Induction of Cross-Reactive Hemagglutination Inhibiting Antibody and Polyfunctional CD4+ T-Cell Responses by a Recombinant Matrix-M-Adjuvanted Hemagglutinin Nanoparticle Influenza Vaccine

Vivek Shinde, Rongman Cai, Joyce Plested, Iksung Cho, Jamie Fiske, Xuan Pham, Mingzhu Zhu, Shane Cloney-Clark, Nan Wang, Haixia Zhou, Bin Zhou, Nita Patel, Michael J Massare, Amy Fix, Michelle Spindler, David Nigel Thomas, Gale Smith, Louis Fries, Gregory M Glenn, Vivek Shinde, Rongman Cai, Joyce Plested, Iksung Cho, Jamie Fiske, Xuan Pham, Mingzhu Zhu, Shane Cloney-Clark, Nan Wang, Haixia Zhou, Bin Zhou, Nita Patel, Michael J Massare, Amy Fix, Michelle Spindler, David Nigel Thomas, Gale Smith, Louis Fries, Gregory M Glenn

Abstract

Background: Recurrent reports of suboptimal influenza vaccine effectiveness have renewed calls to develop improved, broadly cross-protective influenza vaccines. Here, we evaluated the safety and immunogenicity of a novel, saponin (Matrix-M)-adjuvanted, recombinant hemagglutinin (HA) quadrivalent nanoparticle influenza vaccine (qNIV).

Methods: We conducted a randomized, observer-blind, comparator-controlled (trivalent high-dose inactivated influenza vaccine [IIV3-HD] or quadrivalent recombinant influenza vaccine [RIV4]), safety and immunogenicity trial of qNIV (5 doses/formulations) in healthy adults ≥65 years. Vaccine immunogenicity was measured by hemagglutination-inhibition assays using reagents that express wild-type hemagglutination inhibition (wt-HAI) sequences and cell-mediated immune responses.

Results: A total of 1375 participants were randomized, immunized, and followed for safety and immunogenicity. Matrix-M-adjuvanted qNIV induced superior wt-HAI antibody responses against 5 of 6 homologous or drifted strains compared with unadjuvanted qNIV. Adjuvanted qNIV induced post-vaccination wt-HAI antibody responses at day 28 that were statistically higher than IIV3-HD against a panel of homologous or drifted A/H3N2 strains, similar to IIV3-HD against homologous A/H1N1 and B (Victoria) strains and similar to RIV4 against all homologous and drifted strains evaluated. The qNIV formulation with 75 µg Matrix-M adjuvant induced substantially higher post-vaccination geometric mean fold increases of influenza HA-specific polyfunctional CD4+ T cells compared with IIV3-HD or RIV4. Overall, similar frequencies of solicited and unsolicited adverse events were reported in all treatment groups.

Conclusions: qNIV with 75 µg Matrix-M adjuvant was well tolerated and induced robust antibody and cellular responses, notably against both homologous and drifted A/H3N2 viruses. Further investigation in a pivotal phase 3 trial is underway.

Clinical trials registration: NCT03658629.

Keywords: cell-mediated immunity; hemagglutination inhibition; influenza; vaccination.

© The Author(s) 2020. Published by Oxford University Press for the Infectious Diseases Society of America.

