Safety and tolerability of conserved region vaccines vectored by plasmid DNA, simian adenovirus and modified vaccinia virus ankara administered to human immunodeficiency virus type 1-uninfected adults in a randomized, single-blind phase I trial

Emma-Jo Hayton, Annie Rose, Umar Ibrahimsa, Mariarosaria Del Sorbo, Stefania Capone, Alison Crook, Antony P Black, Lucy Dorrell, Tomáš Hanke, Emma-Jo Hayton, Annie Rose, Umar Ibrahimsa, Mariarosaria Del Sorbo, Stefania Capone, Alison Crook, Antony P Black, Lucy Dorrell, Tomáš Hanke

Abstract

Trial design: HIV-1 vaccine development has advanced slowly due to viral antigenic diversity, poor immunogenicity and recently, safety concerns associated with human adenovirus serotype-5 vectors. To tackle HIV-1 variation, we designed a unique T-cell immunogen HIVconsv from functionally conserved regions of the HIV-1 proteome, which were presented to the immune system using a heterologous prime-boost combination of plasmid DNA, a non-replicating simian (chimpanzee) adenovirus ChAdV-63 and a non-replicating poxvirus, modified vaccinia virus Ankara. A block-randomized, single-blind, placebo-controlled phase I trial HIV-CORE 002 administered for the first time candidate HIV-1- vaccines or placebo to 32 healthy HIV-1/2-uninfected adults in Oxford, UK and elicited high frequencies of HIV-1-specific T cells capable of inhibiting HIV-1 replication in vitro. Here, detail safety and tolerability of these vaccines are reported.

Methods: Local and systemic reactogenicity data were collected using structured interviews and study-specific diary cards. Data on all other adverse events were collected using open questions. Serum neutralizing antibody titres to ChAdV-63 were determined before and after vaccination.

Results: Two volunteers withdrew for vaccine-unrelated reasons. No vaccine-related serious adverse events or reactions occurred during 190 person-months of follow-up. Local and systemic events after vaccination occurred in 27/32 individuals and most were mild (severity grade 1) and predominantly transient (<48 hours). Myalgia and flu-like symptoms were more strongly associated with MVA than ChAdV63 or DNA vectors and more common in vaccine recipients than in placebo. There were no intercurrent HIV-1 infections during follow-up. 2/24 volunteers had low ChAdV-63-neutralizing titres at baseline and 7 increased their titres to over 200 with a median (range) of 633 (231-1533) post-vaccination, which is of no safety concern.

Conclusions: These data demonstrate safety and good tolerability of the pSG2.HIVconsv DNA, ChAdV63.HIVconsv and MVA.HIVconsv vaccines and together with their high immunogenicity support their further development towards efficacy studies.

Trial registration: ClinicalTrials.gov NCT01151319.

Conflict of interest statement

Competing Interests: The authors declare no conflict of interest except for MdS and SC, who were employees of OKAIROS AG from 2007 to 2013. Subsequently OKAIROS AG was acquired by GlaxoSmithKline (GSK), who now hold commercial rights on the work described in the manuscript. Presently MdS and SC have no financial or commercial interests and are not GSK employees. LC reports grants from MRC during the conduct of the study, and TH who reports grants from MRC and European and Developing Countries Clinical Trial Partnership obtained during the conduct of the study and is an inventor on patent WO06123256. This does not alter the authors' adherence to PLOS ONE policies on sharing data and materials.

Figures

Figure 1. CONSORT flow diagram for HIV-CORE…
Figure 1. CONSORT flow diagram for HIV-CORE 002.
Each vaccine and placebo dose was delivered by two injections, one into each arm.
Figure 2. Summary of local reactions.
Figure 2. Summary of local reactions.
Volunteers received vaccines pSG2.HIVconsv DNA (D), ChAdV63.HIVcocnsv (c – low dose and C – standard dose) and MVA.HIVconsv (M) in 4 groups using regimens c, (n  =  2), CM, DDDCM and DDDMC (n  =  10) and in Groups 2–4, were randomized between 8 and 2 recipients of vaccine and placebo, respectively. Local reactions were recorded by the volunteers themselves and assessed and scored by the clinical team. ‘Other’ - one placebo recipient reported a ‘slightly stiff left shoulder’.
Figure 3. Summary of systemic reactions.
Figure 3. Summary of systemic reactions.
Following administration of the pSG2.HIVconsv DNA (D), ChAdV63.HIVconsv (c – low dose and C – standard dose) and MVA.HIVconsv (M) vaccines (Groups 1–4) or placebo (Groups 2, 3 and 4), the volunteers themselves and the clinical team recorded and scored systemic reactions. ‘Other’ encompasses allergic reaction, chills/rigors, headache, vomit, stomach cramps and syncope.
Figure 4. Vaccine-elicited neutralizing antibodies to ChAdV-63.
Figure 4. Vaccine-elicited neutralizing antibodies to ChAdV-63.
Volunteers were given ChAdV63.HIVconsv alone or as a part of a heterologous prime-boost regimen and their sera were tested before and 2 weeks after the ChAdV63.HIVconsv administration for ChAdV-63 neutralizing antibodies. Boxed volunteer numbers indicate placebo recipients.

