Evaluation of the efficacy of ChAd63-MVA vectored vaccines expressing circumsporozoite protein and ME-TRAP against controlled human malaria infection in malaria-naive individuals

Susanne H Hodgson, Katie J Ewer, Carly M Bliss, Nick J Edwards, Thomas Rampling, Nicholas A Anagnostou, Eoghan de Barra, Tom Havelock, Georgina Bowyer, Ian D Poulton, Simone de Cassan, Rhea Longley, Joseph J Illingworth, Alexander D Douglas, Pooja B Mange, Katharine A Collins, Rachel Roberts, Stephen Gerry, Eleanor Berrie, Sarah Moyle, Stefano Colloca, Riccardo Cortese, Robert E Sinden, Sarah C Gilbert, Philip Bejon, Alison M Lawrie, Alfredo Nicosia, Saul N Faust, Adrian V S Hill, Susanne H Hodgson, Katie J Ewer, Carly M Bliss, Nick J Edwards, Thomas Rampling, Nicholas A Anagnostou, Eoghan de Barra, Tom Havelock, Georgina Bowyer, Ian D Poulton, Simone de Cassan, Rhea Longley, Joseph J Illingworth, Alexander D Douglas, Pooja B Mange, Katharine A Collins, Rachel Roberts, Stephen Gerry, Eleanor Berrie, Sarah Moyle, Stefano Colloca, Riccardo Cortese, Robert E Sinden, Sarah C Gilbert, Philip Bejon, Alison M Lawrie, Alfredo Nicosia, Saul N Faust, Adrian V S Hill

Abstract

Background: Circumsporozoite protein (CS) is the antigenic target for RTS,S, the most advanced malaria vaccine to date. Heterologous prime-boost with the viral vectors simian adenovirus 63 (ChAd63)-modified vaccinia virus Ankara (MVA) is the most potent inducer of T-cells in humans, demonstrating significant efficacy when expressing the preerythrocytic antigen insert multiple epitope-thrombospondin-related adhesion protein (ME-TRAP). We hypothesized that ChAd63-MVA containing CS may result in a significant clinical protective efficacy.

Methods: We conducted an open-label, 2-site, partially randomized Plasmodium falciparum sporozoite controlled human malaria infection (CHMI) study to compare the clinical efficacy of ChAd63-MVA CS with ChAd63-MVA ME-TRAP.

Results: One of 15 vaccinees (7%) receiving ChAd63-MVA CS and 2 of 15 (13%) receiving ChAd63-MVA ME-TRAP achieved sterile protection after CHMI. Three of 15 vaccinees (20%) receiving ChAd63-MVA CS and 5 of 15 (33%) receiving ChAd63-MVA ME-TRAP demonstrated a delay in time to treatment, compared with unvaccinated controls. In quantitative polymerase chain reaction analyses, ChAd63-MVA CS was estimated to reduce the liver parasite burden by 69%-79%, compared with 79%-84% for ChAd63-MVA ME-TRAP.

Conclusions: ChAd63-MVA CS does reduce the liver parasite burden, but ChAd63-MVA ME-TRAP remains the most promising antigenic insert for a vectored liver-stage vaccine. Detailed analyses of parasite kinetics may allow detection of smaller but biologically important differences in vaccine efficacy that can influence future vaccine development.

Clinical trials registration: NCT01623557.

Keywords: CHMI; CS; ChAd63; ME-TRAP; MVA; P. falciparum; malaria; vaccine.

© The Author 2014. Published by Oxford University Press on behalf of the Infectious Diseases Society of America.

