Phase Ia clinical evaluation of the safety and immunogenicity of the Plasmodium falciparum blood-stage antigen AMA1 in ChAd63 and MVA vaccine vectors

Susanne H Sheehy, Christopher J A Duncan, Sean C Elias, Sumi Biswas, Katharine A Collins, Geraldine A O'Hara, Fenella D Halstead, Katie J Ewer, Tabitha Mahungu, Alexandra J Spencer, Kazutoyo Miura, Ian D Poulton, Matthew D J Dicks, Nick J Edwards, Eleanor Berrie, Sarah Moyle, Stefano Colloca, Riccardo Cortese, Katherine Gantlett, Carole A Long, Alison M Lawrie, Sarah C Gilbert, Tom Doherty, Alfredo Nicosia, Adrian V S Hill, Simon J Draper, Susanne H Sheehy, Christopher J A Duncan, Sean C Elias, Sumi Biswas, Katharine A Collins, Geraldine A O'Hara, Fenella D Halstead, Katie J Ewer, Tabitha Mahungu, Alexandra J Spencer, Kazutoyo Miura, Ian D Poulton, Matthew D J Dicks, Nick J Edwards, Eleanor Berrie, Sarah Moyle, Stefano Colloca, Riccardo Cortese, Katherine Gantlett, Carole A Long, Alison M Lawrie, Sarah C Gilbert, Tom Doherty, Alfredo Nicosia, Adrian V S Hill, Simon J Draper

Abstract

Background: Traditionally, vaccine development against the blood-stage of Plasmodium falciparum infection has focused on recombinant protein-adjuvant formulations in order to induce high-titer growth-inhibitory antibody responses. However, to date no such vaccine encoding a blood-stage antigen(s) alone has induced significant protective efficacy against erythrocytic-stage infection in a pre-specified primary endpoint of a Phase IIa/b clinical trial designed to assess vaccine efficacy. Cell-mediated responses, acting in conjunction with functional antibodies, may be necessary for immunity against blood-stage P. falciparum. The development of a vaccine that could induce both cell-mediated and humoral immune responses would enable important proof-of-concept efficacy studies to be undertaken to address this question.

Methodology: We conducted a Phase Ia, non-randomized clinical trial in 16 healthy, malaria-naïve adults of the chimpanzee adenovirus 63 (ChAd63) and modified vaccinia virus Ankara (MVA) replication-deficient viral vectored vaccines encoding two alleles (3D7 and FVO) of the P. falciparum blood-stage malaria antigen; apical membrane antigen 1 (AMA1). ChAd63-MVA AMA1 administered in a heterologous prime-boost regime was shown to be safe and immunogenic, inducing high-level T cell responses to both alleles 3D7 (median 2036 SFU/million PBMC) and FVO (median 1539 SFU/million PBMC), with a mixed CD4(+)/CD8(+) phenotype, as well as substantial AMA1-specific serum IgG responses (medians of 49 µg/mL and 41 µg/mL for 3D7 and FVO AMA1 respectively) that demonstrated growth inhibitory activity in vitro.

Conclusions: ChAd63-MVA is a safe and highly immunogenic delivery platform for both alleles of the AMA1 antigen in humans which warrants further efficacy testing. ChAd63-MVA is a promising heterologous prime-boost vaccine strategy that could be applied to numerous other diseases where strong cellular and humoral immune responses are required for protection.

Trial registration: ClinicalTrials.gov NCT01095055.

Conflict of interest statement

Competing Interests: AJS, MDJD, SCG, AVSH and SJD are named inventors on US 12/595 574 and UK PCT/GB2008/01262, US 12/595 351 and UK PCT/GB2008/01271 novel adenovirus patent applications covering malaria vectored vaccines and immunization regimes. This does not alter our adherence to all the PLoS ONE policies on sharing data and materials. Authors from Okairòs are employees of and/or shareholders in Okairòs, which is developing vectored vaccines for malaria and other diseases. This does not alter our adherence to all the PLoS ONE policies on sharing data and materials.

