AS03-adjuvanted H5N1 vaccine promotes antibody diversity and affinity maturation, NAI titers, cross-clade H5N1 neutralization, but not H1N1 cross-subtype neutralization

Surender Khurana, Elizabeth M Coyle, Jody Manischewitz, Lisa R King, Jin Gao, Ronald N Germain, Pamela L Schwartzberg, John S Tsang, Hana Golding, and the CHI Consortium, Angelique Biancotto, Julián Candia, Jinguo Chen, Foo Cheung, Howard Dickler, Yuri Kotliarov, Shira Perl, Rongye Shi, Katherine E R Stagliano, Neal S Young, Huizhi Zhou, Surender Khurana, Elizabeth M Coyle, Jody Manischewitz, Lisa R King, Jin Gao, Ronald N Germain, Pamela L Schwartzberg, John S Tsang, Hana Golding, and the CHI Consortium, Angelique Biancotto, Julián Candia, Jinguo Chen, Foo Cheung, Howard Dickler, Yuri Kotliarov, Shira Perl, Rongye Shi, Katherine E R Stagliano, Neal S Young, Huizhi Zhou

Abstract

Immune responses to inactivated vaccines against avian influenza are poor due in part to lack of immune memory. Adjuvants significantly increased virus neutralizing titers. We performed comprehensive analyses of polyclonal antibody responses following FDA-approved adjuvanted H5N1-A/Indonesia vaccine, administered in presence or absence of AS03. Using Whole Genome Fragment Phage Display Libraries, we observed that AS03 induced antibody epitope diversity to viral hemagglutinin (HA) and neuraminidase compared with unadjuvanted vaccine. Furthermore, AS03 promoted significant antibody affinity maturation to properly folded H5-HA1 (but not to HA2) domain, which correlated with neutralization titers against both vaccine and heterologous H5N1 strains. However, no increase in heterosubtypic cross-neutralization of Group1-H1N1 seasonal strains was observed. AS03-H5N1 vaccine also induced higher neuraminidase inhibition antibody titers. This study provides insight into the differential impacts of AS03 adjuvant on H5N1 vaccine-induced antibody responses that may help optimize vaccine platforms for future vaccines with improved protection against seasonal and pandemic influenza strains.

Conflict of interest statement

The authors declare no competing interests.