Figures

Figure 1.
Figure 1.
Flow of study design and volunteer recruitment. Twenty volunteers were excluded following screening for the following reasons: psychiatric history (n = 3), no medical screening letter returned (n = 3), multiple medical problems (n = 2), excessive alcohol use (n = 2), syncope (n = 1), connective tissue disease (n = 1), iron deficiency (n = 1), raised alanine aminotransferase level (n = 1), poor venous access (n = 1), gastrointestinal problems under investigation (n = 1), family history of heart disease (n = 1), lost to follow-up (n = 1), unavailable during challenge (n = 1), and history of recreational drug use (n = 1). Furthermore, 7 volunteers withdrew consent after screening but before enrollment. All immunizations were administered intramuscularly with sequential vaccines administered into the deltoid of alternating arms. No enrolled volunteers withdrew from the study and all volunteers completed study visits as scheduled. Abbreviations: ChAd63, simian adenovirus 63; CS, circumsporozoite protein; ME-TRAP, multiple epitope–thrombospondin-related adhesion protein; MVA, modified vaccinia virus Ankara; pfu, plaque-forming units; vp, viral particles.
Figure 2.
Figure 2.
Antigen-specific T-cell responses to vaccination measured by interferon γ enzyme-linked immunosorbent spot assay. Kinetics of T-cell responses after vaccination with ChAd63-MVA encoding either circumsporozoite protein (CS; group 1; A) or ME-TRAP (group 2; B). Each line represents an individual volunteer. **P < .01 and ***P < .001, by the Kruskal–Wallis test with the Dunn multiple comparison test. C, Median T-cell frequencies for both antigens by group. Mean T-cell frequencies at day 28 after vaccination were 304 and 673 spot-forming cells (SFCs) after ChAd63-MVA CS or ME-TRAP receipt, respectively, and at day 63 peaked at 1378 and 2068 SFCs after ChAd63-MVA CS or ME-TRAP receipt, respectively. Abbreviations: ChAd63, simian adenovirus 63; CHMI, controlled human malaria infection; controls, unvaccinated volunteers undergoing CHMI; ME-TRAP, multiple epitope–thrombospondin-related adhesion protein; MVA, modified vaccinia virus Ankara; PBMC, peripheral blood mononuclear cell.
Figure 3.
Figure 3.
Antibody responses to vaccination measured by enzyme-linked immunosorbent assay (ELISA). A, Anti- circumsporozoite protein (CS) immunoglobulin G (IgG) antibody responses after vaccination with ChAd63-MVA CS (group 1; red) or ME-TRAP (group 2; blue). Lines represent group medians. ***P = <.001 and *P = <.05, by the Friedman test comparing responses before and after vaccination with the Dunn post hoc test. B, Anti-TRAP IgG antibody responses after vaccination with ChAd63 ME-TRAP (group 2). ***P = .0002, by the 2-tailed Wilcoxon matched pairs test. C, Correlation between anti-CS IgG antibodies and CS-specific T-cell immunogenicity the day before challenge in group 1. Spearman r = 0.5; P = .08. Abbreviations: ChAd63, simian adenovirus 63; CHMI, controlled human malaria infection; controls, unvaccinated volunteers undergoing CHMI; ELISPOT, enzyme-linked immunosorbent spot assay; EU, ELISA units; ME-TRAP, multiple epitope–thrombospondin-related adhesion protein; MVA, modified vaccinia virus Ankara; PBMC, peripheral blood mononuclear cell; SFC, spot-forming cell.
Figure 4.
Figure 4.