Figures

Figure 1. Flow chart of the study.
Figure 1. Flow chart of the study.
All vaccinations were administered intramuscularly. ChAd63 AMA1 was administered on day 0 and MVA AMA1 on day 56. Six volunteers were excluded following screening for the following reasons: psychiatric morbidity; recurrent severe urticaria; elevated alanine aminotransferase (87 IU/L); unexplained microscopic haematuria and proteinuria; and withdrawal of consent (two individuals).
Figure 2. Local and systemic AEs deemed…
Figure 2. Local and systemic AEs deemed definitely, probably or possibly related to ChAd63 AMA1.
Only the highest intensity of each AE per subject is listed. Data are combined for all AEs for all volunteers receiving the same vaccine at the stated dose. There were no immunization related serious AEs. (A) Local AEs post ChAd63 AMA1. (B) Systemic AEs post ChAd63 AMA1. ‘Other’ AEs post 5×109 vp ChAd63 AMA1 included cough, coryzal symptoms, abdominal pain and dysmenorrhoea. ‘Other’ AE post 5×1010 vp ChAd63 AMA1 was coryzal symptoms. All ‘other’ AEs were considered possibly related to vaccination due to a temporal association.
Figure 3. Local and systemic AEs deemed…
Figure 3. Local and systemic AEs deemed definitely, probably or possibly related to MVA AMA1.
Only the highest intensity of each AE per subject is listed. Data are combined for all AEs for all volunteers receiving the same vaccine at the stated dose. There were no immunization related serious AEs. (A) Local and systemic AEs post 5×108 pfu MVA AMA1. Local ‘other’ AE was mild bruising at vaccination site. Systemic ‘other’ AEs included two cases of rigor, one case of delirium, loss of appetite and chills. (B) Local and systemic AEs post 2.5×108 pfu MVA AMA1. ‘Other’ AE post 2.5×108 pfu MVA AMA1 was dizziness. (C) Local and systemic AEs post 1.25×108 pfu MVA AMA1.
Figure 4. Cellular immunogenicity of ChAd63 AMA1…
Figure 4. Cellular immunogenicity of ChAd63 AMA1 and ChAd63-MVA AMA1 immunization regimes.
(A) Groups 1A and 2A and (B) groups 1B and 2B median ex-vivo IFN-γ ELISPOT responses in PBMC to the AMA1 insert (summed response across all the individual peptide pools). Individual responses are shown in Figure S1. Note due to allele-specific peptide overlap some responses can be potentially counted twice. Individual breakdowns of the ELISpot responses are shown in Figure S2, and data in Figure S3C show that on average one quarter to one third of the total summed response can be attributed to FVO allele-specific peptides. (C) Median and individual IFN-γ ELISPOT responses at day 63 and (D) at day 140 that are functional to the individual alleles 3D7 (circles) and FVO (squares). (E,F) PBMC from day 84 for group 1B (closed symbols) and group 2B (open symbols) were re-stimulated with a pool of AMA1 peptides or cryopreserved iRBCs. Individual data points and the median are shown for (E) the % CD4+ and (F) CD8+ T cells positive for CD107a, IFN-γ, IL-2 or TNFα. The dotted line indicates the 0.05% level and any response <0.03% is not shown.
Figure 5. Antibody immunogenicity of ChAd63 AMA1…
Figure 5. Antibody immunogenicity of ChAd63 AMA1 and ChAd63-MVA AMA1 immunization regimes.
(A) Groups 1A and 2A and (B) groups 1B and 2B total IgG ELISA responses against 3D7 AMA1 as measured in the serum over time. The geometric mean response is shown for each group and individual responses are shown in Figure S7. The horizontal dotted line indicates the limit of detection of the assay. (C) AMA1-specific ELISA titers against 3D7 (circles) and FVO (squares) in µg/mL for group 1B (n = 4) and group 2B (n = 4) at the peak time-point (day 84). Individual data points and the median are shown. (D) % GIA against 3D7 and FVO parasites at day 0 and day 84 in group 1B (open symbols) and 2B (closed symbols). (E) Relationship between 3D7 strain % GIA and serum 3D7 AMA1-specific IgG ELISA titer at day 0 and day 84 in groups 1B (open symbols) and 2B (closed symbols).

