Protective CD8+ T-cell immunity to human malaria induced by chimpanzee adenovirus-MVA immunisation

Katie J Ewer, Geraldine A O'Hara, Christopher J A Duncan, Katharine A Collins, Susanne H Sheehy, Arturo Reyes-Sandoval, Anna L Goodman, Nick J Edwards, Sean C Elias, Fenella D Halstead, Rhea J Longley, Rosalind Rowland, Ian D Poulton, Simon J Draper, Andrew M Blagborough, Eleanor Berrie, Sarah Moyle, Nicola Williams, Loredana Siani, Antonella Folgori, Stefano Colloca, Robert E Sinden, Alison M Lawrie, Riccardo Cortese, Sarah C Gilbert, Alfredo Nicosia, Adrian V S Hill, Katie J Ewer, Geraldine A O'Hara, Christopher J A Duncan, Katharine A Collins, Susanne H Sheehy, Arturo Reyes-Sandoval, Anna L Goodman, Nick J Edwards, Sean C Elias, Fenella D Halstead, Rhea J Longley, Rosalind Rowland, Ian D Poulton, Simon J Draper, Andrew M Blagborough, Eleanor Berrie, Sarah Moyle, Nicola Williams, Loredana Siani, Antonella Folgori, Stefano Colloca, Robert E Sinden, Alison M Lawrie, Riccardo Cortese, Sarah C Gilbert, Alfredo Nicosia, Adrian V S Hill

Abstract

Induction of antigen-specific CD8(+) T cells offers the prospect of immunization against many infectious diseases, but no subunit vaccine has induced CD8(+) T cells that correlate with efficacy in humans. Here we demonstrate that a replication-deficient chimpanzee adenovirus vector followed by a modified vaccinia virus Ankara booster induces exceptionally high frequency T-cell responses (median >2400 SFC/10(6) peripheral blood mononuclear cells) to the liver-stage Plasmodium falciparum malaria antigen ME-TRAP. It induces sterile protective efficacy against heterologous strain sporozoites in three vaccinees (3/14, 21%), and delays time to patency through substantial reduction of liver-stage parasite burden in five more (5/14, 36%), P=0.008 compared with controls. The frequency of monofunctional interferon-γ-producing CD8(+) T cells, but not antibodies, correlates with sterile protection and delay in time to patency (P(corrected)=0.005). Vaccine-induced CD8(+) T cells provide protection against human malaria, suggesting that a major limitation of previous vaccination approaches has been the insufficient magnitude of induced T cells.

Trial registration: ClinicalTrials.gov NCT00890760.

Conflict of interest statement

Sarah Gilbert, Arturo Reyes-Sandoval, Anna Goodman, Geraldine O'Hara and Adrian Hill are named inventors on patent applications covering malaria vectored vaccines and immunization regimes including: WO/2008/122811-Adenoviral vectors encoding a pathogen or tumour antigen and WO/2008/122769-Adenoviral vector encoding malaria antigen. Authors from Okairos are employees of and/or share holders in Okairos which is developing vectored malaria vaccines. All other authors declare no competing financial interests.