Efficacy of ChAd63-MVA circumsporozoite protein (CS) and ME-TRAP immunization following Plasmodium falciparum 3D7 sporozoite challenge. Kaplan–Meier survival analyses. Log-rank test for significance. A, Kaplan–Meier survival analysis of time to treatment. Median time, 12.0 days for group 1 (CS), 12.5 days for group 2 (ME-TRAP), and 10.3 days for unvaccinated controls. B, Kaplan–Meier survival analysis of time to first sample with >500 parasites/mL detected by quantitative polymerase chain reaction (qPCR). Median time, 10.5 days for group 1 (CS), 12.0 days for group 2 (ME-TRAP), and 7.5 days for unvaccinated controls. C, Kaplan–Meier survival analysis of time to first sample with >20 parasites/mL detected by qPCR. Median time, 7.5 days for group 1 (CS), 9.0 days for group 2 (ME-TRAP), and 7.0 days for unvaccinated controls. Abbreviations: CHMI, controlled human malaria infection; controls, unvaccinated volunteers undergoing CHMI; ME-TRAP, multiple epitope–thrombospondin related adhesion protein.
Figure 5.
Figure 5.
Comparison of mean parasite density, measured by quantitative polymerase chain reaction, 7.5 days after controlled human malaria infection (CHMI) between vaccinees and control volunteers. P values were determined by the Mann–Whitney U test. Abbreviations: ChAd63, simian adenovirus 63; Control, unvaccinated volunteers undergoing CHMI; CS, circumsporozoite protein; group 1, ChAd63-MVA CS recipients; group 2, ChAd63 ME-TRAP recipients; ME-TRAP, multiple epitope–thrombospondin-related adhesion protein; MVA, modified vaccinia virus Ankara.
Figure 6.
Figure 6.
Comparison of areas under the curve (AUCs) of parasite densities, measured by quantitative polymerase chain reaction (PCR), between vaccinees and control volunteers. A, Group mean log-transformed PCR data. The AUC of parasite density over the first 3 replication cycles in infected volunteers was a significant predictor of the time to diagnosis (hazard ratio, 1.000015 [95% confidence interval, 1.000008–1.000022], by Cox proportional hazards regression analysis; P < .000). B, AUC analysis of parasite densities, comparing controls to vaccinees at days 6.5–8 (the first cycle after hepatocyte release), days 8.5–10 (the second cycle), and days 10.5–12 (the third cycle) after controlled human malaria infection (CHMI). Means of log [parasite density + 1] were compared for each vaccine group to those of controls, using a 2-tailed t test. Abbreviations: ChAd63, simian adenovirus 63; controls, unvaccinated volunteers undergoing CHMI; CS, circumsporozoite protein; ME-TRAP, multiple epitope–thrombospondin-related adhesion protein; MVA, modified vaccinia virus Ankara; SP, sterile protection.
Figure 7.
Figure 7.
Associations between immunological outcomes and vaccine efficacy. Correlation between parasite density at day 7.5, measured by quantitative polymerase chain reaction (qPCR), and levels of anti–circumsporozoite protein (CS) immunoglobulin G (IgG) antibody in group 1 (CS; Spearman r = −0.6; P = .03; A) and group 2 (ME-TRAP; Spearman r = −.3; P = .34; B). C, Correlation between parasite density at day 7.5, measured by qPCR, and anti-TRAP IgG antibody responses in group 2 (ME-TRAP; Spearman r = −0.5; P = .05). D, Correlation between interferon γ (IFN-γ)–secreting T-cell frequency to CS measured by enzyme-linked immunosorbent spot (ELISPOT) parasite density at day 7.5 (parasite/mL measured by qPCR) in group 1 (CS; Spearman r = −0.2; P = .50. E, Correlation between IFN-γ–secreting T-cell frequency to ME-TRAP measured by ELISPOT and parasite density at day 7.5 (parasite/mL measured by qPCR) in group 2 (ME-TRAP; Spearman r = 0.1; P = .6). Abbreviations: Black filled points, sterilely protected vaccinees; EU, enzyme-linked immunosorbent assay units; group 1, ChAd63-MVA CS; group 2, ChAd63-MVA ME-TRAP; ME-TRAP, multiple epitope–thrombospondin-related adhesion protein; PBMC, peripheral blood mononuclear cell; SFC, spot-forming cell; unfilled points, vaccinees demonstrating delay to start of antimalarial therapy in comparison to unvaccinated control volunteers.

