Safety and immunogenicity of heterologous prime-boost immunisation with Plasmodium falciparum malaria candidate vaccines, ChAd63 ME-TRAP and MVA ME-TRAP, in healthy Gambian and Kenyan adults

Caroline Ogwang, Muhammed Afolabi, Domtila Kimani, Ya Jankey Jagne, Susanne H Sheehy, Carly M Bliss, Christopher J A Duncan, Katharine A Collins, Miguel A Garcia Knight, Eva Kimani, Nicholas A Anagnostou, Eleanor Berrie, Sarah Moyle, Sarah C Gilbert, Alexandra J Spencer, Peninah Soipei, Jenny Mueller, Joseph Okebe, Stefano Colloca, Riccardo Cortese, Nicola K Viebig, Rachel Roberts, Katherine Gantlett, Alison M Lawrie, Alfredo Nicosia, Egeruan B Imoukhuede, Philip Bejon, Britta C Urban, Katie L Flanagan, Katie J Ewer, Roma Chilengi, Adrian V S Hill, Kalifa Bojang, Caroline Ogwang, Muhammed Afolabi, Domtila Kimani, Ya Jankey Jagne, Susanne H Sheehy, Carly M Bliss, Christopher J A Duncan, Katharine A Collins, Miguel A Garcia Knight, Eva Kimani, Nicholas A Anagnostou, Eleanor Berrie, Sarah Moyle, Sarah C Gilbert, Alexandra J Spencer, Peninah Soipei, Jenny Mueller, Joseph Okebe, Stefano Colloca, Riccardo Cortese, Nicola K Viebig, Rachel Roberts, Katherine Gantlett, Alison M Lawrie, Alfredo Nicosia, Egeruan B Imoukhuede, Philip Bejon, Britta C Urban, Katie L Flanagan, Katie J Ewer, Roma Chilengi, Adrian V S Hill, Kalifa Bojang

Abstract

Background: Heterologous prime boost immunization with chimpanzee adenovirus 63 (ChAd63) and Modified vaccinia Virus Ankara (MVA) vectored vaccines is a strategy recently shown to be capable of inducing strong cell mediated responses against several antigens from the malaria parasite. ChAd63-MVA expressing the Plasmodium falciparum pre-erythrocytic antigen ME-TRAP (multiple epitope string with thrombospondin-related adhesion protein) is a leading malaria vaccine candidate, capable of inducing sterile protection in malaria naïve adults following controlled human malaria infection (CHMI).

Methodology: We conducted two Phase Ib dose escalation clinical trials assessing the safety and immunogenicity of ChAd63-MVA ME-TRAP in 46 healthy malaria exposed adults in two African countries with similar malaria transmission patterns.

Results: ChAd63-MVA ME-TRAP was shown to be safe and immunogenic, inducing high-level T cell responses (median >1300 SFU/million PBMC).

Conclusions: ChAd63-MVA ME-TRAP is a safe and highly immunogenic vaccine regimen in adults with prior exposure to malaria. Further clinical trials to assess safety and immunogenicity in children and infants and protective efficacy in the field are now warranted.

Trial registration: Pactr.org PACTR2010020001771828 Pactr.org PACTR201008000221638 ClinicalTrials.gov NCT01373879 NCT01373879 ClinicalTrials.gov NCT01379430 NCT01379430.

Conflict of interest statement

Competing Interests: AH is a named inventor on the following patent applications on malaria vectored vaccines and immunization regimes (WO2008/122769, Adenoviral vector encoding malaria antigen; and WO 2008/122811 Novel adenovirus vectors). 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 the authors' adherence to all the PLOS ONE policies on sharing data and materials.

