Immunogenicity of chimeric haemagglutinin-based, universal influenza virus vaccine candidates: interim results of a randomised, placebo-controlled, phase 1 clinical trial

David I Bernstein, Jeffrey Guptill, Abdollah Naficy, Raffael Nachbagauer, Francesco Berlanda-Scorza, Jodi Feser, Patrick C Wilson, Alicia Solórzano, Marie Van der Wielen, Emmanuel B Walter, Randy A Albrecht, Kristen N Buschle, Yao-Qing Chen, Carine Claeys, Michelle Dickey, Haley L Dugan, Megan E Ermler, Debra Freeman, Min Gao, Christopher Gast, Jenna J Guthmiller, Rong Hai, Carole Henry, Linda Yu-Ling Lan, Monica McNeal, Anna-Karin E Palm, Dustin G Shaw, Christopher T Stamper, Weina Sun, Victoria Sutton, Micah E Tepora, Rahnuma Wahid, Heather Wenzel, Teddy John Wohlbold, Bruce L Innis, Adolfo García-Sastre, Peter Palese, Florian Krammer, David I Bernstein, Jeffrey Guptill, Abdollah Naficy, Raffael Nachbagauer, Francesco Berlanda-Scorza, Jodi Feser, Patrick C Wilson, Alicia Solórzano, Marie Van der Wielen, Emmanuel B Walter, Randy A Albrecht, Kristen N Buschle, Yao-Qing Chen, Carine Claeys, Michelle Dickey, Haley L Dugan, Megan E Ermler, Debra Freeman, Min Gao, Christopher Gast, Jenna J Guthmiller, Rong Hai, Carole Henry, Linda Yu-Ling Lan, Monica McNeal, Anna-Karin E Palm, Dustin G Shaw, Christopher T Stamper, Weina Sun, Victoria Sutton, Micah E Tepora, Rahnuma Wahid, Heather Wenzel, Teddy John Wohlbold, Bruce L Innis, Adolfo García-Sastre, Peter Palese, Florian Krammer

Abstract

Background: Influenza viruses cause substantial annual morbidity and mortality globally. Current vaccines protect against influenza only when well matched to the circulating strains. However, antigenic drift can cause considerable mismatches between vaccine and circulating strains, substantially reducing vaccine effectiveness. Moreover, current seasonal vaccines are ineffective against pandemic influenza, and production of a vaccine matched to a newly emerging virus strain takes months. Therefore, there is an unmet medical need for a broadly protective influenza virus vaccine. We aimed to test the ability of chimeric H1 haemagglutinin-based universal influenza virus vaccine candidates to induce broadly cross-reactive antibodies targeting the stalk domain of group 1 haemagglutinin-expressing influenza viruses.

Methods: We did a randomised, observer-blinded, phase 1 study in healthy adults in two centres in the USA. Participants were randomly assigned to one of three prime-boost, chimeric haemagglutinin-based vaccine regimens or one of two placebo groups. The vaccine regimens included a chimeric H8/1, intranasal, live-attenuated vaccine on day 1 followed by a non-adjuvanted, chimeric H5/1, intramuscular, inactivated vaccine on day 85; the same regimen but with the inactivated vaccine being adjuvanted with AS03; and an AS03-adjuvanted, chimeric H8/1, intramuscular, inactivated vaccine followed by an AS03-adjuvanted, chimeric H5/1, intramuscular, inactivated vaccine. In this planned interim analysis, the primary endpoints of reactogenicity and safety were assessed by blinded study group. We also assessed anti-H1 haemagglutinin stalk, anti-H2, anti-H9, and anti-H18 IgG antibody titres and plasmablast and memory B-cell responses in peripheral blood. This trial is registered with ClinicalTrials.gov, number NCT03300050.