References

    1. White NJ, Pukrittayakamee S, Hien TT, et al. Malaria. Lancet. 2014;383:723–35.
    1. WHO World Malaria Report. 2013.
    1. Agnandji ST, Lell B, Fernandes JF, et al. A phase 3 trial of RTS,S/AS01 malaria vaccine in African infants. N Engl J Med. 2012;367:2284–95.
    1. Olotu A, Fegan G, Wambua J, et al. Four-year efficacy of RTS,S/AS01E and its interaction with malaria exposure. N Engl J Med. 2013;368:1111–20.
    1. Bejon P, White MT, Olotu A, et al. Efficacy of RTS,S malaria vaccines: individual-participant pooled analysis of phase 2 data. Lancet Infect Dis. 2013;13:319–27.
    1. Agnandji ST, Fendel R, Mestre M, et al. Induction of Plasmodium falciparum-specific CD4+ T cells and memory B cells in Gabonese children vaccinated with RTS,S/AS01(E) and RTS,S/AS02(D) PLoS One. 2011;6:e18559.
    1. Foquet L, Hermsen CC, van Gemert GJ, et al. Vaccine-induced monoclonal antibodies targeting circumsporozoite protein prevent Plasmodium falciparum infection. J Clin Invest. 2014;124:140–4.
    1. Ndungu FM, Mwacharo J, Kimani D, et al. A statistical interaction between circumsporozoite protein-specific T cell and antibody responses and risk of clinical malaria episodes following vaccination with RTS,S/AS01E. PLoS One. 2012;7:e52870.
    1. Birkett AJ, Moorthy VS, Loucq C, Chitnis CE, Kaslow DC. Malaria vaccine R&D in the Decade of Vaccines: breakthroughs, challenges and opportunities. Vaccine. 2013;31(suppl 2):B233–43.
    1. WHO. Malaria Vaccine Technology Roadmap. 2013.
    1. Sedegah M, Hollingdale MR, Farooq F, et al. Sterile immunity to malaria after DNA prime/adenovirus boost immunization is associated with effector memory CD8+T cells targeting AMA1 class I epitopes. PLoS One. 2014;9:e106241.
    1. Schwenk RJ, Richie TL. Protective immunity to pre-erythrocytic stage malaria. Trends Parasitol. 2011;27:306–14.
    1. Hoffman SL, Isenbarger D, Long GW, et al. Sporozoite vaccine induces genetically restricted T cell elimination of malaria from hepatocytes. Science. 1989;244:1078–81.
    1. Khusmith S, Sedegah M, Hoffman SL. Complete protection against Plasmodium yoelii by adoptive transfer of a CD8+ cytotoxic T-cell clone recognizing sporozoite surface protein 2. Infect Immun. 1994;62:2979–83.
    1. Romero P, Maryanski JL, Corradin G, et al. Cloned cytotoxic T cells recognize an epitope in the circumsporozoite protein and protect against malaria. Nature. 1989;341:323–6.
    1. Weiss WR, Mellouk S, Houghten RA, et al. Cytotoxic T cells recognize a peptide from the circumsporozoite protein on malaria-infected hepatocytes. J Exp Med. 1990;171:763–73.
    1. Ewer KJ, O'Hara GA, Duncan CJ, et al. Protective CD8(+) T-cell immunity to human malaria induced by chimpanzee adenovirus-MVA immunisation. Nat Commun. 2013;4:2836.
    1. Hill AV, Reyes-Sandoval A, O'Hara G, et al. Prime-boost vectored malaria vaccines: progress and prospects. Hum Vaccin. 2010;6:78–83.
    1. Chuang I, Sedegah M, Cicatelli S, et al. DNA prime/Adenovirus boost malaria vaccine encoding P. falciparum CSP and AMA1 induces sterile protection associated with cell-mediated immunity. PLoS One. 2013;8:e55571.
    1. Tamminga C, Sedegah M, Maiolatesi S, et al. Human adenovirus 5-vectored Plasmodium falciparum NMRC-M3V-Ad-PfCA vaccine encoding CSP and AMA1 is safe, well-tolerated and immunogenic but does not protect against controlled human malaria infection. Hum Vaccin Immunother. 2013;9:2165–77.
    1. Reyes-Sandoval A, Berthoud T, Alder N, et al. Prime-boost immunization with adenoviral and modified vaccinia virus Ankara vectors enhances the durability and polyfunctionality of protective malaria CD8+ T-cell responses. Infect Immun. 2010;78:145–53.