Figures

Figure 1. Immunogenicity of prime-boost vaccination with…
Figure 1. Immunogenicity of prime-boost vaccination with ChAd63-MVA ME-TRAP.
(a) Median ME-TRAP IFNγ ex vivo ELISPOT responses to the T9/96 strain for each group, P=0.04 for peak immunogenicity (D28 for A (n=10) and D63 for AM (n=14)), *P=0.012 for difference in immunogenicity at D166, both two-tailed Mann–Whitney. (b) Breadth of TRAP-specific ELISPOT responses before (D14, D56) and after boosting (D63) with MVA ME-TRAP, using T9/96 strain peptides, showing mean (±s.e.m.) number of pools recognized. (c) Correlation of peak ELISPOT responses to T9/96 TRAP post-prime and at time of sporozoite challenge. (d) Individual responses to TRAP and ME at time of sporozoite challenge (data points in red represent sterilely protected volunteers). (e) Median with IQR IFNγ ELISPOT responses to TRAP peptides from the heterologous vaccine and challenge strains of P. falciparum. (f) IgG antibodies to TRAP boosted by MVA compared with ChAd63 alone (medians); differences not statistically significant. (g) Spearman’s correlation of pre-existing antibodies to ChAd63 with peak vaccine-induced ELISPOT responses to TRAP (T9/96 strain).
Figure 2. Vaccine efficacy assessments by time…
Figure 2. Vaccine efficacy assessments by time to microscopic patency and PCR measures.
Kaplan–Meier log-rank comparison of days to positive blood film (a) and >20 parasites per ml by PCR (b) between adenovirus-MVA prime-boost (Ad-M) group (n=14), adenovirus-only (Ad) group (n=10) and controls (n=12). Mean days to positivity by blood film: Ad-M 14.6 days (95% CI: 12.3–16.8), adenovirus-alone group (Ad) 11.3 days (95% CI: 10.2–12.5), control group 11.8 days (95% CI: 10.8–12.7); and to 20 parasites per ml by PCR: Ad-M 11.6 days (95% CI: 8.5–14.7), Ad 7.8 days (95% CI: 7.0–8.6), control group 8.1 days (95% CI: 4–8.8). (c) Group mean log-transformed PCR data (error bars represent s.e.m.). Area under curve analysis of parasite densities comparing controls to vaccinees (either including (d) or excluding (e) volunteers that were sterilely protected) at days 6.5–8 (first cycle post hepatocyte release), days 8.5–10 (second cycle) and 10.5–12 (third cycle) post challenge. Over the second and third cycles, there is a significant reduction in vaccinees’ parasite densities compared with controls; the lack of significance at cycle one probably reflects low power due to very low parasite densities. Comparison of areas under curves for all three cycles combined also shows a significant reduction in parasite densities between vaccinees and controls (P=0.003 when including sterilely protected vaccinees, P=0.01 when excluding sterilely protected vaccinees, (two-tailed t-test).
Figure 3. Functionality of TRAP-specific CD4 +…
Figure 3. Functionality of TRAP-specific CD4+ and CD8+ T cells.
(a) Peak immunogenicity (day 63=7 days after boosting) and (b) time of challenge (day 76). N=10 for adenovirus-only group and n=14 for Ad-M group. Frequencies of cytokine-secreting CD4+ and CD8+ T cells. *Staining for CD107a+ expression performed at challenge B only. Mean (with s.e.m.) responses are shown. (c,d) T cell responses shown are grouped according to number of functions at peak (c) and challenge (d). Pie charts summarize the fractions of the total response that are positive for three, two or one functions. All possible combinations of functions are shown in the bar chart stratified by vaccination regimen, with the y axis showing the percentage of CD4+ or CD8+ cells. Data points represent individual volunteers.
Figure 4. Correlates of protective efficacy.
Figure 4. Correlates of protective efficacy.
(a) Correlation of time to parasitaemia with ex vivo ELISPOT responses for Ad-MVA vaccinees, P=0.97. (b) Correlation of time to parasitaemia with frequency of CD8+ IFNγ+/IL-2− /TNFα− for Ad-MVA vaccinees, P=0.0005. For vaccinees receiving Ad alone or Ad-MVA (n=24), correlation of time to parasitaemia with frequency of CD8+ IFNγ+/IL-2− /TNFα−, rs=0.61, P=0.002. Both (a) and (b) were assessed at the time of sporozoite challenge. (c) Correlation of time to parasitaemia with frequency of CD107a+/ IFNγ−/IL-2−/TNFα− CD8+ T cells at day 150 post challenge in challenge A, P=0.02. (d) Correlation of parasite density in second replication cycle with frequency CD8+ IFNγ+/IL-2− /TNFα− for Ad-MVA vaccinees. All correlations were performed using two-tailed Spearman’s correlation.