References

    1. Das P, Horton R. Malaria elimination: worthy, challenging, and just possible. Lancet. 2010;376:1515–1517.
    1. Langhorne J, Ndungu FM, Sponaas AM, Marsh K. Immunity to malaria: more questions than answers. Nat Immunol. 2008;9:725–732.
    1. Goodman AL, Draper SJ. Blood-stage malaria vaccines - recent progress and future challenges. Ann Trop Med Parasitol. 2010;104:189–211.
    1. Sirima SB, Cousens S, Druilhe P. Protection against malaria by MSP3 candidate vaccine. N Engl J Med. 2011;365:1062–1064.
    1. Cech PG, Aebi T, Abdallah MS, Mpina M, Machunda EB, et al. Virosome-formulated Plasmodium falciparum AMA-1 & CSP derived peptides as malaria vaccine: randomized phase 1b trial in semi-immune adults & children. PLoS One. 2011;6:e22273.
    1. Thera MA, Doumbo OK, Coulibaly D, Laurens MB, Ouattara A, et al. A field trial to assess a blood-stage malaria vaccine. N Engl J Med. 2011;365:1004–1013.
    1. Spring MD, Cummings JF, Ockenhouse CF, Dutta S, Reidler R, et al. Phase 1/2a study of the malaria vaccine candidate apical membrane antigen-1 (AMA-1) administered in adjuvant system AS01B or AS02A. PLoS ONE. 2009;4:e5254.
    1. Ellis RD, Sagara I, Doumbo O, Wu Y. Blood stage vaccines for Plasmodium falciparum: Current status and the way forward. Hum Vaccin. 2010;6
    1. Good MF, Engwerda C. Defying malaria: Arming T cells to halt malaria. Nat Med. 2011;17:49–51.
    1. Good MF, Xu H, Wykes M, Engwerda CR. Development and regulation of cell-mediated immune responses to the blood stages of malaria: implications for vaccine research. Annu Rev Immunol. 2005;23:69–99.
    1. Su Z, Stevenson MM. Central role of endogenous gamma interferon in protective immunity against blood-stage Plasmodium chabaudi AS infection. Infect Immun. 2000;68:4399–4406.
    1. Yoneto T, Waki S, Takai T, Tagawa Y, Iwakura Y, et al. A critical role of Fc receptor-mediated antibody-dependent phagocytosis in the host resistance to blood-stage Plasmodium berghei XAT infection. J Immunol. 2001;166:6236–6241.
    1. Joos C, Marrama L, Polson HE, Corre S, Diatta AM, et al. Clinical protection from falciparum malaria correlates with neutrophil respiratory bursts induced by merozoites opsonized with human serum antibodies. PLoS One. 2010;5:e9871.
    1. Bouharoun-Tayoun H, Attanath P, Sabchareon A, Chongsuphajaisiddhi T, Druilhe P. Antibodies that protect humans against Plasmodium falciparum blood stages do not on their own inhibit parasite growth and invasion in vitro, but act in cooperation with monocytes. J Exp Med. 1990;172:1633–1641.
    1. Draper SJ, Goodman AL, Biswas S, Forbes EK, Moore AC, et al. Recombinant viral vaccines expressing merozoite surface protein-1 induce antibody- and T cell-mediated multistage protection against malaria. Cell Host Microbe. 2009;5:95–105.
    1. Kawabata Y, Udono H, Honma K, Ueda M, Mukae H, et al. Merozoite surface protein 1-specific immune response is protective against exoerythrocytic forms of Plasmodium yoelii. Infect Immun. 2002;70:6075–6082.
    1. Belnoue E, Voza T, Costa FT, Gruner AC, Mauduit M, et al. Vaccination with live Plasmodium yoelii blood stage parasites under chloroquine cover induces cross-stage immunity against malaria liver stage. J Immunol. 2008;181:8552–8558.
    1. Hill AVS, Reyes-Sandoval A, O'Hara G, Ewer K, Lawrie AM, et al. Prime-boost vectored malaria vaccines: progress and prospects. Hum Vaccin. 2010;6:78–83.
    1. Biswas S, Dicks MD, Long CA, Remarque EJ, Siani L, et al. Transgene Optimization, Immunogenicity and In Vitro Efficacy of Viral Vectored Vaccines Expressing Two Alleles of Plasmodium falciparum AMA1. PLoS One. 2011;6:e20977.
