Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Article
  • Published:

Structure–function analysis of neutralizing antibodies to H7N9 influenza from naturally infected humans

Abstract

Little is known about the specificities and neutralization breadth of the H7-reactive antibody repertoire induced by natural H7N9 infection in humans. We have isolated and characterized 73 H7-reactive monoclonal antibodies from peripheral B cells from four donors infected in 2013 and 2014. Of these, 45 antibodies were H7-specific, and 17 of these neutralized the virus, albeit with few somatic mutations in their variable domain sequences. An additional set of 28 antibodies, isolated from younger donors born after 1968, cross-reacted between H7 and H3 haemagglutinins in binding assays, and had accumulated significantly more somatic mutations, but were predominantly non-neutralizing in vitro. Crystal structures of three neutralizing and protective antibodies in complex with the H7 haemagglutinin revealed that they recognize overlapping residues surrounding the receptor-binding site of haemagglutinin. One of the antibodies, L4A-14, bound into the sialic acid binding site and made contacts with haemagglutinin residues that were conserved in the great majority of 2016–2017 H7N9 isolates. However, only 3 of the 17 neutralizing antibodies retained activity for the Yangtze River Delta lineage viruses isolated in 2016–2017 that have undergone antigenic change, which emphasizes the need for updated H7N9 vaccines.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: Neutralization titres of convalescent sera from donors with laboratory-confirmed H7N9 infection.
Fig. 2: Structures of L4A-14/H7, L3A-44/H7 and L4B-18/H7 complexes.
Fig. 3: Comparison of epitope residues recognized by L4A-14, L3A-44 and L4B-18 antibodies.
Fig. 4: Protection against lethal infection with H7N9 in mice by antibodies.

Similar content being viewed by others

Data availability

The data that support the findings of this study are available from the corresponding authors upon request. Atomic coordinates for the H7-FabL4A-14, H7-FabL4B-18 and H7-FabL3A-44 complex as well as structure factors have been deposited to the Protein Data Bank under accession codes 6II4, 6II8 and 6II9.

References

  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. 17, 822–832 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  2. Shi, J. et al. H7N9 virulent mutants detected in chickens in China pose an increased threat to humans. Cell Res. 27, 1409–1421 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Yang, L. et al. Genesis and spread of newly emerged highly pathogenic H7N9 avian viruses in mainland China. J. Virol. 91, e01277-17 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  4. Zhang, F. et al. Human infections with recently-emerging highly pathogenic H7N9 avian influenza virus in China. J. Infect. 75, 71–75 (2017).

    Article  PubMed  Google Scholar 

  5. Zhu, W. et al. Biological characterisation of the emerged highly pathogenic avian influenza (HPAI) A(H7N9) viruses in humans, in mainland China, 2016 to 2017. Euro. Surveill. 22, 30533 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  6. Qi, W. et al. Emergence and adaptation of a novel highly pathogenic H7N9 influenza virus in birds and humans from a 2013 human-infecting low-pathogenic ancestor. J. Virol. 92, e00921-17 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  7. Gao, R. et al. Human infection with a novel avian-origin influenza A (H7N9) virus. N. Engl. J. Med. 368, 1888–1897 (2013).

    Article  CAS  PubMed  Google Scholar 

  8. Hu, Y. et al. Association between adverse clinical outcome in human disease caused by novel influenza A H7N9 virus and sustained viral shedding and emergence of antiviral resistance. Lancet 381, 2273–2279 (2013).

    Article  PubMed  Google Scholar 

  9. Hai, R. et al. Influenza A(H7N9) virus gains neuraminidase inhibitor resistance without loss of in vivo virulence or transmissibility. Nat. Commun. 4, 2854 (2013).

    Article  PubMed  Google Scholar 

  10. Lin, P. H. et al. Virological, serological, and antiviral studies in an imported human case of avian influenza A(H7N9) virus in Taiwan. Clin. Infect. Dis. 58, 242–246 (2014).

    Article  CAS  PubMed  Google Scholar 

  11. Marjuki, H. et al. Neuraminidase mutations conferring resistance to oseltamivir in influenza A(H7N9) viruses. J. Virol. 89, 5419–5426 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Imai, M. et al. A highly pathogenic avian H7N9 influenza virus isolated from a human is lethal in some ferrets infected via respiratory droplets. Cell Host Microbe 22, 615–626 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Subbarao, K. Avian influenza H7N9 viruses: a rare second warning. Cell Res. 28, 1–2 (2018).