Figures

Figure 1. Clinical Trial Designs.
Figure 1. Clinical Trial Designs.
Trial A = Phase Ib clinical trial in The Gambia, West Africa. Trial B = Phase Ib clinical trial in Kilifi, Kenya, East Africa. IM = intramuscular administration. ID = intradermal administration. In Trial A, 10 volunteers were excluded following screening for the following reasons: severe thrombocytopenia, severe proteinuria, spastic deformity of arm and withdrawal of consent (seven individuals). In Trial B, 14 volunteers were excluded following screening for the following reasons: hypertension (two individuals), positive serology for HIV (two individuals), positive Hepatitis B surface antigen (four individuals), participation in a previous malaria vaccine trial (2 individuals), peptic ulcer disease, allergic disease, recruitment complete (one participant).
Figure 2. Local and systemic AEs deemed…
Figure 2. Local and systemic AEs deemed definitely, probably or possibly related to ChAd63 ME-TRAP.
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. IM = intramuscular. (A) Local AEs post ChAd63 ME-TRAP. (B) Systemic AEs post ChAd63 ME-TRAP.
Figure 3. Local and systemic AEs deemed…
Figure 3. Local and systemic AEs deemed definitely, probably or possibly related to MVA ME-TRAP.
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. IM = intramuscular. ID = Intradermal. (A) Local AEs post MVA ME-TRAP. (B) Systemic AEs post MVA ME-TRAP.
Figure 4. IFN-γ ELISPOT responses to ChAd63-MVA…
Figure 4. IFN-γ ELISPOT responses to ChAd63-MVA ME-TRAP.
(A) Time course of IFN-γ ELISPOT responses to ChAd63-MVA ME-TRAP. Lines show median immune response to high dose (solid line) and lower dose (dashed line) of ChAd63 ME-TRAP in Trial A (grey line) and Trial B (black line). For Trial B medians are for groups with MVA given IM and ID combined. (B) Peak immune response (day 63–7 days post MVA vaccination) to ChAd63 and MVA ME-TRAP stratified by route of administration of MVA, dose of ChAd63 and trial site. Bar represents geometric mean. Circles represent MVA given IM, squares represent MVA given ID. Closed symbols represent 1×1010 vp ChAd63 and open symbols 5×1010 vp ChAd63.