    1. Draper SJ, Biswas S, Spencer AJ, Remarque EJ, Capone S, et al. Enhancing blood-stage malaria subunit vaccine immunogenicity in rhesus macaques by combining adenovirus, poxvirus, and protein-in-adjuvant vaccines. J Immunol. 2010;185:7583–7595.
    1. Remarque EJ, Faber BW, Kocken CH, Thomas AW. Apical membrane antigen 1: a malaria vaccine candidate in review. Trends Parasitol. 2008;24:74–84.
    1. Takala SL, Coulibaly D, Thera MA, Batchelor AH, Cummings MP, et al. Extreme polymorphism in a vaccine antigen and risk of clinical malaria: implications for vaccine development. Sci Transl Med. 2009;1:2ra5.
    1. Lal AA, Hughes MA, Oliveira DA, Nelson C, Bloland PB, et al. Identification of T-cell determinants in natural immune responses to the Plasmodium falciparum apical membrane antigen (AMA-1) in an adult population exposed to malaria. Infect Immun. 1996;64:1054–1059.
    1. Udhayakumar V, Kariuki S, Kolczack M, Girma M, Roberts JM, et al. Longitudinal study of natural immune responses to the Plasmodium falciparum apical membrane antigen (AMA-1) in a holoendemic region of malaria in western Kenya: Asembo Bay Cohort Project VIII. Am J Trop Med Hyg. 2001;65:100–107.
    1. Bruder JT, Stefaniak ME, Patterson NB, Chen P, Konovalova S, et al. Adenovectors induce functional antibodies capable of potent inhibition of blood stage malaria parasite growth. Vaccine. 2010;28:3201–3210.
    1. Sedegah M, Kim Y, Peters B, McGrath S, Ganeshan H, et al. Identification and localization of minimal MHC-restricted CD8+ T cell epitopes within the Plasmodium falciparum AMA1 protein. Malar J. 2010;9:241.
    1. Sedegah M, Tamminga C, McGrath S, House B, Ganeshan H, et al. Adenovirus 5-Vectored P. falciparum Vaccine Expressing CSP and AMA1. Part A: Safety and Immunogenicity in Seronegative Adults. PLoS One. 2011;6:e24586.
    1. Draper SJ, Heeney JL. Viruses as vaccine vectors for infectious diseases and cancer. Nat Rev Microbiol. 2010;8:62–73.
    1. Barouch DH. Novel adenovirus vector-based vaccines for HIV-1. Curr Opin HIV AIDS. 2010;5:386–390.
    1. Sheehy SH, Duncan CJ, Elias SC, Collins KA, Ewer KJ, et al. Phase Ia Clinical Evaluation of the Plasmodium falciparum Blood-stage Antigen MSP1 in ChAd63 and MVA Vaccine Vectors. Mol Ther 2011
    1. Dutta S, Lee SY, Batchelor AH, Lanar DE. Structural basis of antigenic escape of a malaria vaccine candidate. Proc Natl Acad Sci U S A. 2007;104:12488–12493.
    1. Porter DW, Thompson FM, Berthoud TK, Hutchings CL, Andrews L, et al. A human Phase I/IIa malaria challenge trial of a polyprotein malaria vaccine. Vaccine 2011
    1. Kennedy MC, Wang J, Zhang Y, Miles AP, Chitsaz F, et al. In vitro studies with recombinant Plasmodium falciparum apical membrane antigen 1 (AMA1): production and activity of an AMA1 vaccine and generation of a multiallelic response. Infect Immun. 2002;70:6948–6960.
    1. Roederer M, Nozzi JL, Nason MC. SPICE: exploration and analysis of post-cytometric complex multivariate datasets. Cytometry A. 2011;79:167–174.
    1. Miura K, Orcutt AC, Muratova OV, Miller LH, Saul A, et al. Development and characterization of a standardized ELISA including a reference serum on each plate to detect antibodies induced by experimental malaria vaccines. Vaccine. 2008;26:193–200.
    1. Miura K, Zhou H, Diouf A, Moretz SE, Fay MP, et al. Anti-apical-membrane-antigen-1 antibody is more effective than anti-42-kilodalton-merozoite-surface-protein-1 antibody in inhibiting plasmodium falciparum growth, as determined by the in vitro growth inhibition assay. Clin Vaccine Immunol. 2009;16:963–968.
