Glycan repositioning of influenza hemagglutinin stem facilitates the elicitation of protective cross-group antibody responses

Seyhan Boyoglu-Barnum, Geoffrey B Hutchinson, Jeffrey C Boyington, Syed M Moin, Rebecca A Gillespie, Yaroslav Tsybovsky, Tyler Stephens, John R Vaile, Julia Lederhofer, Kizzmekia S Corbett, Brian E Fisher, Hadi M Yassine, Sarah F Andrews, Michelle C Crank, Adrian B McDermott, John R Mascola, Barney S Graham, Masaru Kanekiyo, Seyhan Boyoglu-Barnum, Geoffrey B Hutchinson, Jeffrey C Boyington, Syed M Moin, Rebecca A Gillespie, Yaroslav Tsybovsky, Tyler Stephens, John R Vaile, Julia Lederhofer, Kizzmekia S Corbett, Brian E Fisher, Hadi M Yassine, Sarah F Andrews, Michelle C Crank, Adrian B McDermott, John R Mascola, Barney S Graham, Masaru Kanekiyo

Abstract

The conserved hemagglutinin (HA) stem has been a focus of universal influenza vaccine efforts. Influenza A group 1 HA stem-nanoparticles have been demonstrated to confer heterosubtypic protection in animals; however, the protection does not extend to group 2 viruses, due in part to differences in glycosylation between group 1 and 2 stems. Here, we show that introducing the group 2 glycan at Asn38HA1 to a group 1 stem-nanoparticle (gN38 variant) based on A/New Caledonia/20/99 (H1N1) broadens antibody responses to cross-react with group 2 HAs. Immunoglobulins elicited by the gN38 variant provide complete protection against group 2 H7N9 virus infection, while the variant loses protection against a group 1 H5N1 virus. The N38HA1 glycan thus is pivotal in directing antibody responses by controlling access to group-determining stem epitopes. Precise targeting of stem-directed antibody responses to the site of vulnerability by glycan repositioning may be a step towards achieving cross-group influenza protection.

Conflict of interest statement

J.C.B., H.M.Y., J.R.M., B.S.G. and M.K. are named inventors of a patent application on stabilized influenza HA stem filed by the National Institutes of Health. All other authors declare no competing interests.

