Safety and immunogenicity of an FP9-vectored candidate tuberculosis vaccine (FP85A), alone and with candidate vaccine MVA85A in BCG-vaccinated healthy adults: a phase I clinical trial

Rosalind Rowland, Ansar A Pathan, Iman Satti, Ian D Poulton, Magali M L Matsumiya, Megan Whittaker, Angela M Minassian, Geraldine A O'Hara, Matthew Hamill, Janet T Scott, Stephanie A Harris, Hazel C Poyntz, Cynthia Bateman, Joel Meyer, Nicola Williams, Sarah C Gilbert, Alison M Lawrie, Adrian V S Hill, Helen McShane, Rosalind Rowland, Ansar A Pathan, Iman Satti, Ian D Poulton, Magali M L Matsumiya, Megan Whittaker, Angela M Minassian, Geraldine A O'Hara, Matthew Hamill, Janet T Scott, Stephanie A Harris, Hazel C Poyntz, Cynthia Bateman, Joel Meyer, Nicola Williams, Sarah C Gilbert, Alison M Lawrie, Adrian V S Hill, Helen McShane

Abstract

The safety and immunogenicity of a new candidate tuberculosis (TB) vaccine, FP85A was evaluated alone and in heterologous prime-boost regimes with another candidate TB vaccine, MVA85A. This was an open label, non-controlled, non-randomized Phase I clinical trial. Healthy previously BCG-vaccinated adult subjects were enrolled sequentially into three groups and vaccinated with FP85A alone, or both FP85A and MVA85A, with a four week interval between vaccinations. Passive and active data on adverse events were collected. Immunogenicity was evaluated by Enzyme Linked Immunospot (ELISpot), flow cytometry and Enzyme Linked Immunosorbent assay (ELISA). Most adverse events were mild and there were no vaccine-related serious adverse events. FP85A vaccination did not enhance antigen 85A-specific cellular immunity. When MVA85A vaccination was preceded by FP85A vaccination, cellular immune responses were lower compared with when MVA85A vaccination was the first immunisation. MVA85A vaccination, but not FP85A vaccination, induced anti-MVA IgG antibodies. Both MVA85A and FP85A vaccinations induced anti-FP9 IgG antibodies. In conclusion, FP85A vaccination was well tolerated but did not induce antigen-specific cellular immune responses. We hypothesize that FP85A induced anti-FP9 IgG antibodies with cross-reactivity for MVA85A, which may have mediated inhibition of the immune response to subsequent MVA85A. ClinicalTrials.gov identification number: NCT00653770.

Keywords: heterologous prime-boost regimes; phase I clinical trial; poxvirus-vectored subunit vaccines; tuberculosis vaccines.