    1. Capone S, Meola A, Ercole BB, Vitelli A, Pezzanera M, et al. A novel adenovirus type 6 (Ad6)-based hepatitis C virus vector that overcomes preexisting anti-ad5 immunity and induces potent and broad cellular immune responses in rhesus macaques. J Virol. 2006;80:1688–1699.
    1. Dudareva M, Andrews L, Gilbert SC, Bejon P, Marsh K, 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. 2009;27:3501–3504.
    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. Keefer MC, Frey SE, Elizaga M, Metch B, De Rosa SC, et al. A phase I trial of preventive HIV vaccination with heterologous poxviral-vectors containing matching HIV-1 inserts in healthy HIV-uninfected subjects. Vaccine. 2011;29:1948–1958.
    1. Douglas AD, de Cassan SC, Dicks MD, Gilbert SC, Hill AV, et al. Tailoring subunit vaccine immunogenicity: Maximizing antibody and T cell responses by using combinations of adenovirus, poxvirus and protein-adjuvant vaccines against Plasmodium falciparum MSP1. Vaccine. 2010;28:7167–7178.
    1. Goodman AL, Epp C, Moss D, Holder AA, Wilson JM, et al. New candidate vaccines against blood-stage Plasmodium falciparum malaria: prime-boost immunization regimens incorporating human and simian adenoviral vectors and poxviral vectors expressing an optimized antigen based on merozoite surface protein 1. Infect Immun. 2010;78:4601–4612.
    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. Lyke KE, Daou M, Diarra I, Kone A, Kouriba B, et al. Cell-mediated immunity elicited by the blood stage malaria vaccine apical membrane antigen 1 in Malian adults: results of a Phase I randomized trial. Vaccine. 2009;27:2171–2176.
    1. Duncan CJ, Sheehy SH, Ewer KJ, Douglas AD, Collins KA, et al. Impact on Malaria Parasite Multiplication Rates in Infected Volunteers of the Protein-in-Adjuvant Vaccine AMA1-C1/Alhydrogel+CPG 7909. PLoS One. 2011;6:e22271.
    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. Pierce MA, Ellis RD, Martin LB, Malkin E, Tierney E, et al. Phase 1 safety and immunogenicity trial of the Plasmodium falciparum blood-stage malaria vaccine AMA1-C1/ISA 720 in Australian adults. Vaccine. 2010;28:2236–2242.
    1. Mullen GE, Ellis RD, Miura K, Malkin E, Nolan C, et al. Phase 1 trial of AMA1-C1/Alhydrogel plus CPG 7909: an asexual blood-stage vaccine for Plasmodium falciparum malaria. PLoS ONE. 2008;3:e2940.
    1. Ellis RD, Mullen GE, Pierce M, Martin LB, Miura K, et al. A Phase 1 study of the blood-stage malaria vaccine candidate AMA1-C1/Alhydrogel with CPG 7909, using two different formulations and dosing intervals. Vaccine. 2009;27:4104–4109.
    1. Bett AJ, Dubey SA, Mehrotra DV, Guan L, Long R, et al. Comparison of T cell immune responses induced by vectored HIV vaccines in non-human primates and humans. Vaccine. 2010;28:7881–7889.
    1. Tamminga C, Sedegah M, Regis D, Chuang I, Epstein JE, 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. 2011;6:e25868.
    1. Draper SJ, Moore AC, Goodman AL, Long CA, Holder AA, et al. Effective induction of high-titer antibodies by viral vector vaccines. Nat Med. 2008;14:819–821.
    1. Silvie O, Franetich JF, Charrin S, Mueller MS, Siau A, et al. A role for apical membrane antigen 1 during invasion of hepatocytes by Plasmodium falciparum sporozoites. J Biol Chem. 2004;279:9490–9496.
    1. Kusi KA, Faber BW, Thomas AW, Remarque EJ. Humoral immune response to mixed PfAMA1 alleles; multivalent PfAMA1 vaccines induce broad specificity. PLoS One. 2009;4:e8110.
    1. de Cassan SC, Forbes EK, Douglas AD, Milicic A, Singh B, et al. The requirement for potent adjuvants to enhance the immunogenicity and protective efficacy of protein vaccines can be overcome by prior immunization with a recombinant adenovirus. J Immunol. 2011;187:2602–2616.

Source: PubMed

3
購読する