    Article  CAS  PubMed  Google Scholar 

  14. Summary of Influenza Risk Assessment Tool (IRAT) Results Web Site (Centers for Disease Control and Prevention, accessed 2 May 2018); https://www.cdc.gov/flu/pandemic-resources/monitoring/irat-virus-summaries.htm.

  15. Wang, D. et al. Two outbreak sources of influenza A (H7N9) viruses have been established in China. J. Virol. 90, 5561–5573 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Kile, J. C. et al. Update: increase in human infections with novel Asian lineage avian influenza A(H7N9) viruses during the fifth epidemic—China, October 1, 2016–August 7, 2017. MMWR. Morb. Mortal. Wkly Rep. 66, 928–932 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  17. Gostic, K. M., Ambrose, M., Worobey, M. & Lloyd-Smith, J. O. Potent protection against H5N1 and H7N9 influenza via childhood hemagglutinin imprinting. Science 354, 722–726 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Ekiert, D. C. et al. A highly conserved neutralizing epitope on group 2 influenza A viruses. Science 333, 843–850 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Guo, L. et al. Human antibody responses to avian influenza A(H7N9) virus, 2013. Emerg. Infect. Dis. 20, 192–200 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  20. Dunand, C. J. et al. Preexisting human antibodies neutralize recently emerged H7N9 influenza strains. J. Clin. Invest. 125, 1255–1268 (2015).

    Article  Google Scholar 

  21. Kallewaard, N. L. et al. Structure and function analysis of an antibody recognizing all influenza A subtypes. Cell 166, 596–608 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Yamayoshi, S. et al. Human protective monoclonal antibodies against the HA stem of group 2 HAs derived from an H3N2 virus-infected human. J. Infect. 76, 177–185 (2018).

    Article  PubMed  Google Scholar 

  23. Stadlbauer, D. et al. Vaccination with a recombinant H7 hemagglutinin-based influenza virus vaccine induces broadly reactive antibodies in humans. mSphere 2, e00502-17 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  24. Stadlbauer, D., Nachbagauer, R., Meade, P. & Krammer, F. Universal influenza virus vaccines: what can we learn from the human immune response following exposure to H7 subtype viruses? Front. Med. 11, 471–479 (2017).

    Article  PubMed  Google Scholar 

  25. Andrews, S. F. et al. Preferential induction of cross-group influenza A hemagglutinin stem-specific memory B cells after H7N9 immunization in humans. Sci. Immunol. 2, eaan2676 (2017).

    Article  PubMed  Google Scholar 

  26. Wang, Z. et al. Recovery from severe H7N9 disease is associated with diverse response mechanisms dominated by CD8+ T cells. Nat. Commun. 6, 6833 (2015).

    Article  CAS  PubMed  Google Scholar 

  27. Baz, M. et al. Nonreplicating influenza A virus vaccines confer broad protection against lethal challenge. mBio 6, e01487-15 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  28. Sutton, T. C. et al. Protective efficacy of influenza group 2 hemagglutinin stem-fragment immunogen vaccines. NPJ Vaccines 2, 35 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  29. Sobhanie, M. et al. Evaluation of the safety and immunogenicity of a candidate pandemic live attenuated influenza vaccine (pLAIV) against influenza A(H7N9). J. Infect. Dis. 213, 922–929 (2016).

    Article  CAS  PubMed  Google Scholar 

  30. Dunand, C. J. et al. Both neutralizing and non-neutralizing human H7N9 influenza vaccine-induced monoclonal antibodies confer protection. Cell Host Microbe 19, 800–813 (2016).

    Article  Google Scholar 

  31. Powell, T. J., Silk, J. D., Sharps, J., Fodor, E. & Townsend, A. R. Pseudotyped influenza A virus as a vaccine for the induction of heterotypic immunity. J. Virol. 86, 13397–13406 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Burke, D. F. & Smith, D. J. A recommended numbering scheme for influenza A HA subtypes. PLoS ONE 9, e112302 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  33. Thornburg, N. J. et al. H7N9 influenza virus neutralizing antibodies that possess few somatic mutations. J. Clin. Invest. 126, 1482–1494 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  34. Matrosovich, M., Matrosovichm, T., Carr, J., Roberts, N. A. & Klenk, H. D. Overexpression of the alpha-2,6-sialyltransferase in MDCK cells increases influenza virus sensitivity to neuraminidase inhibitors. J. Virol. 77, 8418–8425 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Wiley, D. C. & Skehel, J. J. The structure and function of the hemagglutinin membrane glycoprotein of influenza virus. Annu. Rev. Biochem. 56, 365–394 (1987).

