an asexual blood-stage vaccine for Plasmodium falciparum malaria. Infect Immun. 2005;73:3677–3685.
    1. Taliaferro WT, LG. The effect of immunity on the asexual reproduction of Plasmodium brasilianum. The Journal of Infectious Diseases. 1944;75:1–32.
    1. Klotz FW, Hudson DE, Coon HG, Miller LH. Vaccination-induced variation in the 140 kD merozoite surface antigen of Plasmodium knowlesi malaria. J Exp Med. 1987;165:359–367.
    1. Clark IA, Virelizier JL, Carswell EA, Wood PR. Possible importance of macrophage-derived mediators in acute malaria. Infect Immun. 1981;32:1058–1066.
    1. Karunaweera ND, Carter R, Grau GE, Kwiatkowski D, Del Giudice G, et al. Tumour necrosis factor-dependent parasite-killing effects during paroxysms in non-immune Plasmodium vivax malaria patients. Clin Exp Immunol. 1992;88:499–505.
    1. Jensen JB, Boland MT, Akood M. Induction of crisis forms in cultured Plasmodium falciparum with human immune serum from Sudan. Science. 1982;216:1230–1233.
    1. Florens L, Washburn MP, Raine JD, Anthony RM, Grainger M, et al. A proteomic view of the Plasmodium falciparum life cycle. Nature. 2002;419:520–526.
    1. Dunachie SJ, Walther M, Epstein JE, Keating S, Berthoud T, et al. A DNA prime-modified vaccinia virus Ankara boost vaccine encoding thrombospondin-related adhesion protein but not circumsporozoite protein partially protects healthy malaria-naive adults against Plasmodium falciparum sporozoite challenge. Infect Immun. 2006;74:5933–5942.
    1. Walther M, Thompson FM, Dunachie S, Keating S, Todryk S, et al. Safety, immunogenicity, and efficacy of prime-boost immunization with recombinant poxvirus FP9 and modified vaccinia virus Ankara encoding the full-length Plasmodium falciparum circumsporozoite protein. Infect Immun. 2006;74:2706–2716.
    1. Dunachie SJ, Walther M, Vuola JM, Webster DP, Keating SM, et al. A clinical trial of prime-boost immunisation with the candidate malaria vaccines RTS,S/AS02A and MVA-CS. Vaccine. 2006;24:2850–2859.
    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.
    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. Pombo DJ, Lawrence G, Hirunpetcharat C, Rzepczyk C, Bryden M, et al. Immunity to malaria after administration of ultra-low doses of red cells infected with Plasmodium falciparum. Lancet. 2002;360:610–617.

Source: PubMed

3
Abonnere