Figures

Figure 1.
Figure 1.
Flow diagram on screening, enrollment, and disposition of participants through the study. Safety population is defined as all participants who provided consent, were randomized, and received any investigational treatment; used for all descriptive safety analyses. Immunogenicity per protocol population is defined as all participants in the safety population who received the assigned investigational treatment according to the protocol, had wild-type hemagglutination inhibition (HAI) serology results for day 0 and day 28, and had no major protocol deviations that affected the primary immunogenicity outcomes as determined by the sponsor prior to database lock and unblinding; used for all immunogenicity analyses. The ITT population is defined as all participants in the safety population who provided any HAI serology data. Abbreviations: A, influenza A strain hemagglutinin (HA) antigen content in micrograms for each of A/H1N1 and A/H3N2 strains; AE, adverse event; B, influenza B strain HA antigen content in micrograms for each of B/Victoria and B/Yamagata lineage strains; f/u, follow-up; IIV3-HD, trivalent high-dose inactivated influenza vaccine (Fluzone High-Dose); ITT, intent-to-treat population; M, Matrix-M adjuvant content in micrograms; qNIV, quadrivalent recombinant nanoparticle influenza vaccine; RIV4, quadrivalent recombinant influenza vaccine (Flublok Quadrivalent); voluntary*, voluntary withdrawal unrelated to an adverse event.
Figure 2.
Figure 2.
Demonstration of adjuvant effect–baseline adjusted ratio of day 28 wt-HAI geometric mean titers (GMTs; GMTR) (Matrix-M–adjuvanted qNIV [group B]/unadjuvanted qNIV [group E]). Full strain names: A/Singapore/INFIMH-16–0019/2016 (H3N2); A/Switzerland/9715293/2013 (H3N2); A/Wisconsin/19/2017 (H3N2); A/Michigan/45/2015 (H1N1); B/Colorado/06/2017 (Victoria lineage); B/Phuket/3073/2013 (Yamagata lineage). The primary immunogenicity objective of demonstrating an adjuvant effect required establishing immunogenic superiority of group B (qNIV 60 µg hemagglutinin [HA] × 4 strains with 50 µg Matrix-M1 adjuvant) relative to group E (qNIV 60 µg HA × 4 strains without adjuvant) by excluding values ≤1.0 at the lower 95% confidence bound for the baseline-adjusted ratio of day 28 post-vaccination wt-HAI GMTs (ie, GMT of group B [adjuvant]/GMT of group E [no adjuvant] at day 28) for not less than 2 of 6 influenza strains (ie, any 2 of 4 vaccine-homologous strains and/or 2 antigenically drifted influenza strains), while no other strain(s) demonstrated GMTRs that were significantly

Figure 3.

A, qNIV (group B or…

Figure 3.

A, qNIV (group B or C) compared with IIV3-HD–baseline adjusted ratio of day…
Figure 3.
A, qNIV (group B or C) compared with IIV3-HD–baseline adjusted ratio of day 28 wt-HAI geometric mean titers (GMTs; GMTR) (qNIV [group B or C]/IIV3-HD [group F]). Full strain names: A/Singapore/INFIMH-16–0019/2016 (H3N2); A/Switzerland/9715293/2013 (H3N2); A/Wisconsin/19/2017 (H3N2); A/Michigan/45/2015 (H1N1); B/Colorado/06/2017 (Victoria lineage); B/Phuket/3073/2013 (Yamagata lineage). B, qNIV (group B or C) compared with RIV4–baseline adjusted ratio of day 28 wt-HAI GMTs (GMTR) (qNIV [group B or C]/RIV4 [group G]). Since day 56 samples were tested separately from day 0 and day 28 samples, day 56 titers were adjusted for the long-term assay variability. The adjustment was based on retesting of a randomly selected subset, 50 participants, of day 0 samples concurrently with day 56 samples. Abbreviations: B, group B; C, group C; F, group F; IIV3-HD, trivalent high-dose inactivated influenza vaccine; qNIV, quadrivalent recombinant nanoparticle influenza vaccine; wt-HAI, wild-type sequenced hemagglutinin inhibition antibody.

Figure 4.

Log 10 scale counts of…

Figure 4.

Log 10 scale counts of double- or triple-cytokine producing strain-specific CD4+ T cells…