References

    1. Robinson H. L. & Amara R. R. T cell vaccines for microbial infections. Nat. Med. 11, S25–S32 (2005).
    1. Topham D. J., Tripp R. A. & Doherty P. C. CD8+ T cells clear influenza virus by perforin or Fas-dependent processes. J. Immunol. 159, 5197–5200 (1997).
    1. Gillespie G. M. et al. Functional heterogeneity and high frequencies of cytomegalovirus-specific CD8(+) T lymphocytes in healthy seropositive donors. J. Virol. 74, 8140–8150 (2000).
    1. Callan M. F. The evolution of antigen-specific CD8+ T cell responses after natural primary infection of humans with Epstein-Barr virus. Viral Immunol. 16, 3–16 (2003).
    1. Borrow P., Lewicki H., Hahn B. H., Shaw G. M. & Oldstone M. B. Virus-specific CD8+ cytotoxic T-lymphocyte activity associated with control of viremia in primary human immunodeficiency virus type 1 infection. J. Virol. 68, 6103–6110 (1994).
    1. Betts M. R. et al. Analysis of total human immunodeficiency virus (HIV)-specific CD4(+) and CD8(+) T-cell responses: relationship to viral load in untreated HIV infection. J. Virol. 75, 11983–11991 (2001).
    1. Arens R. & Schoenberger S. P. Plasticity in programming of effector and memory CD8 T-cell formation. Immunol. Rev. 235, 190–205 (2010).
    1. Whelan K. T. et al. Safety and immunogenicity of boosting BCG vaccinated subjects with BCG: comparison with boosting with a new TB vaccine, MVA85A. PLoS One 4, e5934 (2009).
    1. Darrah P. A. et al. Multifunctional TH1 cells define a correlate of vaccine-mediated protection against Leishmania major. Nat. Med. 13, 843–850 (2007).
    1. Schmidt N. W. et al. Memory CD8 T cell responses exceeding a large but definable threshold provide long-term immunity to malaria. Proc. Natl Acad. Sci. USA 105, 14017–14022 (2008).
    1. Reyes-Sandoval A. 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. 78, 145–153 (2010).
    1. Liu J. et al. Immune control of an SIV challenge by a T-cell-based vaccine in rhesus monkeys. Nature 457, 87–91 (2009).
    1. Doolan D. L. et al. DNA vaccines for malaria: the past, the present, & the future. Ind. J. Med. Res. 106, 109–119 (1997).
    1. Schneider J. 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. 4, 397–402 (1998).
    1. Li S. et al. Priming with recombinant influenza virus followed by administration of recombinant vaccinia virus induces CD8+ T-cell-mediated protective immunity against malaria. Proc. Natl Acad. Sci. USA 90, 5214–5218 (1993).
    1. Gilbert S. C. et al. Enhanced CD8 T cell immunogenicity and protective efficacy in a mouse malaria model using a recombinant adenoviral vaccine in heterologous prime-boost immunisation regimes. Vaccine 20, 1039–1045 (2002).
    1. McConkey S. J. et al. Enhanced T-cell immunogenicity of plasmid DNA vaccines boosted by recombinant modified vaccinia virus Ankara in humans. Nat. Med. 9, 729–735 (2003).
    1. Webster D. P. 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. USA 102, 4836–4841 (2005).
    1. Colloca S. et al. Vaccine vectors derived from a large collection of simian adenoviruses induce potent cellular immunity across multiple species. Sci. Transl. Med. 4, (2012).
    1. RTS,S Clinical Trials Partnership. A Phase 3 Trial of RTS,S/AS01 Malaria Vaccine in African Infants. N. Engl. J. Med. 367, 2284–2295 (2012).
    1. Agnandji S. T. et al. First results of phase 3 trial of RTS,S/AS01 malaria vaccine in African children. N. Engl. J. Med. 365, 1863–1875 (2011).
    1. Hutchings C. L., Birkett A. J., Moore A. C. & Hill A. V. Combination of protein and viral vaccines induces potent cellular and humoral immune responses and enhanced protection from murine malaria challenge. Infect. Immun. 75, 5819–5826 (2007).
    1. O’Hara G. A. et al. Clinical assessment of a recombinant simian adenovirus ChAd63: a potent new vaccine vector. J. Infect. Dis. 205, 772–781 (2012).