Findings: Between Oct 10, 2017, and Nov 27, 2017, 65 participants were enrolled and randomly assigned. The adjuvanted inactivated vaccine, but not the live-attenuated vaccine, induced a substantial serum IgG antibody response after the prime immunisation, with a seven times increase in anti-H1 stalk antibody titres on day 29. After boost immunisation, all vaccine regimens induced detectable anti-H1 stalk antibody (2·2-5·6 times induction over baseline), cross-reactive serum IgG antibody, and peripheral blood plasmablast responses. An unsolicited adverse event was reported for 29 (48%) of 61 participants. Solicited local adverse events were reported in 12 (48%) of 25 participants following prime vaccination with intramuscular study product or placebo, in 12 (33%) of 36 after prime immunisation with intranasal study product or placebo, and in 18 (32%) of 56 following booster doses of study product or placebo. Solicited systemic adverse events were reported in 14 (56%) of 25 after prime immunisation with intramuscular study product or placebo, in 22 (61%) of 36 after immunisation with intranasal study product or placebo, and in 21 (38%) of 56 after booster doses of study product or placebo. Disaggregated safety data were not available at the time of this interim analysis.

Interpretation: The tested chimeric haemagglutinin-based, universal influenza virus vaccine regimens elicited cross-reactive serum IgG antibodies that targeted the conserved haemagglutinin stalk domain. This is the first proof-of-principle study to show that high anti-stalk titres can be induced by a rationally designed vaccine in humans and opens up avenues for further development of universal influenza virus vaccines. On the basis of the blinded study group, the vaccine regimens were tolerable and no safety concerns were observed.

Funding: Bill & Melinda Gates Foundation.

Copyright © 2020 The Author(s). Published by Elsevier Ltd. This is an Open Access article under the CC BY 4.0 license. Published by Elsevier Ltd.. All rights reserved.

Figures

Figure 1
Figure 1
Schematic of the chimeric HA vaccination regimens and trial design (A) Vaccination strategy. Adults have pre-existing antibodies targeting both the membrane-distal head domain (top) and the membrane-proximal stalk domain (bottom) of H1 HA (green) due to previous exposure to influenza viruses. Vaccination with a chimeric H8/1 construct is expected to elicit some antibodies against the head domain (yellow), to which humans are naive, while substantially boosting H1 stalk antibodies. An additional booster vaccination with chimeric H5/1 HA was expected to provide an additional increase in antibodies targeting the HA stalk domain. Structures were adapted from RCSB Protein Data Bank ID 1RU7 and visualised in Protein Workshop.12 (B) Vaccination and blood collection schedule. (C) A phylogenetic tree based on percentage amino acid difference was constructed to illustrate the evolutionary distance of the antigens used for the ELISA analysis. The H1 (blue) stalk domain was used in the vaccines. H2 is closely related to H1, whereas H9 and H18 (all highlighted in green) are distantly related HAs within influenza A group 1. HA subtypes that donated heads to the vaccine constructs (H5 and H8) are shown in purple. Group 1 HAs are shaded in purple and group 2 in orange. HA clades are indicated within the groups. The scale bar represents a 5% difference in amino acid sequence. IIV=inactivated influenza vaccine. LAIV=live-attenuated influenza vaccine. PBS=phosphate-buffered saline. HA=haemagglutinin.
Figure 2
Figure 2
Trial profile Randomisation into inpatient (LAIV8-IIV5/AS03, LAIV8-IIV5, and sterile saline placebo control) and outpatient (IIV8/AS03-IIV5/AS03 and phosphate buffered saline placebo control) groups is shown. *66 randomly assigned, with one ineligible participant included in error and excluded after randomisation.
Figure 3
Figure 3
Titres of antibodies targeting the H1 stalk domain and heterosubtypic group 1 haemagglutinins Geometric mean ELISA antibody titres (ELISA units per mL) are plotted on the y axis (log10) for the timepoints indicated on the x axis. Error bars show the upper and lower limits of the 95% CIs. Vaccination timepoints for the LAIV8-IIV5/AS03, LAIV8-IIV5, IIV8/AS03-IIV5/AS03, and PBS groups are indicated below the x-axis. Group sizes are 19 for the LAIV8-IIV5/AS03 group, 14 for the LAIV8-IIV5 group, 15 for the IIV8/AS03-IIV5/AS03 group, and ten for the PBS group. IIV=inactivated influenza vaccine. LAIV=live-attenuated influenza vaccine. PBS=phosphate-buffered saline.
Figure 4
Figure 4
Plasmablast and memory B-cell responses to the H1 stalk and wild-type H1 haemagglutinins The error bars indicate the upper and lower limits of the 95% CIs. The lower limit of detection was four spot forming units per 106 PBMCs. Plasmablasts were tested for H1 stalk-specific IgG (A), IgA (C), and Cal09 H1 IgA plus IgG (E) secretion on days 8 and 92 (7 days after vaccination). Memory B cells were tested for H1 stalk-specific IgG (B), IgA (D), and Cal09 H1 IgA plus IgG (F) secretion on days 1, 29, 85, and 113 (vaccination timepoints and 4-week post-vaccination timepoints). Group sizes are 19 for the LAIV8-IIV5/AS03 group, 14 for the LAIV8-IIV5 group, 15 for the IIV8/AS03-IIV5/AS03 group, and ten in the PBS group. IIV=inactivated influenza vaccine. LAIV=live-attenuated influenza vaccine. PBMC=peripheral blood mononuclear cells. PBS=phosphate-buffered saline.

