Preliminary assessment of the efficacy of a T-cell-based influenza vaccine, MVA-NP+M1, in humans

Patrick J Lillie, Tamara K Berthoud, Timothy J Powell, Teresa Lambe, Caitlin Mullarkey, Alexandra J Spencer, Matthew Hamill, Yanchun Peng, Marie-Eve Blais, Christopher J A Duncan, Susanne H Sheehy, Tom Havelock, Saul N Faust, Rob Lambkin Williams, Anthony Gilbert, John Oxford, Tao Dong, Adrian V S Hill, Sarah C Gilbert, Patrick J Lillie, Tamara K Berthoud, Timothy J Powell, Teresa Lambe, Caitlin Mullarkey, Alexandra J Spencer, Matthew Hamill, Yanchun Peng, Marie-Eve Blais, Christopher J A Duncan, Susanne H Sheehy, Tom Havelock, Saul N Faust, Rob Lambkin Williams, Anthony Gilbert, John Oxford, Tao Dong, Adrian V S Hill, Sarah C Gilbert

Abstract

Background: The novel influenza vaccine MVA-NP+M1 is designed to boost cross-reactive T-cell responses to internal antigens of the influenza A virus that are conserved across all subtypes, providing protection against both influenza disease and virus shedding against all influenza A viruses. Following a phase 1 clinical study that demonstrated vaccine safety and immunogenicity, a phase 2a vaccination and influenza challenge study has been conducted in healthy adult volunteers.

Methods: Volunteers with no measurable serum antibodies to influenza A/Wisconsin/67/2005 received either a single vaccination with MVA-NP+M1 or no vaccination. T-cell responses to the vaccine antigens were measured at enrollment and again prior to virus challenge. All volunteers underwent intranasal administration of influenza A/Wisconsin/67/2005 while in a quarantine unit and were monitored for symptoms of influenza disease and virus shedding.

Results: Volunteers had a significantly increased T-cell response to the vaccine antigens following a single dose of the vaccine, with an increase in cytolytic effector molecules. Intranasal influenza challenge was undertaken without safety issues. Two of 11 vaccinees and 5 of 11 control subjects developed laboratory-confirmed influenza (symptoms plus virus shedding). Symptoms of influenza were less pronounced in the vaccinees and there was a significant reduction in the number of days of virus shedding in those vaccinees who developed influenza (mean, 1.09 days in controls, 0.45 days in vaccinees, P = .036).

Conclusions: This study provides the first demonstration of clinical efficacy of a T-cell-based influenza vaccine and indicates that further clinical development should be undertaken.

Clinical trials registration: NCT00993083.

Figures

Figure 1.
Figure 1.
Ex vivo interferon γ enzyme-linked immunosorbent spot assay responses to nucleoprotein (NP) and matrix protein 1 (M1). The graph represents the summed response to NP and M1 antigens in vaccinees (circles) and controls (squares) at the relevant time points; lines represent the median per group and open symbols represent subjects who developed laboratory-confirmed influenza. Control subjects were not assayed at day 0 or day 21. Vaccination took place on day 0 and influenza challenge on day 30. Data were analyzed with a Kruskal-Wallis 1-way analysis of variance with selected pairs of data analyzed with a Dunn positive test. No significant difference between the median response in the vaccinated and control group was observed at time of screening (day 0 for vaccinees, day 29 for controls). A significant increase in the response was observed in vaccinees between days 0 and 21 and days 0 and 29 (< .001,< .05, respectively). A significant difference between vaccinees and controls was observed at day 29 (P < .05). Abbreviations: PBMC, peripheral blood mononuclear cell; SFU, spot-forming units.
Figure 2.
Figure 2.
Responses to M158–66 in human leukocyte antigen A2–positive volunteers. Whole blood drawn 1 day prior to virus challenge was labeled for tetramer (A*0201/GILGFVFTL) followed by perforin or granzyme A staining. Values shown are the percentage of CD8+ T cells or Tet+ cells; individuals are shown as a single point with lines representing the median per group. Open symbols represent samples from volunteers who subsequently developed laboratory-confirmed influenza. For each marker the data were analyzed with an unpaired t test; P values are shown for statistically significant differences between vaccinees and controls.
Figure 3.
Figure 3.
Total of symptom scores at each time point following challenge (A) or total grade 2 and 3 symptom and examination scores (B) for vaccinees (circles) and controls (squares), with the group mean indicated by a line. Open symbols denote subjects who developed laboratory-confirmed influenza after challenge.