Figures

Fig. 1
Fig. 1
Endpoint microneutralization titers following vaccination of adults with H5N1 with and without AS03 adjuvant against diverse H5N1 strains. a Schematic design of the heterologous H5N1 prime-boost vaccine immunization is shown. The number of individuals in each group: Unadjuvanted = 20; AS03-adjuvanted = 22. b−f End-point microneutralization titers of the individuals against various H5N1 virus strains at day 0 (prevaccination), day 21 (post first vaccination), day 42 (post second vaccination) and day 100 postvaccination are shown for AS03-adjuvanted group (in red) and unadjuvanted group (in blue) against b homologous H5N1 A/Indonesia/5/2005 (clade 2.1) vaccine strain, c A/Anhui/1/2005 (clade 2.3.4). d A/Turkey/15/2006 (clade 2.2), e A/Egypt/3072/2010 (clade 2.2), and f A/Vietnam/1194/2004 (clade 1). The pairwise comparison of endpoint neutralization titers that were statistically significant with p values of <0.05 (*), or <0.005 (**), or <0.0005 (***), or <0.0001 (****) are shown
Fig. 2
Fig. 2
Elucidation of antibody repertoires elicited in humans following vaccination with unadjuvanted and adjuvanted subunit H5N1-A/Indonesia vaccine. a Distribution of phage clones after affinity selection on post-H5N1 vaccination sera. b Schematic alignment of the peptides recognized by post-second vaccination sera in the NIAID sponsored trial, identified by panning with H5N1-GFPDL A/Indonesia/5/2005. Amino acid designation is based on the HA + NA protein sequence (Fig. S1). Bars with arrows indicate identified inserts in the 5′-3′ orientation in HA1 (red bars), HA2 (blue bars), and NA (black bars). Only clones with frequency of ≥2 are shown. Yellow bars indicate inserts spanning the receptor binding domain (RBD) in HA1. The pie charts represent the ratio of frequency of clones selected in HA1 (red) vs. HA2 (blue) domain by various vaccine groups are depicted. The thickness of bars represents the frequency of phages containing each insert. All bars shown represent phages that were affinity selected at least twice
Fig. 3
Fig. 3
Binding of post-H5N1 vaccination human sera to properly folded HA1 and HA2 domains. ac Steady-state equilibrium analysis of human vaccine sera at day 0 (prevaccination), day 21 (post first vaccination), day 42 (post second vaccination) and day 100 postvaccination are shown for AS03-adjuvanted group and unadjuvanted group against properly folded homologous H5-A/Indonesia/5/05 (Clade 2.1) HA1 (a) and HA2 (b) domains and heterologous H5N1-A/Vietnam/1203/2004 (Clade 1) HA1 domain (c) were measured using SPR. Recombinant HA1 and HA2 domains were immobilized on a sensor chip through the free amine group. Binding of the antibodies to the immobilized protein is shown as resonance unit (RU) values. Mean with standard deviation (SD) is also shown. The pairwise comparison of serum polyclonal antibody binding titers that were statistically significant with p values of <0.05 (*), or <0.005 (**), or <0.0005 (***), or <0.0001 (****) are shown
Fig. 4
Fig. 4
Kinetics of antibody affinity maturation following vaccination with adjuvanted and unadjuvanted H5N1 subunit vaccine. Sequential SPR analysis of vaccine sera (pre-, post first and post second vaccination with unadjuvanted or AS03-adjuvanted H5N1 vaccine) with properly folded homologous H5-A/Indonesia/5/05 HA1 (a) and HA2 (b) domains. Ten-fold and 100-fold diluted individual serum from each participant in vaccine trial at prevaccination (day 0) and at 21 days after each immunization, as well as at day 100 were evaluated. Serum antibody off-rate constants were determined as described in the Material and Methods. Mean with standard deviation (SD) is also shown. The pairwise comparison of polyclonal antibody off-rates that were statistically significant with p values of <0.05 (*), or <0.005 (**), or <0.0005 (***), or <0.0001 (****) are shown. c, d Correlation between in vitro neutralizing titers against homologous H5N1-A/Indonesia/5/05 and anti-HA1 antibody affinity (c) and anti-HA2 binding affinity (d) in human sera before and after H5N1 vaccination is shown. eh Correlation statistics of the off-rate constants and peak MN titers on day 42 (post second vaccine) for heterologous H5N1 strains are shown and found to be highly significant for e A/Anhui/1/2005 (clade 2.3.4), f A/Turkey/15/2006 (clade 2.2), g A/Egypt/3072/2010 (clade 2.2), with p < 0.05 but not for highly divergent h A/Vietnam/1194/2004 (clade 1) virus
Fig. 5
Fig. 5
No increase in neutralization titers against Group 1 seasonal H1N1 strains following vaccination with AS03-adjuvanted H5N1 vaccine in adults. To ascertain the generation of stem-specific neutralizing antibodies that will cross neutralize Groups 1 H1N1 strains following vaccination with AS03-adjuvanted H5N1 vaccine, a modified microneutralization assay was performed (as described in the Materials and Methods) with serum samples from day 0 (prevaccination) and day 7 post first vaccination (demonstrated to have peak HA2 stem cross-reactive titers following heterologous H5N1 vaccination). No significantly higher fold change (day 7/day 0) in neutralization titers were observed for any of the Gp 1 H1N1 (A/New Caledonia/22/99, A/Solomon Islands/3/06, A/Brisbane/59/07, A/California/07/2009), or Gp 2 H3N2 (A/Switzerland/9715293/2013) viruses following vaccination AS03-adjuvanted H5N1 (a) or unadjuvanted H5N1 vaccine (b). Mean with standard deviation (SD) is also shown
Fig. 6
Fig. 6
AS03 adjuvant promotes H5N1 neuraminidase-inhibiting antibodies. Neuraminidase enzymatic activity inhibition (NAI) titers for prevaccination (day 0) and post second vaccination (day 42) human sera were measured as described in the Material and Methods. The pairwise comparison of sera from AS03-adjuvanted vaccine recipients gave significantly higher fold change in NAI titers compared to unadjuvanted H5N1 vaccines with p values of <0.05, respectively. Mean with standard deviation (SD) is also shown