Figures

Fig. 1. Design and characterization of group…
Fig. 1. Design and characterization of group 1 H1 stem nanoparticle variants.
a Comparison of N-linked glycosylation pattern of group 1 and group 2 HA stem. Surface renderings of HA trimer for a representative group 1 HA (A/Solomon Islands/06 (H1N1), PDB: 3SM5, left) and representative group 2 HA (A/Finland/486/04 (H3N2), PDB: 2YP5, right). One monomer is colored in blue for visibility. The terminal N-acetylglucosamine (GlcNAc) moiety for each glycan is modeled in green and residue positions are labeled according to the H3 numbering. Yellow areas designate the approximate location of the CR6261 epitope on each HA. b Amino acid sequence alignment of N-terminal portion of HA1 of H1 (A/New Caledonia/20/99), H3 (A/Finland/486/04) and designed variants gN38 and R38. Dots and dashes indicate residues identical to H1 and gaps, respectively. c Cryo-electron microscopy structures of H1 stem nanoparticle (H1ssF WT) and its variants H1ssF gN38 and H1ssF R38. Nanoparticles are depicted along the 2-fold symmetry axis. Side views of HA stem trimeric spikes are shown below each nanoparticle. The maps were low-pass filtered to a resolution of 7 Å for comparison. Scale bar indicates 5 nm. d Antigenicity of H1 stem nanoparticle variants. ELISA binding curves are shown for mAbs specific to group 1 stem (CR6261, 02-1H01 and C179), group 2 stem (CR8020), both group 1 and 2 stems (FI6v3, MEDI8852), or irrelevant RSV F protein (D25). Source data are provided as Source Data file.
Fig. 2. Immunogenicity of H1 stem nanoparticle…
Fig. 2. Immunogenicity of H1 stem nanoparticle variants in mice.
BALB/c mice (N = 10) were immunized with 2 µg of H1ssF WT, H1ssF gN38 or H1ssF R38 on weeks 0, 4, and 8. Serum antibody titers to HAs were measured at 2 weeks after the third immunization (week 10). a ELISA endpoint titers to H1N1 NC99 HA (left) and its gN38 variant (right). b Serum neutralizing antibody titers to NC99, SG86, and CA09 (H1N1) pseudoviruses. Shown are half-maximal inhibitory dilution of serum (IC50). c ELISA endpoint titers to H5N1 VN04 (left) and H7N9 AN13 HAs (right). d Serum neutralizing antibody titers to H5N1 VN04 and H7N9 AN13 pseudoviruses. Dotted lines indicate the lower detection limit of the assay. Data are presented as scattered dot plots with horizontal lines indicating geometric mean for each group. Statistical analysis was carried out by using nonparametric Kruskal–Wallis test with Dunn’s multiple comparisons. Displayed results are representative of two independent experiments (H1ssF WT and H1ssF gN38) or based on one experiment (H1ssF R38). Source data are provided as Source Data file.
Fig. 3. Specificity of serum neutralizing activity…
Fig. 3. Specificity of serum neutralizing activity elicited by H1 stem nanoparticle variants.
a Serum pseudovirus-neutralization activity in the presence of competitor proteins. Immune sera were pre-incubated with either NC99 WT HA, HA gN38, or irrelevant RSV F proteins prior to evaluation of SG86 (H1N1) pseudovirus neutralization. Serum neutralization IC50 titers were calculated in the absence or presence of competitor proteins. b Percent inhibition of virus neutralization was calculated for each competitor protein. Dotted lines indicate the lower detection limit of the assay. Data are presented as scattered dot plots with horizontal lines indicating geometric mean for each group. Statistical analysis was carried out by using nonparametric Kruskal–Wallis test with Dunn’s multiple comparisons. Displayed results are representative of two independent experiments. Source data are provided as Source Data file.
Fig. 4. Homotypic, heterosubtypic, and cross-group protection…
Fig. 4. Homotypic, heterosubtypic, and cross-group protection of mice immunized with H1 stem nanoparticle variants.
BALB/c mice (N = 10) were immunized with 2 µg of H1ssF WT, H1ssF gN38 or H1ssF R38 on weeks 0, 4, and 8, and challenged between weeks 16 and 18 with 20 × LD50 of A/California/07/09 (H1N1) virus (a), 25 × LD50 of A/Vietnam/1203/04 (H5N1) virus (b), or 10 × LD50 of A/Anhui/1/13 (H7N9) virus (c). All viruses were given intranasal inoculation. Mice were monitored twice daily for their weight and activity for 14 days post infection. Survival curve (left) and associated weight loss curve (right) were plotted for each virus challenge (ac). For weight loss curves, lines indicate group mean ± s.d. (shaded). Statistical test to compare multiple Kaplan–Meier curves was carried out by using Mantel-Cox log-rank test with Bonferroni correction. Displayed results are representative of two independent experiments (H1N1 and H5N1) or cumulative of two experiments (H7N9). Source data are provided as Source Data file.
Fig. 5. Cross-group neutralization and protective capacity…
Fig. 5. Cross-group neutralization and protective capacity of immunoglobulins isolated from mice immunized with H1 stem nanoparticle gN38 variant.
Immune Ig were obtained from H1ssF gN38-immunized mice by affinity-purifying with H7 (AN13) HA. a ELISA binding profile of purified Ig to HA from NC99, H5N1 IN05, H3N2 HK68, H7N9 AN13, and H10N8 JD13. b Neutralization profile of purified Ig to H5N1 VN04 (left), H3N2 HK68 (center), and H7N9 AN13 (right) pseudoviruses. Data are shown as mean ± s.d. c Passive immunization of purified Ig in naïve BALB/c mice (N = 10). Mice were given 0.2 mg of purified Ig from either naïve or H1ssF gN38-immunized mice, or bNAb FI6v3 (0.1 mg) intraperitoneally 24 h prior to H7N9 virus challenge (10 × LD50). Mice were monitored twice daily for their weight and activity for 14 days post infection. Statistical test to compare multiple Kaplan–Meier curves was carried out by using Mantel-Cox log-rank test with Bonferroni correction. Displayed results are from one experiment. Source data are provided as Source Data file.