References

    1. Osterholm MT, Kelley NS, Sommer A, Belongia EA. Efficacy and effectiveness of influenza vaccines: a systematic review and meta-analysis. Lancet Infect Dis. 2012;12:36–44.
    1. Kresse H, Rovini H. Influenza vaccine market dynamics. Nat Rev Drug Discov. 2009;8:841–2.
    1. McMichael AJ, Gotch F, Cullen P, Askonas B, Webster RG. The human cytotoxic T cell response to influenza A vaccination. Clin Exp Immunol. 1981;43:276–84.
    1. Epstein SL. Prior H1N1 influenza infection and susceptibility of Cleveland Family Study participants during the H2N2 pandemic of 1957: an experiment of nature. J Infect Dis. 2006;193:49–53.
    1. He XS, Holmes TH, Zhang C, et al. Cellular immune responses in children and adults receiving inactivated or live attenuated influenza vaccines. J Virol. 2006;80:11756–66.
    1. Lee LY, Ha do LA, Simmons C, et al. Memory T cells established by seasonal human influenza A infection cross-react with avian influenza A (H5N1) in healthy individuals. J Clin Invest. 2008;118:3478–90.
    1. Berthoud TK, Hamill M, Lillie PJ, et al. Potent CD8+ T-cell immunogenicity in humans of a novel heterosubtypic influenza A vaccine, MVA-NP+M1. Clin Infect Dis. 2011;52:1–7.
    1. Makedonas G, Banerjee PP, Pandey R, et al. Rapid up-regulation and granule-independent transport of perforin to the immunological synapse define a novel mechanism of antigen-specific CD8+ T cell cytotoxic activity. J Immunol. 2009;182:5560–9.
    1. Forrest BD, Pride MW, Dunning AJ, et al. Correlation of cellular immune responses with protection against culture-confirmed influenza virus in young children. Clin Vaccine Immunol. 2008;15:1042–53.
    1. Laddy DJ, Yan J, Kutzler M, et al. Heterosubtypic protection against pathogenic human and avian influenza viruses via in vivo electroporation of synthetic consensus DNA antigens. PLoS One. 2008;3:e2517.
    1. Wesley RD, Tang M, Lager KM. Protection of weaned pigs by vaccination with human adenovirus 5 recombinant viruses expressing the hemagglutinin and the nucleoprotein of H3N2 swine influenza virus. Vaccine. 2004;22:3427–34.
    1. CDC. Interim within-season estimate of the effectiveness of trivalent inactivated influenza vaccine–Marshfield, Wisconsin, 2007–08 influenza season. MMWR Morb Mortal Wkly Rep. 2008;57:393–8.
    1. Breathnach CC, Clark HJ, Clark RC, Olsen CW, Townsend HG, Lunn DP. Immunization with recombinant modified vaccinia Ankara (rMVA) constructs encoding the HA or NP gene protects ponies from equine influenza virus challenge. Vaccine. 2005;24:1180–90.
    1. Donnelly JJ, Friedman A, Martinez D, et al. Preclinical efficacy of a prototype DNA vaccine: enhanced protection against antigenic drift in influenza virus. Nat Med. 1995;1:583–7.
    1. Epstein SL, Tumpey TM, Misplon JA, et al. DNA vaccine expressing conserved influenza virus proteins protective against H5N1 challenge infection in mice. Emerg Infect Dis. 2002;8:796–801.
    1. Laddy DJ, Yan J, Khan AS, et al. Electroporation of synthetic DNA antigens offers protection in nonhuman primates challenged with highly pathogenic avian influenza virus. J Virol. 2009;83:4624–30.

Source: PubMed

3
Tilaa