References

    1. Zeng H, et al. Highly pathogenic avian influenza H5N1 viruses elicit an attenuated type i interferon response in polarized human bronchial epithelial cells. J. Virol. 2007;81:12439–12449. doi: 10.1128/JVI.01134-07.
    1. Zitzow LA, et al. Pathogenesis of avian influenza A (H5N1) viruses in ferrets. J. Virol. 2002;76:4420–4429. doi: 10.1128/JVI.76.9.4420-4429.2002.
    1. Wang X, et al. Epidemiology of avian influenza A H7N9 virus in human beings across five epidemics in mainland China, 2013−17: an epidemiological study of laboratory-confirmed case series. Lancet Infect. Dis. 2017;17:822–832. doi: 10.1016/S1473-3099(17)30323-7.
    1. Zhang F, et al. Human infections with recently-emerging highly pathogenic H7N9 avian influenza virus in China. J. Infect. 2017;75:71–75. doi: 10.1016/j.jinf.2017.04.001.
    1. Ke C, et al. Human infection with highly pathogenic avian influenza A (H7N9) virus, China. Emerg. Infect. Dis. 2017;23:1332–1340. doi: 10.3201/eid2308.170600.
    1. Treanor JJ, et al. Safety and immunogenicity of a recombinant hemagglutinin vaccine for H5 influenza in humans. Vaccine. 2001;19:1732–1737. doi: 10.1016/S0264-410X(00)00395-9.
    1. Couch RB, Patel SM, Wade-Bowers CL, Nino D. A randomized clinical trial of an inactivated avian influenza A (H7N7) vaccine. PLoS ONE. 2012;7:e49704. doi: 10.1371/journal.pone.0049704.
    1. Garcon N, Vaughn DW, Didierlaurent AM. Development and evaluation of AS03, an adjuvant system containing alpha-tocopherol and squalene in an oil-in-water emulsion. Expert Rev. Vaccin. 2012;11:349–366. doi: 10.1586/erv.11.192.
    1. Morel S, et al. Adjuvant System AS03 containing alpha-tocopherol modulates innate immune response and leads to improved adaptive immunity. Vaccine. 2011;29:2461–2473. doi: 10.1016/j.vaccine.2011.01.011.
    1. Galli G, et al. Adjuvanted H5N1 vaccine induces early CD4+ T cell response that predicts long-term persistence of protective antibody levels. Proc. Natl. Acad. Sci. USA. 2009;106:3877–3882. doi: 10.1073/pnas.0813390106.
    1. Galli G, et al. Fast rise of broadly cross-reactive antibodies after boosting long-lived human memory B cells primed by an MF59 adjuvanted prepandemic vaccine. Proc. Natl. Acad. Sci. USA. 2009;106:7962–7967. doi: 10.1073/pnas.0903181106.
    1. Langley JM, et al. Safety and cross-reactive immunogenicity of candidate AS03-adjuvanted prepandemic H5N1 influenza vaccines: a randomized controlled phase 1/2 trial in adults. J. Infect. Dis. 2010;201:1644–1653. doi: 10.1086/652701.
    1. Langley JM, et al. Dose-sparing H5N1 A/Indonesia/05/2005 pre-pandemic influenza vaccine in adults and elderly adults: a phase III, placebo-controlled, randomized study. J. Infect. Dis. 2011;203:1729–1738. doi: 10.1093/infdis/jir172.
    1. Gillard P, et al. Long-term booster schedules with AS03A-adjuvanted heterologous H5N1 vaccines induces rapid and broad immune responses in Asian adults. Bmc Infect. Dis. 2014;14:142. doi: 10.1186/1471-2334-14-142.
    1. Chada KE, Forshee R, Golding H, Anderson S, Yang H. A systematic review and meta-analysis of cross-reactivity of antibodies induced by oil-in-water emulsion adjuvanted influenza H5N1 virus monovalent vaccines. Vaccine. 2017;35:3162–3170. doi: 10.1016/j.vaccine.2017.04.029.