References

    1. Skehel JJ, Wiley DC. Receptor binding and membrane fusion in virus entry: the influenza hemagglutinin. Annu Rev. Biochem. 2000;69:531–569. doi: 10.1146/annurev.biochem.69.1.531.
    1. Wharton, S. A., Weis, W., Skehel, J. J. & Wiley, D. C. In The Influenza Viruses 153–173 (Springer, Boston, MA, 1989).
    1. Gamblin SJ, Skehel JJ. Influenza hemagglutinin and neuraminidase membrane glycoproteins. J. Biol. Chem. 2010;285:28403–28409. doi: 10.1074/jbc.R110.129809.
    1. Wu Y, Wu Y, Tefsen B, Shi Y, Gao GF. Bat-derived influenza-like viruses H17N10 and H18N11. Trends Microbiol. 2014;22:183–191. doi: 10.1016/j.tim.2014.01.010.
    1. Nabel GJ, Fauci AS. Induction of unnatural immunity: prospects for a broadly protective universal influenza vaccine. Nat. Med. 2010;16:1389–1391. doi: 10.1038/nm1210-1389.
    1. Wu NC, Wilson IA. Structural insights into the design of novel anti-influenza therapies. Nat. Struct. Mol. Biol. 2018;25:115–121. doi: 10.1038/s41594-018-0025-9.
    1. Air GM. Influenza virus antigenicity and broadly neutralizing epitopes. Curr. Opin. Virol. 2015;11:113–121. doi: 10.1016/j.coviro.2015.03.006.
    1. Lee PS, Wilson IA. Structural characterization of viral epitopes recognized by broadly cross-reactive antibodies. Curr. Top. Microbiol. Immunol. 2015;386:323–341.
    1. Corti D, et al. Tackling influenza with broadly neutralizing antibodies. Curr. Opin. Virol. 2017;24:60–69. doi: 10.1016/j.coviro.2017.03.002.
    1. Corti D, et al. A neutralizing antibody selected from plasma cells that binds to group 1 and group 2 influenza A hemagglutinins. Science. 2011;333:850–856. doi: 10.1126/science.1205669.
    1. Dreyfus C, et al. Highly conserved protective epitopes on influenza B viruses. Science. 2012;337:1343–1348. doi: 10.1126/science.1222908.
    1. Ekiert DC, et al. Antibody recognition of a highly conserved influenza virus epitope. Science. 2009;324:246–251. doi: 10.1126/science.1171491.
    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. Sui J, et al. Structural and functional bases for broad-spectrum neutralization of avian and human influenza A viruses. Nat. Struct. Mol. Biol. 2009;16:265–273. doi: 10.1038/nsmb.1566.
    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. Wrammert J, et al. Broadly cross-reactive antibodies dominate the human B cell response against 2009 pandemic H1N1 influenza virus infection. J. Exp. Med. 2011;208:181–193. doi: 10.1084/jem.20101352.
    1. Henry Dunand CJ, et al. Preexisting human antibodies neutralize recently emerged H7N9 influenza strains. J. Clin. Invest. 2015;125:1255–1268. doi: 10.1172/JCI74374.
    1. Friesen RH, et al. A common solution to group 2 influenza virus neutralization. Proc. Natl Acad. Sci. USA. 2014;111:445–450. doi: 10.1073/pnas.1319058110.
    1. Tan GS, et al. Characterization of a broadly neutralizing monoclonal antibody that targets the fusion domain of group 2 influenza A virus hemagglutinin. J. Virol. 2014;88:13580–13592. doi: 10.1128/JVI.02289-14.
    1. Wu Y, et al. A potent broad-spectrum protective human monoclonal antibody crosslinking two haemagglutinin monomers of influenza A virus. Nat. Commun. 2015;6:7708. doi: 10.1038/ncomms8708.