    1. Colloca S, Barnes E, Folgori A, et al. Vaccine vectors derived from a large collection of simian adenoviruses induce potent cellular immunity across multiple species. Sci Transl Med. 2012;4 115ra112.
    1. Sauerwein RW, Roestenberg M, Moorthy VS. Experimental human challenge infections can accelerate clinical malaria vaccine development. Nat Rev Immunol. 2011;11:57–64.
    1. Thompson FM, Porter DW, Okitsu SL, et al. Evidence of blood stage efficacy with a virosomal malaria vaccine in a phase IIa clinical trial. PLoS One. 2008;3:e1493.
    1. O'Hara GA, Duncan CJ, Ewer KJ, et al. Clinical assessment of a recombinant simian adenovirus ChAd63: a potent new vaccine vector. J Infect Dis. 2012;205:772–81.
    1. Dunachie SJ, Walther M, Epstein JE, 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;74:5933–42.
    1. Walther M, Thompson FM, Dunachie 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;74:2706–16.
    1. Imoukhuede EB, Berthoud T, Milligan P, et al. Safety and immunogenicity of the malaria candidate vaccines FP9 CS and MVA CS in adult Gambian men. Vaccine. 2006;24:6526–33.
    1. Bejon P, Peshu N, Gilbert SC, 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;42:1102–10.
    1. Bruna-Romero O, Rocha CD, Tsuji M, Gazzinelli RT. Enhanced protective immunity against malaria by vaccination with a recombinant adenovirus encoding the circumsporozoite protein of Plasmodium lacking the GPI-anchoring motif. Vaccine. 2004;22:3575–84.
    1. Coppi A, Pinzon-Ortiz C, Hutter C, Sinnis P. The Plasmodium circumsporozoite protein is proteolytically processed during cell invasion. J Exp Med. 2005;201:27–33.
    1. Singh AP, Buscaglia CA, Wang Q, et al. Plasmodium circumsporozoite protein promotes the development of the liver stages of the parasite. Cell. 2007;131:492–504.
    1. Cottingham MG, Carroll F, Morris SJ, et al. Preventing spontaneous genetic rearrangements in the transgene cassettes of adenovirus vectors. Biotechnol Bioeng. 2012;109:719–28.
    1. Epstein JE, Tewari K, Lyke KE, et al. Live attenuated malaria vaccine designed to protect through hepatic CD8(+) T cell immunity. Science. 2011;334:475–80.
    1. Sheehy SH, Duncan CJ, Elias SC, et al. ChAd63-MVA-vectored blood-stage malaria vaccines targeting MSP1 and AMA1: assessment of efficacy against mosquito bite challenge in humans. Mol Ther. 2012;20:2355–68.
    1. Bejon P, Andrews L, Andersen RF, 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;191:619–26.
    1. Sheehy SH, Duncan CJ, Elias SC, et al. Phase Ia clinical evaluation of the Plasmodium falciparum blood-stage antigen MSP1 in ChAd63 and MVA vaccine vectors. Mol Ther. 2011;19:2269–76.
    1. Sheehy SH, Duncan CJ, Elias SC, et al. Phase Ia clinical evaluation of the safety and immunogenicity of the Plasmodium falciparum blood-stage antigen AMA1 in ChAd63 and MVA vaccine vectors. PLoS One. 2012;7:e31208.
    1. Sedegah M, Kim Y, Ganeshan H, et al. Identification of minimal human MHC-restricted CD8+ T-cell epitopes within the Plasmodium falciparum circumsporozoite protein (CSP) Malar J. 2013;12:185.
    1. Epstein JE, Rao S, Williams F, et al. Safety and clinical outcome of experimental challenge of human volunteers with Plasmodium falciparum-infected mosquitoes: an update. J Infect Dis. 2007;196:145–54.
    1. Kester KE, Cummings JF, Ofori-Anyinam O, et al. Randomized, double-blind, phase 2a trial of falciparum malaria vaccines RTS,S/AS01B and RTS,S/AS02A in malaria-naive adults: safety, efficacy, and immunologic associates of protection. J Infect Dis. 2009;200:337–46.
    1. Webster DP, Dunachie S, Vuola JM, 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;102:4836–41.

Source: PubMed

3
Subskrybuj