    1. Hoelscher M, Gangappa S, Zhong W, Jayashankar L, Sambhara S. Vaccines against epidemic and pandemic influenza. Expert Opin. Drug Deliv. 2008;5:1139–1157. doi: 10.1517/17425247.5.10.1139.
    1. Jennings LC, Monto AS, Chan PK, Szucs TD, Nicholson KG. Stockpiling prepandemic influenza vaccines: a new cornerstone of pandemic preparedness plans. Lancet Infect. Dis. 2008;8:650–658. doi: 10.1016/S1473-3099(08)70232-9.
    1. Leroux-Roels I, et al. Priming with AS03 A-adjuvanted H5N1 influenza vaccine improves the kinetics, magnitude and durability of the immune response after a heterologous booster vaccination: an open non-randomised extension of a double-blind randomised primary study. Vaccine. 2010;28:849–857. doi: 10.1016/j.vaccine.2009.10.017.
    1. Leroux-Roels I, et al. Evaluation of the safety and immunogenicity of two antigen concentrations of the Mtb72F/AS02(A) candidate tuberculosis vaccine in purified protein derivative-negative adults. Clin. Vaccin. Immunol. 2010;17:1763–1771. doi: 10.1128/CVI.00133-10.
    1. Khurana S, et al. Antigenic fingerprinting of H5N1 avian influenza using convalescent sera and monoclonal antibodies reveals potential vaccine and diagnostic targets. PLoS Med. 2009;6:e1000049. doi: 10.1371/journal.pmed.1000049.
    1. Khurana S, et al. Vaccines with MF59 adjuvant expand the antibody repertoire to target protective sites of pandemic avian H5N1 influenza virus. Sci. Transl. Med. 2010;2:15ra15. doi: 10.1126/scitranslmed.3000624.
    1. Khurana S, et al. MF59 adjuvant enhances diversity and affinity of antibody-mediated immune response to pandemic influenza vaccines. Sci. Transl. Med. 2011;3:85ra48. doi: 10.1126/scitranslmed.3002336.
    1. Khurana S, et al. Bacterial HA1 vaccine against pandemic H5N1 influenza virus: evidence of oligomerization, hemagglutination, and cross-protective immunity in ferrets. J. Virol. 2010;85:1246–1256. doi: 10.1128/JVI.02107-10.
    1. Verma S, et al. Oligomeric recombinant H5 HA1 vaccine produced in bacteria protects ferrets from homologous and heterologous wild-type H5N1 influenza challenge and controls viral loads better than subunit H5N1 vaccine by eliciting high-affinity antibodies. J. Virol. 2012;86:12283–12293. doi: 10.1128/JVI.01596-12.
    1. Wan H, et al. Molecular basis for broad neuraminidase immunity: conserved epitopes in seasonal and pandemic H1N1 as well as H5N1 influenza viruses. J. Virol. 2013;87:9290–9300. doi: 10.1128/JVI.01203-13.
    1. Wiley DC, Wilson IA, Skehel JJ. Structural identification of the antibody-binding sites of Hong Kong influenza haemagglutinin and their involvement in antigenic variation. Nature. 1981;289:373–378. doi: 10.1038/289373a0.
    1. Bizebard T, et al. Structure of influenza virus haemagglutinin complexed with a neutralizing antibody. Nature. 1995;376:92–94. doi: 10.1038/376092a0.
    1. Corti D, et al. Heterosubtypic neutralizing antibodies are produced by individuals immunized with a seasonal influenza vaccine. J. Clin. Invest. 2010;120:1663–1673. doi: 10.1172/JCI41902.
    1. Dreyfus C, et al. Highly conserved protective epitopes on influenza B viruses. Science. 2012;337:1343–1348. doi: 10.1126/science.1222908.
    1. Throsby M, et al. Heterosubtypic neutralizing monoclonal antibodies cross-protective against H5N1 and H1N1 recovered from human IgM+ memory B cells. PLoS ONE. 2008;3:e3942. doi: 10.1371/journal.pone.0003942.