References

    1. Letourneau S, Im E-J, Mashishi T, Brereton C, Bridgeman A, et al. (2007) Design and pre-clinical evaluation of a universal HIV-1 vaccine. PLoS ONE 2: e984.
    1. Altfeld M, Addo MM, Rosenberg ES, Hecht FM, Lee PK, et al. (2003) Influence of HLA-B57 on clinical presentation and viral control during acute HIV-1 infection. AIDS 17: 2581–2591.
    1. Ferguson AL, Mann JK, Omarjee S, Ndung'u T, Walker BD, et al. (2013) Translating HIV Sequences into Quantitative Fitness Landscapes Predicts Viral Vulnerabilities for Rational Immunogen Design. Immunity 38: 606–617.
    1. Kelleher AD, Long C, Holmes EC, Allen RL, Wilson J, et al. (2001) Clustered mutations in HIV-1 gag are consistently required for escape from HLA-B27-restricted cytotoxic T lymphocyte responses. J Exp Med 193: 375–386.
    1. Leslie AJ, Pfafferott KJ, Chetty P, Draenert R, Addo MM, et al. (2004) HIV evolution: CTL escape mutation and reversion after transmission. Nat Med 10: 282–289.
    1. Colloca S, Barnes E, Folgori A, Ammendola V, Capone S, et al. (2012) Vaccine vectors derived from a large collection of simian adenoviruses induce potent cellular immunity across multiple species. Sci Transl Med 4: 1–9.
    1. Rosario M, Bridgeman A, Quakkelaar ED, Quigley MF, Hill BJ, et al. (2010) Long peptides induce polyfunctional T cells against conserved regions of HIV-1 with superior breadth to single-gene vaccines in macaques. Eur J Immunol 40: 1973–1984.
    1. Knudsen ML, Mbewe-Mvula A, Rosario M, Johansson DX, Kakoulidou M, et al. (2012) Superior induction of T cell responses to conserved HIV-1 regions by electroporated alphavirus replicon DNA compared to conventional plasmid DNA vaccine. J Virol 86: 4082–4090.
    1. Rosario M, Borthwick N, Stewart-Jones GB, Mbewe-Mwula A, Bridgeman A, et al. (2012) Prime-boost regimens with adjuvanted synthetic long peptides elicit T cells and antibodies to conserved regions of HIV-1 in macaques. AIDS 26: 275–284.
    1. Ondondo B, Brennan C, Nicosia A, Crome S, Hanke T (2013) Absence of systemic toxicity changes following intramuscular administration of novel pSG2.HIVconsv DNA, ChAdV63.HIVconsv and MVA.HIVconsv vaccines to BALB/c mice Vaccine. 31: 5594–5601.
    1. Borthwick N, Ahmed T, Ondondo B, Hayes P, Rose A, et al. (2014) Vaccine-elicited human T cells recognizing conserved protein regions inhibit HIV-1. Mol Ther 22: 464–475.
    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. Cebere I, Dorrell L, McShane H, Simmons A, McCormack S, et al. (2006) Phase I clinical trial safety of DNA- and modified virus Ankara-vectored human immunodeficiency virus type 1 (HIV-1) vaccines administered alone and in a prime-boost regime to healthy HIV-1-uninfected volunteers. Vaccine 24: 417–425.
    1. Currier JR, Ngauy V, de Souza MS, Ratto-Kim S, Cox JH, et al. (2010) Phase I safety and immunogenicity evaluation of MVA-CMDR, a multigenic, recombinant modified vaccinia Ankara-HIV-1 vaccine candidate. PLoS One 5: e13983.
    1. Dorrell L, Williams P, Suttill A, Brown D, Roberts J, et al. (2007) Safety and tolerability of recombinant modified vaccinia virus Ankara expressing an HIV-1 gag/multiepitope immunogen (MVA.HIVA) in HIV-1-infected persons receiving combination antiretroviral therapy. Vaccine 25: 3277–3283.
    1. Graham B, Koup R, Roederer M, Bailer R, Enama M, et al. (2006) Phase I Safety and Immunogenicity Evaluation of a Multiclade HIV-1 Candidate DNA Vaccine. J Infect Dis 194: 1650–1660.
    1. Jaoko W, Nakwagala FN, Anzala O, Manyonyi GO, Birungi J, et al. (2008) Safety and immunogenicity of recombinant low-dosage HIV-1 A vaccine candidates vectored by plasmid pTHr DNA or modified vaccinia virus Ankara (MVA) in humans in East Africa. Vaccine 26: 2788–2795.
    1. McCormack S, Stohr W, Barber T, Bart PA, Harari A, et al. (2008) EV02: a Phase I trial to compare the safety and immunogenicity of HIV DNA-C prime-NYVAC-C boost to NYVAC-C alone. Vaccine 26: 3162–3174.
    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. 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.