References

    1. WHO (2011) World Malaria Report. World Health Organisation.
    1. Murray CJ, Rosenfeld LC, Lim SS, Andrews KG, Foreman KJ, et al. (2012) Global malaria mortality between 1980 and 2010: a systematic analysis. Lancet 379: 413–431.
    1. Das P, Horton R (2010) Malaria elimination: worthy, challenging, and just possible. Lancet 376: 1515–1517.
    1. Cohen J, Nussenzweig V, Nussenzweig R, Vekemans J, Leach A (2010) From the circumsporozoite protein to the RTS, S/AS candidate vaccine. Hum Vaccin 6: 90–96.
    1. Agnandji ST, Lell B, Soulanoudjingar SS, Fernandes JF, Abossolo BP, et al. (2011) First results of phase 3 trial of RTS,S/AS01 malaria vaccine in African children. N Engl J Med 365: 1863–1875.
    1. Agnandji ST, Lell B, Fernandes JF, Abossolo BP, Methogo BG, et al. (2012) A phase 3 trial of RTS,S/AS01 malaria vaccine in African infants. N Engl J Med 367: 2284–2295.
    1. Hill AV (2012) RTS,S/AS01 malaria vaccine in African children. N Engl J Med 366: 764; author reply 765–766.
    1. Hill AV (2011) Vaccines against malaria. Philos Trans R Soc Lond B Biol Sci 366: 2806–2814.
    1. Stoute JA, Kester KE, Krzych U, Wellde BT, Hall T, et al. (1998) Long-term efficacy and immune responses following immunization with the RTS, S malaria vaccine. J Infect Dis 178: 1139–1144.
    1. Stoute JA, Slaoui M, Heppner DG, Momin P, Kester KE, et al. (1997) A preliminary evaluation of a recombinant circumsporozoite protein vaccine against Plasmodium falciparum malaria. RTS,S Malaria Vaccine Evaluation Group. N Engl J Med 336: 86–91.
    1. Aponte JJ, Aide P, Renom M, Mandomando I, Bassat Q, et al. (2007) Safety of the RTS,S/AS02D candidate malaria vaccine in infants living in a highly endemic area of Mozambique: a double blind randomised controlled phase I/IIb trial. Lancet 370: 1543–1551.
    1. Greenwood B (2011) Immunological correlates of protection for the RTS,S candidate malaria vaccine. Lancet Infect Dis 11: 75–76.
    1. Lalvani A, Moris P, Voss G, Pathan AA, Kester KE, et al. (1999) Potent induction of focused Th1-type cellular and humoral immune responses by RTS,S/SBAS2, a recombinant Plasmodium falciparum malaria vaccine. J Infect Dis 180: 1656–1664.
    1. Schwenk RJ, Richie TL (2011) Protective immunity to pre-erythrocytic stage malaria. Trends Parasitol.
    1. Hoffman SL, Isenbarger D, Long GW, Sedegah M, Szarfman A, et al. (1989) Sporozoite vaccine induces genetically restricted T cell elimination of malaria from hepatocytes. Science 244: 1078–1081.
    1. Khusmith S, Sedegah M, Hoffman SL (1994) Complete protection against Plasmodium yoelii by adoptive transfer of a CD8+ cytotoxic T-cell clone recognizing sporozoite surface protein 2. Infect Immun 62: 2979–2983.
    1. Romero P, Maryanski JL, Corradin G, Nussenzweig RS, Nussenzweig V, et al. (1989) Cloned cytotoxic T cells recognize an epitope in the circumsporozoite protein and protect against malaria. Nature 341: 323–326.
    1. Schofield L, Villaquiran J, Ferreira A, Schellekens H, Nussenzweig R, et al. (1987) Gamma interferon, CD8+ T cells and antibodies required for immunity to malaria sporozoites. Nature 330: 664–666.
    1. Weiss WR, Mellouk S, Houghten RA, Sedegah M, Kumar S, et al. (1990) Cytotoxic T cells recognize a peptide from the circumsporozoite protein on malaria-infected hepatocytes. J Exp Med 171: 763–773.
    1. Hill AV, Reyes-Sandoval A, O′Hara G, Ewer K, Lawrie A, et al. (2010) Prime-boost vectored malaria vaccines: progress and prospects. Hum Vaccin 6: 78–83.
    1. Webster DP, Dunachie S, Vuola JM, Berthoud T, Keating S, et al. (2005) 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 102: 4836–4841.
    1. Gilbert SC, Plebanski M, Harris SJ, Allsopp CE, Thomas R, et al. (1997) A protein particle vaccine containing multiple malaria epitopes. Nat Biotechnol 15: 1280–1284.
    1. Prieur E, Gilbert SC, Schneider J, Moore AC, Sheu EG, et al. (2004) A Plasmodium falciparum candidate vaccine based on a six-antigen polyprotein encoded by recombinant poxviruses. Proc Natl Acad Sci U S A 101: 290–295.
    1. McConkey SJ, Reece WH, Moorthy VS, Webster D, Dunachie S, et al. (2003) Enhanced T-cell immunogenicity of plasmid DNA vaccines boosted by recombinant modified vaccinia virus Ankara in humans. Nat Med 9: 729–735.
    1. Moorthy VS, McConkey S, Roberts M, Gothard P, Arulanantham N, et al. (2003) Safety of DNA and modified vaccinia virus Ankara vaccines against liver-stage P. falciparum malaria in non-immune volunteers. Vaccine 21: 1995–2002.