Figure 4.
Log10 scale counts of double- or triple-cytokine producing strain-specific CD4+ T cells by treatment group, time point, and strain. Cell-mediated immune (CMI) responses were measured by intracellular cytokine staining. Counts of peripheral blood CD4+ T cells producing interleukin-2 (IL-2), interferon gamma (IFN-γ), and/or tumor necrosis factor alpha (TNF-α) cytokines were measured following in vitro restimulation with vaccine-homologous (A/Singapore/FIMH-16–0019/2016 [H3N2]; A/Michigan/45/2015 [H1N1]; /Colorado/06/2017 [Victoria]), or drifted (A/Wisconsin/19/2017 [H3N2]) strain-specific recombinant wild-type sequence hemagglutinins (HAs). A, CMI responses against A/Singapore and A/Wisconsin. B, CMI responses against B/Colorado and A/Michigan. Box plots are shown for counts of double-cytokine producing (any 2 of IFN-γ, TNF-α, or IL-2) or triple-cytokine producing (all 3 of IFN-γ, TNF-α, and IL-2) strain-specific CD4+ T-cell responses across the 4 strains evaluated using peripheral blood mononuclear cells obtained from a subgroup of participants on day 0 (pre-vaccination) and day 7 (post-vaccination). The box plots represent the interquartile range (±3 standard deviations), the solid horizontal black line represents the median, the number in red indicates the median count of double- or triple-cytokine producing CD4+ T cells, respectively, and the open diamond represents the mean. Group B is qNIV 60 µg HA × 4 strains with 50 µg Matrix-M1 adjuvant; group C is qNIV 60 µg HA × 4 strains with 75 µg Matrix-M1 adjuvant; group F is IIV3-HD; and group G is RIV4. Note that the number of strains tested for a given participant’s sample was dependent on the number of cells available; thus, not all samples could be tested across all 4 strains. Abbreviations: IIV3-HD, trivalent high-dose inactivated influenza vaccine; qNIV, quadrivalent recombinant nanoparticle influenza vaccine; RIV4, quadrivalent recombinant influenza vaccine.
Figure 3.
Figure 3.
A, qNIV (group B or C) compared with IIV3-HD–baseline adjusted ratio of day 28 wt-HAI geometric mean titers (GMTs; GMTR) (qNIV [group B or C]/IIV3-HD [group F]). Full strain names: A/Singapore/INFIMH-16–0019/2016 (H3N2); A/Switzerland/9715293/2013 (H3N2); A/Wisconsin/19/2017 (H3N2); A/Michigan/45/2015 (H1N1); B/Colorado/06/2017 (Victoria lineage); B/Phuket/3073/2013 (Yamagata lineage). B, qNIV (group B or C) compared with RIV4–baseline adjusted ratio of day 28 wt-HAI GMTs (GMTR) (qNIV [group B or C]/RIV4 [group G]). Since day 56 samples were tested separately from day 0 and day 28 samples, day 56 titers were adjusted for the long-term assay variability. The adjustment was based on retesting of a randomly selected subset, 50 participants, of day 0 samples concurrently with day 56 samples. Abbreviations: B, group B; C, group C; F, group F; IIV3-HD, trivalent high-dose inactivated influenza vaccine; qNIV, quadrivalent recombinant nanoparticle influenza vaccine; wt-HAI, wild-type sequenced hemagglutinin inhibition antibody.
Figure 4.
Figure 4.
Log10 scale counts of double- or triple-cytokine producing strain-specific CD4+ T cells by treatment group, time point, and strain. Cell-mediated immune (CMI) responses were measured by intracellular cytokine staining. Counts of peripheral blood CD4+ T cells producing interleukin-2 (IL-2), interferon gamma (IFN-γ), and/or tumor necrosis factor alpha (TNF-α) cytokines were measured following in vitro restimulation with vaccine-homologous (A/Singapore/FIMH-16–0019/2016 [H3N2]; A/Michigan/45/2015 [H1N1]; /Colorado/06/2017 [Victoria]), or drifted (A/Wisconsin/19/2017 [H3N2]) strain-specific recombinant wild-type sequence hemagglutinins (HAs). A, CMI responses against A/Singapore and A/Wisconsin. B, CMI responses against B/Colorado and A/Michigan. Box plots are shown for counts of double-cytokine producing (any 2 of IFN-γ, TNF-α, or IL-2) or triple-cytokine producing (all 3 of IFN-γ, TNF-α, and IL-2) strain-specific CD4+ T-cell responses across the 4 strains evaluated using peripheral blood mononuclear cells obtained from a subgroup of participants on day 0 (pre-vaccination) and day 7 (post-vaccination). The box plots represent the interquartile range (±3 standard deviations), the solid horizontal black line represents the median, the number in red indicates the median count of double- or triple-cytokine producing CD4+ T cells, respectively, and the open diamond represents the mean. Group B is qNIV 60 µg HA × 4 strains with 50 µg Matrix-M1 adjuvant; group C is qNIV 60 µg HA × 4 strains with 75 µg Matrix-M1 adjuvant; group F is IIV3-HD; and group G is RIV4. Note that the number of strains tested for a given participant’s sample was dependent on the number of cells available; thus, not all samples could be tested across all 4 strains. Abbreviations: IIV3-HD, trivalent high-dose inactivated influenza vaccine; qNIV, quadrivalent recombinant nanoparticle influenza vaccine; RIV4, quadrivalent recombinant influenza vaccine.