    1. Kallewaard NL, et al. Structure and function analysis of an antibody recognizing all influenza A subtypes. Cell. 2016;166:596–608. doi: 10.1016/j.cell.2016.05.073.
    1. Joyce MG, et al. Vaccine-induced antibodies that neutralize group 1 and group 2 influenza A viruses. Cell. 2016;166:609–623. doi: 10.1016/j.cell.2016.06.043.
    1. Khanna M, Sharma S, Kumar B, Rajput R. Protective immunity based on the conserved hemagglutinin stalk domain and its prospects for universal influenza vaccine development. Biomed. Res. Int. 2014;2014:546274. doi: 10.1155/2014/546274.
    1. Krammer F, Palese P. Advances in the development of influenza virus vaccines. Nat. Rev. Drug Disco. 2015;14:167–182. doi: 10.1038/nrd4529.
    1. Laursen NS, Wilson IA. Broadly neutralizing antibodies against influenza viruses. Antivir. Res. 2013;98:476–483. doi: 10.1016/j.antiviral.2013.03.021.
    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;87:6542–6550. doi: 10.1128/JVI.00641-13.
    1. Eggink D, Goff PH, Palese P. Guiding the immune response against influenza virus hemagglutinin toward the conserved stalk domain by hyperglycosylation of the globular head domain. J. Virol. 2014;88:699–704. doi: 10.1128/JVI.02608-13.
    1. Margine I, et al. Hemagglutinin stalk-based universal vaccine constructs protect against group 2 influenza A viruses. J. Virol. 2013;87:10435–10446. doi: 10.1128/JVI.01715-13.
    1. Gocnik M, et al. Antibodies induced by the HA2 glycopolypeptide of influenza virus haemagglutinin improve recovery from influenza A virus infection. J. Gen. Virol. 2008;89:958–967. doi: 10.1099/vir.0.83524-0.
    1. Sagawa H, Ohshima A, Kato I, Okuno Y, Isegawa Y. The immunological activity of a deletion mutant of influenza virus haemagglutinin lacking the globular region. J. Gen. Virol. 1996;77:1483–1487. doi: 10.1099/0022-1317-77-7-1483.
    1. Steel J, et al. Influenza virus vaccine based on the conserved hemagglutinin stalk domain. MBio. 2010;1:e00018–e00020. doi: 10.1128/mBio.00018-10.
    1. Yassine HM, et al. Hemagglutinin-stem nanoparticles generate heterosubtypic influenza protection. Nat. Med. 2015;21:1065–1070. doi: 10.1038/nm.3927.
    1. Impagliazzo A, et al. A stable trimeric influenza hemagglutinin stem as a broadly protective immunogen. Science. 2015;349:1301–1306. doi: 10.1126/science.aac7263.
    1. Sutton TC, et al. Protective efficacy of influenza group 2 hemagglutinin stem-fragment immunogen vaccines. NPJ Vaccines. 2017;2:35. doi: 10.1038/s41541-017-0036-2.
    1. Andrews SF, Graham BS, Mascola JR, McDermott AB. Is it possible to develop a “universal” influenza virus vaccine? Immunogenetic considerations underlying B-cell biology in the development of a pan-subtype influenza A vaccine targeting the hemagglutinin stem. Cold Spring Harb. Perspect. Biol. 2018;10:a029413. doi: 10.1101/cshperspect.a029413.
    1. Lingwood D, et al. Structural and genetic basis for development of broadly neutralizing influenza antibodies. Nature. 2012;489:566–570. doi: 10.1038/nature11371.
    1. Whittle JR, et al. Flow cytometry reveals that H5N1 vaccination elicits cross-reactive stem-directed antibodies from multiple Ig heavy-chain lineages. J. Virol. 2014;88:4047–4057. doi: 10.1128/JVI.03422-13.