References

    1. WHO Influenza (seasonal). Geneva: World Health Organization, 2018. (accessed Oct 2, 2019).
    1. Krammer F, Palese P. Advances in the development of influenza virus vaccines. Nat Rev Drug Discov 2015; 14: 167–82.
    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. Paules CI, Marston HD, Eisinger RW, Baltimore D, Fauci AS. The pathway to a universal influenza vaccine. Immunity 2017; 47: 599–603.
    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. Heaton NS, Sachs D, Chen CJ, Hai R, Palese P. Genome-wide mutagenesis of influenza virus reveals unique plasticity of the hemagglutinin and NS1 proteins. Proc Natl Acad Sci USA 2013; 110: 20248–53.
    1. Doud MB, Bloom JD. Accurate measurement of the effects of all amino-acid mutations on influenza hemagglutinin. Viruses 2016; 8: e155.
    1. Nachbagauer R, Choi A, Izikson R, Cox MM, Palese P, Krammer F. Age dependence and isotype specificity of influenza virus hemagglutinin stalk-reactive antibodies in humans. mBio 2016; 7: e01996-15.
    1. Sui J, Sheehan J, Hwang WC, et al. . Wide prevalence of heterosubtypic broadly neutralizing human anti-influenza a antibodies. Clin Infect Dis 2011; 52: 1003–09.
    1. Henry Dunand CJ, Leon PE, Kaur K, et al. . Preexisting human antibodies neutralize recently emerged H7N9 influenza strains. J Clin Invest 2015; 125: 1255–68.
    1. Wu NC, Wilson IA. Structural insights into the design of novel anti-influenza therapies. Nat Struct Mol Biol 2018; 25: 115–21.
    1. Gamblin S, Haire L, Russell R, et al. . The structure and receptor binding properties of the 1918 influenza hemagglutinin. Science 2004; 303: 1838–42.
    1. Hai R, Krammer F, Tan GS, et al. . Influenza viruses expressing chimeric hemagglutinins: globular head and stalk domains derived from different subtypes. J Virol 2012; 86: 5774–81.
    1. Chen CJ, Ermler ME, Tan GS, Krammer F, Palese P, Hai R. Influenza A viruses expressing intra- or intergroup chimeric hemagglutinins. J Virol 2016; 90: 3789–93.
    1. Ermler ME, Kirkpatrick E, Sun W, et al. . Chimeric hemagglutinin constructs induce broad protection against influenza B virus challenge in the mouse model. J Virol 2017; 91: e00286-17.
    1. Krammer F, Pica N, Hai R, Margine I, Palese P. Chimeric hemagglutinin influenza virus vaccine constructs elicit broadly protective stalk-specific antibodies. J Virol 2013; 87: 6542–50.
    1. Margine I, Krammer F, Hai R, et al. . Hemagglutinin stalk-based universal vaccine constructs protect against group 2 influenza A viruses. J Virol 2013; 87: 10435–46.
    1. Nachbagauer R, Liu W-C, Choi A, et al. . A universal influenza virus vaccine candidate confers protection against pandemic H1N1 infection in preclinical ferret studies. npj Vaccines 2017; 2: 26.
    1. Nachbagauer R, Kinzler D, Choi A, et al. . A chimeric haemagglutinin-based influenza split virion vaccine adjuvanted with AS03 induces protective stalk-reactive antibodies in mice. 2016; 1: 16015.
    1. Garçon N, Vaughn DW, Didierlaurent AM. Development and evaluation of AS03, an Adjuvant System containing α-tocopherol and squalene in an oil-in-water emulsion. Expert Rev Vaccines 2012; 11: 349–66.
    1. Talaat KR, Luke CJ, Khurana S, et al. . A live attenuated influenza A(H5N1) vaccine induces long-term immunity in the absence of a primary antibody response. J Infect Dis 2014; 209: 1860–09.