References

    1. Grohskopf LA, Alyanak E, Broder KR, Walter EB, Fry AM, Jernigan DB. Prevention and control of seasonal influenza with vaccines: recommendations of the Advisory Committee on Immunization Practices—United States, 2019–20 influenza season. MMWR Recomm Rep 2019; 68:1–20.
    1. Paget J, Spreeuwenberg P, Charu V, et al. ; Global Seasonal Influenza-associated Mortality Collaborator Network and GLaMOR Collaborating Teams . Global mortality associated with seasonal influenza epidemics: new burden estimates and predictors from the GLaMOR Project. J Glob Health 2019; 9:020421.
    1. Centers for Disease Control and Prevention. Past weekly surveillance reports. Available at: . Accessed 7 February 2020.
    1. Kissling E, Pozo F, Buda S, et al. . Low 2018/19 vaccine effectiveness against influenza A(H3N2) among 15–64-year-olds in Europe: exploration by birth cohort. Euro Surveill 2019; 24:1900604.
    1. Kissling E, Pozo F, Buda S, et al. ; I-MOVE/I-MOVE+ Study Team . Effectiveness of influenza vaccine against influenza A in Europe in seasons of different A(H1N1)pdm09 and the same A(H3N2) vaccine components (2016–17 and 2017–18). Vaccine X 2019; 3:100042.
    1. Skowronski DM, Chambers C, De Serres G, et al. . Early season co-circulation of influenza A(H3N2) and B(Yamagata): interim estimates of 2017/18 vaccine effectiveness, Canada, January 2018. Euro Surveill 2018; 23:18-00035.
    1. Skowronski DM, Sabaiduc S, Leir S, et al. . Paradoxical clade- and age-specific vaccine effectiveness during the 2018/19 influenza A(H3N2) epidemic in Canada: potential imprint-regulated effect of vaccine (I-REV). Euro Surveill 2019; 24:1900585.
    1. Flannery B, Kondor RJG, Chung JR, et al. . Spread of antigenically drifted influenza A(H3N2) viruses and vaccine effectiveness in the United States during the 2018–2019 season. J Infect Dis 2020; 221:8–15.
    1. Rolfes MA, Flannery B, Chung JR, et al. . Effects of influenza vaccination in the United States during the 2017–2018 influenza season. Clin Infect Dis 2019; 69:1845–53.
    1. Flannery B, Chung JR, Monto AS, et al. . Influenza vaccine effectiveness in the United States during the 2016–2017 season. Clin Infect Dis 2019; 68:1798–806.
    1. Russell K, Chung JR, Monto AS, et al. . Influenza vaccine effectiveness in older adults compared with younger adults over five seasons. Vaccine 2018; 36:1272–8.
    1. Zost SJ, Parkhouse K, Gumina ME, et al. . Contemporary H3N2 influenza viruses have a glycosylation site that alters binding of antibodies elicited by egg-adapted vaccine strains. Proc Natl Acad Sci U S A 2017; 114:12578–83.
    1. Wu NC, Zost SJ, Thompson AJ, et al. . A structural explanation for the low effectiveness of the seasonal influenza H3N2 vaccine. PLoS Pathog 2017; 13:e1006682.
    1. Skowronski DM, Janjua NZ, De Serres G, et al. . Low 2012-13 influenza vaccine effectiveness associated with mutation in the egg-adapted H3N2 vaccine strain not antigenic drift in circulating viruses. PLoS One 2014; 9:e92153.
    1. Levine MZ, Martin ET, Petrie JG, et al. . Antibodies against egg- and cell-grown influenza A(H3N2) viruses in adults hospitalized during the 2017–2018 influenza season. J Infect Dis 2019; 219:1904–12.
    1. Subbarao K, Barr I. A tale of two mutations: beginning to understand the problems with egg-based influenza vaccines? Cell Host Microbe 2019; 25:773–5.
    1. Allen JD, Ross TM. H3N2 influenza viruses in humans: viral mechanisms, evolution, and evaluation. Hum Vaccin Immunother 2018; 14:1840–7.
    1. Paules CI, Sullivan SG, Subbarao K, Fauci AS. Chasing seasonal influenza—the need for a universal influenza vaccine. N Engl J Med 2018; 378:7–9.
    1. Erbelding EJ, Post DJ, Stemmy EJ, et al. . A universal influenza vaccine: the strategic plan for the National Institute of Allergy and Infectious Diseases. J Infect Dis 2018; 218:347–54.
    1. Belongia EA, McLean HQ. Influenza vaccine effectiveness: defining the H3N2 problem. Clin Infect Dis 2019; 69:1817–23.