    1. Sheehy SH, Duncan CJ, Elias SC, Choudhary P, Biswas S, et al. (2013) ChAd63-MVA-vectored blood-stage malaria vaccines targeting MSP1 and AMA1: assessment of efficacy against mosquito bite challenge in humans. Mol Ther 20: 2355–2368.
    1. Sheehy SH, Duncan CJ, Elias SC, Collins KA, Ewer KJ, et al. (2012) Phase Ia clinical evaluation of the Plasmodium falciparum blood-stage antigen MSP1 in ChAd63 and MVA vaccine vectors. Mol Ther 19: 2269–2276.
    1. Ogwang C, Afolabi M, Kimani D, Jagne YJ, Sheehy SH, et al. (2013) 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. PLoS ONE 8: e57726.
    1. Aste-Amezaga M, Bett AJ, Wang F, Casimiro DR, Antonello JM, et al. (2004) Quantitative adenovirus neutralization assays based on the secreted alkaline phosphatase reporter gene: application in epidemiologic studies and in the design of adenovector vaccines. Hum Gene Ther 15: 293–304.
    1. Buchbinder SP, Mehrotra DV, Duerr A, Fitzgerald DW, Mogg R, et al. (2008) 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.
    1. Paris R, Kuschner RA, Binn L, Thomas SJ, Colloca S, et al... (2014) Adenovirus type 4 and type 7 vaccination or type 4 respiratory infection elicit minimal cross-reactive antibody to non-human adenovirus vaccine vectors. Clin Vaccine Immunol.
    1. Hanke T (2014) Conserved immunogens in prime-bost strategies for te next-generation HIV-1 vaccines. Expert Opin Biol Ther 14: 601–616.
    1. Estcourt MJ, McMichael AJ, Hanke T (2004) DNA vaccines against human immunodeficiency virus type 1. Immunol Rev 199: 144–155.
    1. Gilbert SC, Moorthy VS, Andrews L, Pathan AA, McConkey SJ, et al. (2006) Synergistic DNA-MVA prime-boost vaccination regimes for malaria and tuberculosis. Vaccine 24: 4554–4561.
    1. Im E-J, Hanke T (2004) MVA as a vector for vaccines against HIV-1. Expert Rev Vaccines 3: S89–97.
    1. Mehendale S, Thakar M, Sahay S, Kumar M, Shete A, et al. (2013) Safety and immunogenicity of DNA and MVA HIV-1 subtype C vaccine prime-boost regimens: a phase I randomised Trial in HIV-uninfected Indian volunteers. PLoS One 8: e55831.
    1. Ramanathan VD, Kumar M, Mahalingam J, Sathyamoorthy P, Narayanan PR, et al. (2009) A Phase 1 study to evaluate the safety and immunogenicity of a recombinant HIV type 1 subtype C-modified vaccinia Ankara virus vaccine candidate in Indian volunteers. AIDS Res Hum Retroviruses 25: 1107–1116.
    1. Sutter G, Staib C (2003) Vaccinia vectors as candidate vaccines: the development of modified vaccinia virus Ankara for antigen delivery. Curr Drug Targets Infect Disord 3: 263–271.
    1. Gilbert P, Follmann D (2013) Presentation of meta-analysis. Mini-Summit on Adenovirus Platforms for HIV Vaccines. Bethesda, Maryland, U.S.A. .
    1. Duerr A, Huang Y, Buchbinder S, Coombs RW, Sanchez J, et al. (2013) Reply to Richie and Villasante. J Infect Dis 207: 690–692.
    1. Duerr A, Wasserheit JN, Corey L (2006) HIV vaccines: new frontiers in vaccine development. Clin Infect Dis 43: 500–511.
    1. Gray GE, Moodie Z, Metch B, Gilbert PB, Bekker LG, et al... (2014) Recombinant adenovirus type 5 HIV gag/pol/nef vaccine in South Africa: unblinded, long-term follow-up of the phase 2b HVTN 503/Phambili study. Lancet Infect Dis.
    1. Richie TL, Villasante EF (2013) Use of adenovirus serotype 5 vaccine vectors in seropositive, uncircumcised men: safety lessons from the step trial. J Infect Dis 207: 689–690.
    1. Hammer SM, Sobieszczyk ME, Janes H, Karuna ST, Mulligan MJ, et al... (2013) Efficacy Trial of a DNA/rAd5 HIV-1 Preventive Vaccine. N Engl J Med.
    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. Hutnick NA, Carnathan D, Demers K, Makedonas G, Ertl HC, et al. (2010) Adenovirus-specific human T cells are pervasive, polyfunctional, and cross-reactive. Vaccine 28: 1932–1941.
    1. Frahm N, DeCamp AC, Friedrich DP, Carter DK, Defawe OD, et al. (2012) Human adenovirus-specific T cells modulate HIV-specific T cell responses to an Ad5-vectored HIV-1 vaccine. J Clin Invest 122: 359–367.
    1. Keefer MC, Gilmour J, Hayes P, Gill D, Kopycinski J, et al. (2012) A phase I double blind, placebo-controlled, randomized study of a multigenic HIV-1 adenovirus subtype 35 vector vaccine in healthy uninfected adults. PLoS One 7: e41936.

Source: PubMed

3
Abonnere