Figures

https://www.ncbi.nlm.nih.gov/pmc/articles/instance/3667946/bin/hvi-9-50-g1.jpg
Figure 1. CONSORT flow diagram. Figure 1 shows the flow of subjects through the trial. Subjects were not randomized, but allocated sequentially into Group 1, then Group 2, then Group 3, in order of enrolment. All enrolled subjects completed follow up.
https://www.ncbi.nlm.nih.gov/pmc/articles/instance/3667946/bin/hvi-9-50-g2.jpg
Figure 2. IFNγ ELISpot responses to 85A and soluble serum cytokines. (A) Longitudinal IFNγ ELISpot responses to the single 85A peptide pool. Each dot represents an individual subjects’ response and median responses are connected by lines. Group 1 = FP85A vaccination week 0; Group 2 = MVA85A vaccination week 0, FP85A vaccination week four; Group 3 = FP85A vaccination week 0, MVA85A vaccination week four. No increases in responses to antigen 85A were seen after FP85A vaccination in Group 1. MVA85A vaccination induced strong IFNγ T cell responses to antigen 85A in Group 2, which were maintained throughout the 52 week follow up, but were not boosted by subsequent FP85A vaccination at week four. There were no responses after FP85A vaccination in Group 3, but subsequent MVA85A vaccination at week four induced moderate IFNγ T cell responses to antigen 85A. (B) Proportion of subjects with detectable soluble serum cytokines. The bars show the proportion of subjects within each group, in whose serum, any cytokines were detectable. Group 1 = FP85A vaccination week 0; Group 2 = MVA85A vaccination week 0, FP85A vaccination week four; Group 3 = FP85A vaccination week 0, MVA85A vaccination week four; days = days since enrolment. Serum IFNγ and TNFα were detected in no more than one subject’s serum at any one time point. IL-8 was detected in all Group 2 subjects’ serum by day seven (one week after MVA85A vaccination).
https://www.ncbi.nlm.nih.gov/pmc/articles/instance/3667946/bin/hvi-9-50-g3.jpg
Figure 3. IFNγ ELISpot responses to Vaccinia CD4+ and CD8+ T cell epitopes. (A) Longitudinal IFNγ ELISpot responses to known Vaccinia CD4 and CD8 epitopes Each dot represents an individual subjects’ response and median responses are connected by lines. Group 1 = FP85A vaccination week 0; Group 2 = MVA85A vaccination week 0, FP85A vaccination week four; Group 3 = FP85A vaccination week 0, MVA85A vaccination week four. Minimal, transient increases in Vaccinia CD8+ responses compared with baseline were observed after FP85A vaccination in Group 1. Transient responses to Vaccinia CD4+ and CD8+ epitopes were observed after MVA85A, but not FP85A vaccination in Group 2. In Group 3, responses to Vaccinia epitopes did not significantly increase after either FP85A or MVA85A vaccinations. (B) Correlation between IFNγ ELISpot responses to Vaccinia CD4+ and CD8+ at the time of MVA85A vaccination and IFNγ ELISpot responses to antigen 85A one week after MVA85A vaccination. Each dot represents an individual subjects’ responses. Pre-vaccination responses were from the day of MVA85A vaccination (Group 2 = week 0; Group 3 = week four). Post-vaccination responses were from samples taken one week after MVA5A vaccination (Group 2 = week one; Group 3 = week five). Circles = responses to Vaccinia CD4+ epitopes; diamonds = responses to Vaccinia CD8+ epitopes. No relationship between pre-vaccination anti-vector responses (y axes) and post-vaccination T cell responses (x axes) for MVA85A vaccination was found.
https://www.ncbi.nlm.nih.gov/pmc/articles/instance/3667946/bin/hvi-9-50-g4.jpg
Figure 4. Serum IgG ELISA responses to r85A, MVA and FP9. (A) Longitudinal r85A IgG, FP9 IgG and MVA IgG responses detected by ELISA Each dot represents an individual subjects’ response and median responses are connected by lines. Group 1 = FP85A vaccination week 0; Group 2 = MVA85A vaccination week 0, FP85A vaccination week four; Group 3 = FP85A vaccination week 0, MVA85A vaccination week four. Anti-vector antibody responses were generally stronger than antigen-specific anti-85A IgG responses. In Group 1, FP85A vaccination induced an FP9 IgG response, but no MVA IgG antibodies. In Group 2, MVA85A vaccination induced FP9 IgG and MVA IgG responses. Subsequent FP85A vaccination boosted the FP9 IgG response but did not boost the MVA IgG response. In Group 3, MVA85A vaccination induced an MVA IgG response, but did not boost the FP9 IgG response to prior FP85A vaccination. (B) Correlation between FP9 IgG and MVA IgG levels detectable by ELISA at the time of MVA85A vaccination and IFNγ ELISpot responses to antigen 85A one week after MVA85A vaccination. Each dot represents an individual subjects’ responses. Pre-vaccination responses were from the day of MVA85A vaccination (Group 2 = week 0; Group 3 = week four). Post-vaccination responses were from samples taken after MVA5A vaccination (Group 2 = week four; Group 3 = week 12). Circles = FP9 IgG levels; diamonds = MVA IgG levels. There were trends toward negative correlations between pre-MVA85A FP9 and MVA IgG levels and post-MVA85A IFNγ ELISpot responses to single pool 85A in Groups 2 and 3.