    1. Rudenko L, Naykhin A, Donina S, et al. . Assessment of immune responses to H5N1 inactivated influenza vaccine among individuals previously primed with H5N2 live attenuated influenza vaccine. Hum Vaccin Immunother 2015; 11: 2839–48.
    1. Jegaskanda S, Mason RD, Andrews SF, et al. . Intranasal live influenza vaccine priming elicits localized B cell responses in mediastinal lymph nodes. J Virol 2018; 92: e01970-17.
    1. Pitisuttithum P, Boonnak K, Chamnanchanunt S, et al. . Safety and immunogenicity of a live attenuated influenza H5 candidate vaccine strain A/17/turkey/Turkey/05/133 H5N2 and its priming effects for potential pre-pandemic use: a randomised, double-blind, placebo-controlled trial. Lancet Infect Dis 2017; 17: 833–42.
    1. Nachbagauer R, Krammer F, Albrecht RA. A live-attenuated prime, inactivated boost vaccination strategy with chimeric hemagglutinin-based universal influenza virus vaccines provides protection in ferrets: a confirmatory study. Vaccines (Basel) 2018; 6: e47.
    1. Isakova-Sivak I, Chen LM, Matsuoka Y, et al. . Genetic bases of the temperature-sensitive phenotype of a master donor virus used in live attenuated influenza vaccines: A/Leningrad/134/17/57 (H2N2). Virology 2011; 412: 297–305.
    1. Choi A, Bouzya B, Cortés Franco KD, et al. . Chimeric hemagglutinin-based influenza virus vaccines induce protective stalk-specific humoral immunity and cellular responses in mice. Immunohorizons 2019; 3: 133–48.
    1. Hong M, Lee PS, Hoffman RM, et al. . Antibody recognition of the pandemic H1N1 influenza virus hemagglutinin receptor binding site. J Virol 2013; 87: 12471–80.
    1. Nachbagauer R, Wohlbold TJ, Hirsh A, et al. . Induction of broadly reactive anti-hemagglutinin stalk antibodies by an H5N1 vaccine in humans. J Virol 2014; 88: 13260–68.
    1. Jacobsen H, Rajendran M, Choi A, et al. . Influenza virus hemagglutinin stalk-specific antibodies in human serum are a surrogate marker for in vivo protection in a serum transfer mouse challenge model. mBio 2017; 8: e01463-17.
    1. Ng S, Nachbagauer R, Balmaseda A, et al. . Novel correlates of protection against pandemic H1N1 influenza A virus infection. Nat Med 2019; 25: 962–67.
    1. Park JK, Han A, Czajkowski L, et al. . Evaluation of preexisting anti-hemagglutinin stalk antibody as a correlate of protection in a healthy volunteer challenge with influenza A/H1N1pdm virus. mBio 2018; 9: e02284-17.
    1. Couch RB, Atmar RL, Keitel WA, et al. . Randomized comparative study of the serum antihemagglutinin and antineuraminidase antibody responses to six licensed trivalent influenza vaccines. Vaccine 2012; 31: 190–95.
    1. Barría MI, Garrido JL, Stein C, et al. . Localized mucosal response to intranasal live attenuated influenza vaccine in adults. J Infect Dis 2013; 207: 115–24.
    1. Karron RA, Talaat K, Luke C, et al. . Evaluation of two live attenuated cold-adapted H5N1 influenza virus vaccines in healthy adults. Vaccine 2009; 27: 4953–60.
    1. Talaat KR, Karron RA, Luke CJ, et al. . An open label Phase I trial of a live attenuated H6N1 influenza virus vaccine in healthy adults. Vaccine 2011; 29: 3144–48.
    1. Talaat KR, Karron RA, Liang PH, et al. . An open-label phase I trial of a live attenuated H2N2 influenza virus vaccine in healthy adults. Influenza Other Respir Viruses 2013; 7: 66–73.
    1. Krammer F. The human antibody response to influenza A virus infection and vaccination. Nat Rev Immunol 2019; 19: 383–97.

Source: PubMed

3
Iratkozz fel