    1. Wheatley AK, et al. H5N1 vaccine-elicited memory B cells are genetically constrained by the IGHV locus in the recognition of a neutralizing epitope in the hemagglutinin stem. J. Immunol. 2015;195:602–610. doi: 10.4049/jimmunol.1402835.
    1. Wilson IA, Skehel JJ, Wiley DC. Structure of the haemagglutinin membrane glycoprotein of influenza virus at 3 A resolution. Nature. 1981;289:366–373. doi: 10.1038/289366a0.
    1. Corbett KS, et al. Design of nanoparticulate group 2 influenza virus hemagglutinin stem antigens that activate unmutated ancestor B cell receptors of broadly neutralizing antibody lineages. MBio. 2019;10:e02810–e02818. doi: 10.1128/mBio.02810-18.
    1. Okuno Y, Isegawa Y, Sasao F, Ueda S. A common neutralizing epitope conserved between the hemagglutinins of influenza A virus H1 and H2 strains. J. Virol. 1993;67:2552–2558. doi: 10.1128/JVI.67.5.2552-2558.1993.
    1. Ekiert DC, et al. A highly conserved neutralizing epitope on group 2 influenza A viruses. Science. 2011;333:843–850. doi: 10.1126/science.1204839.
    1. Kwakkenbos MJ, et al. Generation of stable monoclonal antibody-producing B cell receptor-positive human memory B cells by genetic programming. Nat. Med. 2010;16:123–128. doi: 10.1038/nm.2071.
    1. McLellan JS, et al. Structure-based design of a fusion glycoprotein vaccine for respiratory syncytial virus. Science. 2013;342:592–598. doi: 10.1126/science.1243283.
    1. Clementi N, et al. A human monoclonal antibody with neutralizing activity against highly divergent influenza subtypes. PLoS One. 2011;6:e28001. doi: 10.1371/journal.pone.0028001.
    1. Hu W, et al. Fully human broadly neutralizing monoclonal antibodies against influenza A viruses generated from the memory B cells of a 2009 pandemic H1N1 influenza vaccine recipient. Virology. 2013;435:320–328. doi: 10.1016/j.virol.2012.09.034.
    1. Nachbagauer R, et al. Hemagglutinin stalk immunity reduces influenza virus replication and transmission in ferrets. J. Virol. 2015;90:3268–3273. doi: 10.1128/JVI.02481-15.
    1. Lang S, et al. Antibody 27F3 broadly targets influenza A group 1 and 2 hemagglutinins through a further variation in VH1-69 antibody orientation on the HA stem. Cell Rep. 2017;20:2935–2943. doi: 10.1016/j.celrep.2017.08.084.
    1. Andrews SF, et al. Preferential induction of cross-group influenza A hemagglutinin stem-specific memory B cells after H7N9 immunization in humans. Sci. Immunol. 2017;2:eaan2676. doi: 10.1126/sciimmunol.aan2676.
    1. Liu Y, et al. Cross-lineage protection by human antibodies binding the influenza B hemagglutinin. Nat. Commun. 2019;10:324. doi: 10.1038/s41467-018-08165-y.
    1. Pappas L, et al. Rapid development of broadly influenza neutralizing antibodies through redundant mutations. Nature. 2014;516:418–422. doi: 10.1038/nature13764.
    1. Avnir Y, et al. Molecular signatures of hemagglutinin stem-directed heterosubtypic human neutralizing antibodies against influenza A viruses. PLoS Pathog. 2014;10:e1004103. doi: 10.1371/journal.ppat.1004103.