References

    1. Global Tuberculosis Control. WHO report 2011. Geneva: World Health Organisation, 2011.
    1. Trunz BB, Fine P, Dye C. Effect of BCG vaccination on childhood tuberculous meningitis and miliary tuberculosis worldwide: a meta-analysis and assessment of cost-effectiveness. Lancet. 2006;367:1173–80. doi: 10.1016/S0140-6736(06)68507-3.
    1. Colditz GA, Brewer TF, Berkey CS, Wilson ME, Burdick E, Fineberg HV, et al. Efficacy of BCG vaccine in the prevention of tuberculosis. Meta-analysis of the published literature. JAMA. 1994;271:698–702. doi: 10.1001/jama.1994.03510330076038.
    1. Revised BCG vaccination guidelines for infants at risk for HIV infection. Wkly Epidemiol Rec. 2007;82:193–6.
    1. Flynn JL. Immunology of tuberculosis and implications in vaccine development. Tuberculosis (Edinb) 2004;84:93–101. doi: 10.1016/j.tube.2003.08.010.
    1. Vuola JM, Keating S, Webster DP, Berthoud T, Dunachie S, Gilbert SC, et al. Differential immunogenicity of various heterologous prime-boost vaccine regimens using DNA and viral vectors in healthy volunteers. J Immunol. 2005;174:449–55.
    1. Scriba TJ, Tameris M, Smit E, van der Merwe L, Hughes EJ, Kadira B, et al. A phase IIa trial of the new tuberculosis vaccine, MVA85A, in HIV- and/or Mycobacterium tuberculosis-infected adults. Am J Respir Crit Care Med. 2012;185:769–78. doi: 10.1164/rccm.201108-1548OC.
    1. Minassian AM, Rowland R, Beveridge NE, Poulton ID, Satti I, Harris S, et al. A Phase I study evaluating the safety and immunogenicity of MVA85A, a candidate TB vaccine, in HIV-infected adults. BMJ Open. 2011;1:e000223. doi: 10.1136/bmjopen-2011-000223.
    1. Ota MO, Odutola AA, Owiafe PK, Donkor S, Owolabi OA, Brittain NJ, et al. Immunogenicity of the tuberculosis vaccine MVA85A is reduced by coadministration with EPI vaccines in a randomized controlled trial in Gambian infants. Sci Transl Med. 2011;3:88ra56. doi: 10.1126/scitranslmed.3002461.
    1. Scriba TJ, Tameris M, Mansoor N, Smit E, van der Merwe L, Mauff K, et al. Dose-finding study of the novel tuberculosis vaccine, MVA85A, in healthy BCG-vaccinated infants. J Infect Dis. 2011;203:1832–43. doi: 10.1093/infdis/jir195.
    1. Sander CR, Pathan AA, Beveridge NE, Poulton I, Minassian A, Alder N, et al. Safety and immunogenicity of a new tuberculosis vaccine, MVA85A, in Mycobacterium tuberculosis-infected individuals. Am J Respir Crit Care Med. 2009;179:724–33. doi: 10.1164/rccm.200809-1486OC.
    1. Brookes RH, Hill PC, Owiafe PK, Ibanga HB, Jeffries DJ, Donkor SA, et al. Safety and immunogenicity of the candidate tuberculosis vaccine MVA85A in West Africa. PLoS One. 2008;3:e2921. doi: 10.1371/journal.pone.0002921.
    1. Hawkridge T, Scriba TJ, Gelderbloem S, Smit E, Tameris M, Moyo S, et al. Safety and immunogenicity of a new tuberculosis vaccine, MVA85A, in healthy adults in South Africa. J Infect Dis. 2008;198:544–52. doi: 10.1086/590185.
    1. Beveridge NE, Price DA, Casazza JP, Pathan AA, Sander CR, Asher TE, et al. Immunisation with BCG and recombinant MVA85A induces long-lasting, polyfunctional Mycobacterium tuberculosis-specific CD4+ memory T lymphocyte populations. Eur J Immunol. 2007;37:3089–100. doi: 10.1002/eji.200737504.