    1. Schmidt AG, et al. Preconfiguration of the antigen-binding site during affinity maturation of a broadly neutralizing influenza virus antibody. Proc. Natl. Acad. Sci. USA. 2013;110:264–269. doi: 10.1073/pnas.1218256109.
    1. Paules CI, Marston HD, Eisinger RW, Baltimore D, Fauci AS. The pathway to a universal influenza vaccine. Immunity. 2017;47:599–603. doi: 10.1016/j.immuni.2017.09.007.
    1. Nachbagauer R, Krammer F. Universal influenza virus vaccines and therapeutic antibodies. Clin. Microbiol. Infect. 2017;23:222–228. doi: 10.1016/j.cmi.2017.02.009.
    1. Jacobsen H, 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. doi: 10.1128/mBio.01463-17.
    1. Nakamura G, et al. An in vivo human-plasmablast enrichment technique allows rapid identification of therapeutic influenza A antibodies. Cell Host Microbe. 2013;14:93–103. doi: 10.1016/j.chom.2013.06.004.
    1. Andrews SF, et al. Immune history profoundly affects broadly protective B cell responses to influenza. Sci. Transl. Med. 2015;7:316ra192. doi: 10.1126/scitranslmed.aad0522.
    1. Koudstaal W, et al. Pre- and postexposure use of human monoclonal antibody against H5N1 and H1N1 influenza virus in mice: viable alternative to oseltamivir. J. Infect. Dis. 2009;200:1870–1873. doi: 10.1086/648378.
    1. Murphy BR, Kasel JA, Chanock RM. Association of serum anti-neuraminidase antibody with resistance to influenza in man. N. Engl. J. Med. 1972;286:1329–1332. doi: 10.1056/NEJM197206222862502.
    1. Kilbourne ED. Influenza pandemics of the 20th century. Emerg. Infect. Dis. 2006;12:9–14. doi: 10.3201/eid1201.051254.
    1. Monto AS, et al. Antibody to influenza virus neuraminidase: an independent correlate of protection. J. Infect. Dis. 2015;212:1191–1199. doi: 10.1093/infdis/jiv195.
    1. Memoli MJ, et al. Evaluation of antihemagglutinin and antineuraminidase antibodies as correlates of protection in an influenza A/H1N1 virus healthy human challenge model. mBio. 2016;7:e00417-00416. doi: 10.1128/mBio.00417-16.
    1. Gao J., Couzens L. & Eichelberger M. C. Measuring influenza neuraminidase inhibition antibody titers by enzyme-linked lectin assay. J. Vis. Exp. (2016), Sep 6;(115), 10.3791/54573.
    1. Treanor JJ, Campbell JD, Zangwill KM, Rowe T, Wolff M. Safety and immunogenicity of an inactivated subvirion influenza A (H5N1) vaccine. N. Engl. J. Med. 2006;354:1343–1351. doi: 10.1056/NEJMoa055778.
    1. Madan A, et al. Immunogenicity and safety of an AS03-adjuvanted H7N9 pandemic influenza vaccine in a randomized trial in healthy adults. J. Infect. Dis. 2016;214:1717–1727. doi: 10.1093/infdis/jiw414.
    1. Jackson LA, et al. Effect of varying doses of a monovalent H7N9 influenza vaccine with and without AS03 and MF59 adjuvants on immune response: a randomized clinical trial. JAMA. 2015;314:237–246. doi: 10.1001/jama.2015.7916.
    1. Tete SM, et al. Impact of pre-existing immunity on the induction of functional cross-reactive anti-hemagglutinin stalk antibodies following vaccination with an AS03 adjuvanted pandemic H1N1 vaccine. Vaccine. 2018;36:2213–2219. doi: 10.1016/j.vaccine.2018.02.022.
    1. Dilillo DJ, Tan GS, Palese P, Ravetch JV. Broadly neutralizing hemagglutinin stalk-specific antibodies require FcgammaR interactions for protection against influenza virus in vivo. Nat. Med. 2014;20:143–151. doi: 10.1038/nm.3443.