    1. Ohshima N, Kubota-Koketsu R, Iba Y, Okuno Y, Kurosawa Y. Two types of antibodies are induced by vaccination with A/California/2009 pdm virus: binding near the sialic acid-binding pocket and neutralizing both H1N1 and H5N1 viruses. PLoS One. 2014;9:e87305. doi: 10.1371/journal.pone.0087305.
    1. Dreyfus C, Ekiert DC, Wilson IA. Structure of a classical broadly neutralizing stem antibody in complex with a pandemic H2 influenza virus hemagglutinin. J. Virol. 2013;87:7149–7154. doi: 10.1128/JVI.02975-12.
    1. Wada Y, et al. A humanized mouse model identifies key amino acids for low immunogenicity of H7N9 vaccines. Sci. Rep. 2017;7:1283. doi: 10.1038/s41598-017-01372-5.
    1. Mulligan MJ, et al. Serological responses to an avian influenza A/H7N9 vaccine mixed at the point-of-use with MF59 adjuvant: a randomized clinical trial. JAMA. 2014;312:1409–1419. doi: 10.1001/jama.2014.12854.
    1. Mendoza M, et al. Generation and testing anti-influenza human monoclonal antibodies in a new humanized mouse model (DRAGA: HLA-A2. HLA-DR4. Rag1 KO. IL-2Rgammac KO. NOD) Hum. Vaccin Immunother. 2018;14:345–360. doi: 10.1080/21645515.2017.1403703.
    1. Ivanova II, et al. Targeting of influenza viral epitopes to antigen-presenting cells by genetically engineered chimeric molecules in a humanized NOD SCID Gamma Transfer Model. Hum. Gene Ther. 2018;29:1056–1070. doi: 10.1089/hum.2018.100.
    1. Sasaki E, et al. Development of a preclinical humanized mouse model to evaluate acute toxicity of an influenza vaccine. Oncotarget. 2018;9:25751–25763.
    1. Matsuda K, et al. Prolonged evolution of the memory B cell response induced by a replicating adenovirus-influenza H5 vaccine. Sci. Immunol. 2019;4:eaau2710. doi: 10.1126/sciimmunol.aau2710.
    1. Gostic KM, Ambrose M, Worobey M, Lloyd-Smith JO. Potent protection against H5N1 and H7N9 influenza via childhood hemagglutinin imprinting. Science. 2016;354:722–726. doi: 10.1126/science.aag1322.
    1. Cobey S, Hensley SE. Immune history and influenza virus susceptibility. Curr. Opin. Virol. 2017;22:105–111. doi: 10.1016/j.coviro.2016.12.004.
    1. Scheres SH. RELION: implementation of a Bayesian approach to cryo-EM structure determination. J. Struct. Biol. 2012;180:519–530. doi: 10.1016/j.jsb.2012.09.006.
    1. Zheng SQ, et al. MotionCor2: anisotropic correction of beam-induced motion for improved cryo-electron microscopy. Nat. Methods. 2017;14:331–332. doi: 10.1038/nmeth.4193.
    1. Rohou A, Grigorieff N. CTFFIND4: fast and accurate defocus estimation from electron micrographs. J. Struct. Biol. 2015;192:216–221. doi: 10.1016/j.jsb.2015.08.008.
    1. Tang G, et al. EMAN2: an extensible image processing suite for electron microscopy. J. Struct. Biol. 2007;157:38–46. doi: 10.1016/j.jsb.2006.05.009.
    1. Pettersen EF, et al. UCSF Chimera—a visualization system for exploratory research and analysis. J. Comput Chem. 2004;25:1605–1612. doi: 10.1002/jcc.20084.
    1. Kong WP, et al. Protective immunity to lethal challenge of the 1918 pandemic influenza virus by vaccination. Proc. Natl Acad. Sci. USA. 2006;103:15987–15991. doi: 10.1073/pnas.0607564103.
    1. Yang ZY, et al. Immunization by avian H5 influenza hemagglutinin mutants with altered receptor binding specificity. Science. 2007;317:825–828. doi: 10.1126/science.1135165.

Source: PubMed

3
Prenumerera