    1. Pathan AA, Sander CR, Fletcher HA, Poulton I, Alder NC, Beveridge NE, et al. Boosting BCG with recombinant modified vaccinia ankara expressing antigen 85A: different boosting intervals and implications for efficacy trials. PLoS One. 2007;2:e1052. doi: 10.1371/journal.pone.0001052.
    1. McShane H, Pathan AA, Sander CR, Keating SM, Gilbert SC, Huygen K, et al. Recombinant modified vaccinia virus Ankara expressing antigen 85A boosts BCG-primed and naturally acquired antimycobacterial immunity in humans. Nat Med. 2004;10:1240–4. doi: 10.1038/nm1128.
    1. Williams A, Goonetilleke NP, McShane H, Clark SO, Hatch G, Gilbert SC, et al. Boosting with poxviruses enhances Mycobacterium bovis BCG efficacy against tuberculosis in guinea pigs. Infect Immun. 2005;73:3814–6. doi: 10.1128/IAI.73.6.3814-3816.2005.
    1. Walther M, Woodruff J, Edele F, Jeffries D, Tongren JE, King E, et al. Innate immune responses to human malaria: heterogeneous cytokine responses to blood-stage Plasmodium falciparum correlate with parasitological and clinical outcomes. J Immunol. 2006;177:5736–45.
    1. Webster DP, Dunachie S, McConkey S, Poulton I, Moore AC, Walther M, et al. Safety of recombinant fowlpox strain FP9 and modified vaccinia virus Ankara vaccines against liver-stage P. falciparum malaria in non-immune volunteers. Vaccine. 2006;24:3026–34. doi: 10.1016/j.vaccine.2005.10.058.
    1. Rowland R, Brittain N, Poulton ID, Minassian AM, Sander C, Porter DW, et al. A review of the tolerability of the candidate TB vaccine, MVA85A compared with BCG and Yellow Fever vaccines, and correlation between MVA85A vaccine reactogenicity and cellular immunogenicity. Trials in Vaccinology. 2012;1:27–35. doi: 10.1016/j.trivac.2012.07.001.
    1. Keefer MC, Frey SE, Elizaga M, Metch B, De Rosa SC, Barroso PF, et al. NIAID HIV Vaccine Trials Network A phase I trial of preventive HIV vaccination with heterologous poxviral-vectors containing matching HIV-1 inserts in healthy HIV-uninfected subjects. Vaccine. 2011;29:1948–58. doi: 10.1016/j.vaccine.2010.12.104.
    1. Porter DW, Thompson FM, Berthoud TK, Hutchings CL, Andrews L, Biswas S, et al. A human Phase I/IIa malaria challenge trial of a polyprotein malaria vaccine. Vaccine. 2011;29:7514–22. doi: 10.1016/j.vaccine.2011.03.083.
    1. Currier JR, Ngauy V, de Souza MS, Ratto-Kim S, Cox JH, Polonis VR, et al. Phase I safety and immunogenicity evaluation of MVA-CMDR, a multigenic, recombinant modified vaccinia Ankara-HIV-1 vaccine candidate. PLoS One. 2010;5:e13983. doi: 10.1371/journal.pone.0013983.
    1. Guerra S, López-Fernández LA, Conde R, Pascual-Montano A, Harshman K, Esteban M. Microarray analysis reveals characteristic changes of host cell gene expression in response to attenuated modified vaccinia virus Ankara infection of human HeLa cells. J Virol. 2004;78:5820–34. doi: 10.1128/JVI.78.11.5820-5834.2004.
    1. Sawant KV, Cho H, Lyons M, Ly LH, McMurray DN. Guinea pig neutrophil-macrophage interactions during infection with Mycobacterium tuberculosis. Microbes Infect. 2010;12:828–37. doi: 10.1016/j.micinf.2010.05.009.