    1. Capone S. et al. Immune responses against a liver-stage malaria antigen induced by simian adenoviral vector AdCh63 and MVA prime-boost immunisation in non-human primates. Vaccine 29, 256–265 (2010).
    1. Buchbinder S. P. et al. Efficacy assessment of a cell-mediated immunity HIV-1 vaccine (the Step Study): a double-blind, randomised, placebo-controlled, test-of-concept trial. Lancet 372, 1881–1893 (2008).
    1. Dangoor A. et al. Clinical and immunological responses in metastatic melanoma patients vaccinated with a high-dose poly-epitope vaccine. Cancer Immunol. Immunother 59, 863–873 (2010).
    1. Tamminga C. et al. Adenovirus-5-vectored P. falciparum vaccine expressing CSP and AMA1. Part B: safety, immunogenicity and protective efficacy of the CSP component. PLoS One 6, e25868 (2011).
    1. Dunachie S. J. 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. 74, 5933–5942 (2006).
    1. Bejon P. 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. 191, 619–626 (2005).
    1. Andrews L. et al. Quantitative real-time polymerase chain reaction for malaria diagnosis and its use in malaria vaccine clinical trials. Am. J. Trop. Med. Hyg. 73, 191–198 (2005).
    1. Wang R. et al. Induction of antigen-specific cytotoxic T lymphocytes in humans by a malaria DNA vaccine. Science 282, 476–480 (1998).
    1. Epstein J. E. et al. Live attenuated malaria vaccine designed to protect through hepatic CD8(+) T cell immunity. Science 334, 475–480 (2011).
    1. Bett A. J. et al. Comparison of T cell immune responses induced by vectored HIV vaccines in non-human primates and humans. Vaccine 28, 7881–7889 (2010).
    1. Dudareva M. et al. Prevalence of serum neutralizing antibodies against chimpanzee adenovirus 63 and human adenovirus 5 in Kenyan children, in the context of vaccine vector efficacy. Vaccine 27, 3501–3504 (2009).
    1. Chen H. et al. Adenovirus-based vaccines: comparison of vectors from three species of adenoviridae. J. Virol. 84, 10522–10532 (2010).
    1. Kester K. E. et al. Phase 2a trial of 0, 1, and 3 month and 0, 7, and 28 day immunization schedules of malaria vaccine RTS,S/AS02 in malaria-naive adults at the Walter Reed Army Institute of Research. Vaccine 26, 2191–2202 (2008).
    1. Kester K. E. 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. 200, 337–346 (2009).
    1. Barouch D. H. et al. Protective efficacy of a single immunization of a chimeric adenovirus vector-based vaccine against simian immunodeficiency virus challenge in rhesus monkeys. J. Virol. 83, 9584–9590 (2009).
    1. Sheehy S. H. 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 7, e31208 (2012).
    1. Sheehy S. H. et al. Phase Ia clinical evaluation of the Plasmodium falciparum blood-stage antigen MSP1 in ChAd63 and MVA vaccine vectors. Mol. Ther. 19, 2269–2276 (2011).
    1. Barnes E. et al. Novel adenovirus-based vaccines induce broad and sustained T cell responses to HCV in man. Sci. Transl. Med. 4, (2012).
    1. Plotkin S. A. Vaccines: correlates of vaccine-induced immunity. Clin. Infect. Dis. 47, 401–409 (2008).
    1. Moorthy V. S. & Ballou W. R. Immunological mechanisms underlying protection mediated by RTS,S: a review of the available data. Malar. J. 8, 312 (2009).
    1. Bakshi S. & Imoukhuede E. B. Malaria Vectored Vaccines Consortium (MVVC). Hum. Vaccin. 6, 433–434 (2010).
    1. Hermsen C. C. et al. Detection of Plasmodium falciparum malaria parasites in vivo by real-time quantitative PCR. Mol. Biochem. Parasitol. 118, 247–251 (2001).
    1. Wang C. W. et al. The Plasmodium falciparum var gene transcription strategy at the onset of blood stage infection in a human volunteer. Parasitol. Int. 58, 478–480 (2009).
    1. Sauerwein R. W., Roestenberg M. & Moorthy V. S. Experimental human challenge infections can accelerate clinical malaria vaccine development. Nat. Rev. Immunol. 11, 57–64 (2011).
    1. Roestenberg M. et al. Comparison of clinical and parasitological data from controlled human malaria infection trials. PLoS One 7, e38434 (2012).

Source: PubMed

3
Předplatit