    1. Matias G, Taylor R, Haguinet F, Schuck-Paim C, Lustig R, Shinde V. Estimates of mortality attributable to influenza and RSV in the United States during 1997–2009 by influenza type or subtype, age, cause of death, and risk status. Influenza Other Respir Viruses 2014; 8:507–15.
    1. Matias G, Taylor R, Haguinet F, Schuck-Paim C, Lustig R, Shinde V. Estimates of hospitalization attributable to influenza and RSV in the US during 1997–2009, by age and risk status. BMC Public Health 2017; 17:271.
    1. Ferdinands JM, Fry AM, Reynolds S, et al. . Intraseason waning of influenza vaccine protection: evidence from the US Influenza Vaccine Effectiveness Network, 2011-12 through 2014-15. Clin Infect Dis 2017; 64:544–50.
    1. Kumar A, McElhaney JE, Walrond L, et al. . Cellular immune responses of older adults to four influenza vaccines: results of a randomized, controlled comparison. Hum Vaccin Immunother 2017; 13:2048–57.
    1. Cowling BJ, Perera RAPM, Valkenburg SA, et al. . Comparative immunogenicity of several enhanced influenza vaccine options for older adults: a randomized, controlled trial. Clin Infect Dis 2020; 71:1704–14.
    1. Smith G, Liu Y, Flyer D, et al. . Novel hemagglutinin nanoparticle influenza vaccine with Matrix-M™ adjuvant induces hemagglutination inhibition, neutralizing, and protective responses in ferrets against homologous and drifted A(H3N2) subtypes. Vaccine 2017; 35:5366–72.
    1. Portnoff AD, Patel N, Massare MJ, et al. . Influenza hemagglutinin nanoparticle vaccine elicits broadly neutralizing antibodies against structurally distinct domains of H3N2 HA. Vaccines (Basel) 2020; 8:99.
    1. Shinde V, Fries L, Wu Y, et al. . Improved titers against influenza drift variants with a nanoparticle vaccine. N Engl J Med 2018; 378:2346–8.
    1. Couch RB, Bayas JM, Caso C, et al. . Superior antigen-specific CD4+ T-cell response with AS03-adjuvantation of a trivalent influenza vaccine in a randomised trial of adults aged 65 and older. BMC Infect Dis 2014; 14:425.
    1. Belongia EA, Simpson MD, King JP, et al. . Variable influenza vaccine effectiveness by subtype: a systematic review and meta-analysis of test-negative design studies. Lancet Infect Dis 2016; 16:942–51.
    1. Centers for Disease Control and Prevention. 2010–11 through 2018–19 influenza seasons vaccination coverage trend report. Available at: . Accessed 7 February 2020.
    1. Belongia EA, Kieke BA, Donahue JG, et al. . Effectiveness of inactivated influenza vaccines varied substantially with antigenic match from the 2004–2005 season to the 2006–2007 season. J Infect Dis 2009; 199:159–67.
    1. Coughlan L, Palese P. Overcoming barriers in the path to a universal influenza virus vaccine. Cell Host Microbe 2018; 24:18–24.
    1. Ward BJ, Pillet S, Charland N, Trepanier S, Couillard J, Landry N. The establishment of surrogates and correlates of protection: useful tools for the licensure of effective influenza vaccines? Hum Vaccin Immunother 2018; 14:647–56.
    1. Wilkinson TM, Li CKF, Chui CSC, et al. . Preexisting influenza-specific CD4+ T cells correlate with disease protection against influenza challenge in humans. Nat Med 2012; 18:274–80.
    1. Sridhar S, Begom S, Bermingham A, et al. . Cellular immune correlates of protection against symptomatic pandemic influenza. Nat Med 2013; 19:1305–12.
    1. Clemens EB, Van De Sandt C, Wong SS, Wakim LM, Valkenburg SA. Harnessing the power of T cells: the promising hope for a universal influenza vaccine. Vaccines (Basel) 2018; 6:18.
    1. Effros RB. Role of T lymphocyte replicative senescence in vaccine efficacy. Vaccine 2007; 25:599–604.
    1. Andrew MK, Shinde V, Ye L, et al. ; Serious Outcomes Surveillance Network of the Public Health Agency of Canada/Canadian Institutes of Health Research Influenza Research Network and the Toronto Invasive Bacterial Diseases Network . The importance of frailty in the assessment of influenza vaccine effectiveness against influenza-related hospitalization in elderly people. J Infect Dis 2017; 216:405–14.
    1. Shahid Z, Kleppinger A, Gentleman B, Falsey AR, McElhaney JE. Clinical and immunologic predictors of influenza illness among vaccinated older adults. Vaccine 2010; 28:6145–51.

Source: PubMed

3
Abonnere