    1. Sterling TR, Dorman SE, Chaisson RE, Ding L, Hackman J, Moore K, et al. Human immunodeficiency virus-seronegative adults with extrapulmonary tuberculosis have abnormal innate immune responses. Clin Infect Dis. 2001;33:976–82. doi: 10.1086/322670.
    1. Tuberculosis. Green Book: Department of Health, 2011:403-7.
    1. Laidlaw SM, Skinner MA. Comparison of the genome sequence of FP9, an attenuated, tissue culture-adapted European strain of Fowlpox virus, with those of virulent American and European viruses. J Gen Virol. 2004;85:305–22. doi: 10.1099/vir.0.19568-0.
    1. Qingzhong Y, Barrett T, Brown TD, Cook JK, Green P, Skinner MA, et al. Protection against turkey rhinotracheitis pneumovirus (TRTV) induced by a fowlpox virus recombinant expressing the TRTV fusion glycoprotein (F) Vaccine. 1994;12:569–73. doi: 10.1016/0264-410X(94)90319-0.
    1. McShane H, Behboudi S, Goonetilleke N, Brookes R, Hill AV. Protective immunity against Mycobacterium tuberculosis induced by dendritic cells pulsed with both CD8(+)- and CD4(+)-T-cell epitopes from antigen 85A. Infect Immun. 2002;70:1623–6. doi: 10.1128/IAI.70.3.1623-1626.2002.
    1. Howles S, Guimarães-Walker A, Yang H, Hancock G, di Gleria K, Tarragona-Fiol T, et al. Vaccination with a modified vaccinia virus Ankara (MVA)-vectored HIV-1 immunogen induces modest vector-specific T cell responses in human subjects. Vaccine. 2010;28:7306–12. doi: 10.1016/j.vaccine.2010.08.077.
    1. Meyer VS, Kastenmuller W, Gasteiger G, Franz-Wachtel M, Lamkemeyer T, Rammensee HG, et al. Long-term immunity against actual poxviral HLA ligands as identified by differential stable isotope labeling. J Immunol. 2008;181:6371–83.
    1. Strug I, Calvo-Calle JM, Green KM, Cruz J, Ennis FA, Evans JE, et al. Vaccinia peptides eluted from HLA-DR1 isolated from virus-infected cells are recognized by CD4+ T cells from a vaccinated donor. J Proteome Res. 2008;7:2703–11. doi: 10.1021/pr700780x.
    1. Calvo-Calle JM, Strug I, Nastke MD, Baker SP, Stern LJ. Human CD4+ T cell epitopes from vaccinia virus induced by vaccination or infection. PLoS Pathog. 2007;3:1511–29. doi: 10.1371/journal.ppat.0030144.
    1. Terajima M, Cruz J, Leporati AM, Demkowicz WE, Jr., Kennedy JS, Ennis FA. Identification of vaccinia CD8+ T-cell epitopes conserved among vaccinia and variola viruses restricted by common MHC class I molecules, HLA-A2 or HLA-B7. Hum Immunol. 2006;67:512–20. doi: 10.1016/j.humimm.2005.12.004.
    1. Oseroff C, Kos F, Bui HH, Peters B, Pasquetto V, Glenn J, et al. HLA class I-restricted responses to vaccinia recognize a broad array of proteins mainly involved in virulence and viral gene regulation. Proc Natl Acad Sci U S A. 2005;102:13980–5. doi: 10.1073/pnas.0506768102.
    1. Drexler I, Staib C, Kastenmuller W, Stevanović S, Schmidt B, Lemonnier FA, et al. Identification of vaccinia virus epitope-specific HLA-A*0201-restricted T cells and comparative analysis of smallpox vaccines. Proc Natl Acad Sci U S A. 2003;100:217–22. doi: 10.1073/pnas.262668999.

Source: PubMed

Подписаться