    1. Jegaskanda S, et al. Cross-reactive influenza-specific antibody-dependent cellular cytotoxicity antibodies in the absence of neutralizing antibodies. J. Immunol. 2013;190:1837–1848. doi: 10.4049/jimmunol.1201574.
    1. Madhavi V, et al. Antibody-dependent effector functions against HIV decline in subjects receiving antiretroviral therapy. J. Infect. Dis. 2015;211:529–538. doi: 10.1093/infdis/jiu486.
    1. Shen C, et al. A multimechanistic antibody targeting the receptor binding site potently cross-protects against influenza B viruses. Sci. Transl. Med. 2017;9:eaam5752. doi: 10.1126/scitranslmed.aam5752.
    1. Wong TM, et al. Computationally optimized broadly reactive hemagglutinin elicits hemagglutination inhibition antibodies against a panel of H3N2 influenza virus cocirculating variants. J. Virol. 2017;91:e01581-17. doi: 10.1128/JVI.01581-17.
    1. Carter DM, et al. Elicitation of protective antibodies against a broad panel of H1N1 viruses in ferrets preimmune to historical H1N1 influenza viruses. J. Virol. 2017;91:e01283. doi: 10.1128/JVI.01283-17.
    1. Carter DM, et al. Design and characterization of a computationally optimized broadly reactive hemagglutinin vaccine for H1N1 influenza viruses. J. Virol. 2016;90:4720–4734. doi: 10.1128/JVI.03152-15.
    1. Sutton, T. C. et al. In vitro neutralization is not predictive of prophylactic efficacy of broadly neutralizing monoclonal antibodies CR6261 and CR9114 against lethal H2 influenza virus challenge in mice. J. Virol.91 (2017), 2017 Nov 30;91(24), 10.1128/JVI.01603-17, Print 2017 Dec 15.
    1. Park JK, 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. doi: 10.1128/mBio.02284-17.
    1. Khurana S, et al. Vaccine-induced anti-HA2 antibodies promote virus fusion and enhance influenza virus respiratory disease. Sci. Transl. Med. 2013;5:200ra114. doi: 10.1126/scitranslmed.3006366.
    1. Co MD, et al. Relationship of preexisting influenza hemagglutination inhibition, complement-dependent lytic, and antibody-dependent cellular cytotoxicity antibodies to the development of clinical illness in a prospective study of A(H1N1)pdm09 Influenza in children. Viral Immunol. 2014;27:375–382. doi: 10.1089/vim.2014.0061.
    1. Crowe JE., Jr. Universal flu vaccines: primum non nocere. Sci. Transl. Med. 2013;5:200fs234. doi: 10.1126/scitranslmed.3007118.
    1. Khurana S. Development and regulation of novel influenza virus vaccines: a United States Young Scientist Perspective. Vaccin. (Basel) 2018;6:E24. doi: 10.3390/vaccines6020024.
    1. Galson JD, Truck J, Kelly DF, van der Most R. Investigating the effect of AS03 adjuvant on the plasma cell repertoire following pH1N1 influenza vaccination. Sci. Rep. 2016;6:37229. doi: 10.1038/srep37229.
    1. Sandbulte MR, et al. Discordant antigenic drift of neuraminidase and hemagglutinin in H1N1 and H3N2 influenza viruses. Proc. Natl. Acad. Sci. USA. 2011;108:20748–20753. doi: 10.1073/pnas.1113801108.
    1. Sandbulte MR, Gao J, Straight TM, Eichelberger MC. A miniaturized assay for influenza neuraminidase-inhibiting antibodies utilizing reverse genetics-derived antigens. Influenza Other Respir. Virus. 2009;3:233–240. doi: 10.1111/j.1750-2659.2009.00094.x.

Source: PubMed

3
Tilaa