    1. Vuola JM, Keating S, Webster DP, Berthoud T, Dunachie S, et al. (2005) Differential immunogenicity of various heterologous prime-boost vaccine regimens using DNA and viral vectors in healthy volunteers. J Immunol 174: 449–455.
    1. Webster DP, Dunachie S, McConkey S, Poulton I, Moore AC, et al. (2006) Safety of recombinant fowlpox strain FP9 and modified vaccinia virus Ankara vaccines against liver-stage P. falciparum malaria in non-immune volunteers. Vaccine 24: 3026–3034.
    1. Moorthy VS, Pinder M, Reece WH, Watkins K, Atabani S, et al. (2003) Safety and immunogenicity of DNA/modified vaccinia virus ankara malaria vaccination in African adults. J Infect Dis 188: 1239–1244.
    1. Moorthy VS, Imoukhuede EB, Keating S, Pinder M, Webster D, et al. (2004) 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 189: 2213–2219.
    1. Bejon P, Peshu N, Gilbert SC, Lowe BS, Molyneux CS, et al. (2006) 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 42: 1102–1110.
    1. Bejon P, Mwacharo J, Kai OK, Todryk S, Keating S, et al. (2006) 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 24: 4709–4715.
    1. Bejon P, Mwacharo J, Kai O, Mwangi T, Milligan P, et al. (2006) A phase 2b randomised trial of the candidate malaria vaccines FP9 ME-TRAP and MVA ME-TRAP among children in Kenya. PLoS Clin Trials 1: e29.
    1. Bejon P, Ogada E, Mwangi T, Milligan P, Lang T, et al. (2007) Extended follow-up following a phase 2b randomized trial of the candidate malaria vaccines FP9 ME-TRAP and MVA ME-TRAP among children in Kenya. PLoS One 2: e707.
    1. Imoukhuede EB, Berthoud T, Milligan P, Bojang K, Ismaili J, et al. (2006) Safety and immunogenicity of the malaria candidate vaccines FP9 CS and MVA CS in adult Gambian men. Vaccine 24: 6526–6533.
    1. O′Hara GA, Duncan CJ, Ewer KJ, Collins KA, Elias SC, et al. (2012) Clinical assessment of a recombinant simian adenovirus ChAd63: a potent new vaccine vector. J Infect Dis 205: 772–781.
    1. Greenwood BM, Pickering H (1993) A malaria control trial using insecticide-treated bed nets and targeted chemoprophylaxis in a rural area of The Gambia, west Africa. 1. A review of the epidemiology and control of malaria in The Gambia, west Africa. Trans R Soc Trop Med Hyg 87 Suppl 23–11.
    1. Mwangi TW, Ross A, Snow RW, Marsh K (2005) Case definitions of clinical malaria under different transmission conditions in Kilifi District, Kenya. J Infect Dis 191: 1932–1939.
    1. Okiro EA, Hay SI, Gikandi PW, Sharif SK, Noor AM, et al. (2007) The decline in paediatric malaria admissions on the coast of Kenya. Malar J 6: 151.
    1. Ceesay SJ, Casals-Pascual C, Nwakanma DC, Walther M, Gomez-Escobar N, et al. (2010) Continued decline of malaria in The Gambia with implications for elimination. PLoS One 5: e12242.
    1. Douglas AD, Williams AR, Illingworth JJ, Kamuyu G, Biswas S, et al. (2011) The blood-stage malaria antigen PfRH5 is susceptible to vaccine-inducible cross-strain neutralizing antibody. Nat Commun 2: 601.
    1. Draper SJ, Biswas S, Spencer AJ, Remarque EJ, Capone S, et al. (2010) Enhancing blood-stage malaria subunit vaccine immunogenicity in rhesus macaques by combining adenovirus, poxvirus, and protein-in-adjuvant vaccines. J Immunol 185: 7583–7595.
    1. WHO (2000) Bench Aids for the diagnosis of malaria infections.
    1. Sheehy SH, Duncan CJ, Elias SC, Collins KA, Ewer KJ, et al. (2011) Phase Ia clinical evaluation of the Plasmodium falciparum blood-stage antigen MSP1 in ChAd63 and MVA vaccine vectors. Mol Ther 19: 2269–2276.
    1. Sheehy SH, Duncan CJ, Elias SC, Choudhary P, Biswas S, et al... (2012) ChAd63-MVA-vectored Blood-stage Malaria Vaccines Targeting MSP1 and AMA1: Assessment of Efficacy Against Mosquito Bite Challenge in Humans. Mol Ther.
    1. Duerr A, Huang Y, Buchbinder S, Coombs RW, Sanchez J, et al. (2012) Extended follow-up confirms early vaccine-enhanced risk of HIV acquisition and demonstrates waning effect over time among participants in a randomized trial of recombinant adenovirus HIV vaccine (Step Study). J Infect Dis 206: 258–266.
    1. Dudareva M, Andrews L, Gilbert SC, Bejon P, Marsh K, et al. (2009) 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.
    1. Barnes E, Folgori A, Capone S, Swadling L, Aston S, et al. (2012) Novel adenovirus-based vaccines induce broad and sustained T cell responses to HCV in man. Sci Transl Med 4: 115ra111.
    1. Sheehy SH, Duncan CJ, Elias SC, Biswas S, Collins KA, et al. (2012) 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.

Source: PubMed

3
S'abonner