    Article  CAS  PubMed  Google Scholar 

  36. Chen, C. et al. Structural insight into a human neutralizing antibody against influenza virus H7N9. J. Virol. 92, e01850-17 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  37. Shi, Y. et al. Structures and receptor binding of hemagglutinins from human-infecting H7N9 influenza viruses. Science 342, 243–247 (2013).

    Article  CAS  PubMed  Google Scholar 

  38. Ohshima, N. et al. Naturally occurring antibodies in humans can neutralize a variety of influenza virus strains, including H3, H1, H2, and H5. J. Virol. 85, 11048–11057 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Lee, P. S. et al. Receptor mimicry by antibody F045-092 facilitates universal binding to the H3 subtype of influenza virus. Nat. Commun. 5, 3614 (2014).

    Article  PubMed  Google Scholar 

  40. McCarthy, K. R. et al. Memory B cells that cross-react with Group 1 and Group 2 influenza A viruses are abundant in adult human repertoires. Immunity 48, 174–184 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Ekiert, D. C. et al. Cross-neutralization of influenza A viruses mediated by a single antibody loop. Nature 489, 526–532 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Huang, K. Y. et al. Focused antibody response to influenza linked to antigenic drift. J. Clin. Invest. 125, 2631–2645 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  43. Smith, K. et al. Rapid generation of fully human monoclonal antibodies specific to a vaccinating antigen. Nat. Protoc. 4, 372–384 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Xiao, J. H. et al. Characterization of influenza virus pseudotyped with Ebolavirus glycoprotein. J. Virol. 92, e00941-17 (2018).

    PubMed  PubMed Central  Google Scholar 

  45. Martínez-Sobrido, L. et al. Hemagglutinin-pseudotyped green fluorescent protein-expressing influenza viruses for the detection of influenza virus neutralizing antibodies. J. Virol. 84, 2157–2163 (2010).

    Article  PubMed  Google Scholar 

  46. Rowe, T. et al. Detection of antibody to avian influenza A (H5N1) virus in human serum by using a combination of serologic assays. J. Clin. Microbiol. 37, 937–943 (1999).

    CAS  PubMed  PubMed Central  Google Scholar 

  47. Peacock, T. et al. Antigenic mapping of an H9N2 avian influenza virus reveals two discrete antigenic sites and a novel mechanism of immune escape. Sci. Rep. 6, 18745 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Zhang, W. et al. Crystal structure of the swine-origin A (H1N1)-2009 influenza A virus hemagglutinin (HA) reveals similar antigenicity to that of the 1918 pandemic virus. Protein Cell 1, 459–467 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Aad, G. et al. Search for new particles in two-jet final states in 7Â TeV proton-proton collisions with the ATLAS detector at the LHC. Phys. Rev. Lett. 105, 161801 (2010).

    Article  CAS  PubMed  Google Scholar 

  50. Yang, J.-R. et al. Characterization of influenza A (H7N9) viruses isolated from human cases imported into Taiwan. PLoS ONE 10, e0119792 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

The authors acknowledge support from the laboratories that provided the sequences of reference antibody MEDI885221 for the comparisons in the tables and figures. The authors also acknowledge the BD FACSAria cell sorter service provided by the Core Instrument Center of Chang Gung University, and C. Waugh for FACS sorting B cells and MDCK-SIAT1 cell lines at WIMM Oxford. These studies were funded by the Townsend-Jeantet Charitable Trust (registered charity no. 1011770), the Emergency Technology Research Issue on Prevention and Control for Human infection with A (H7N9) Avian Influenza Virus (10600100000015001206), the Strategic Priority Research Program of the Chinese Academy of Sciences (CAS) (XDB29010000), the National Science and Technology Major Project (2018ZX10101004), the Human Immunology Unit (MRC), Oxford University, Chang Gung Medical Research Program (CMRPG3G0921, CMRPG3G0922) and the Ministry of Science and Technology of Taiwan (MOST 104-2320-B-182A-002-MY2, MOST 105-2320-B-182A-008- and MOST 107-2321-B-182A-003-). G.F.G. is supported partly as a leading principal investigator of the NSFC Innovative Research Group (81621091). Y.S. is supported by the Excellent Young Scientist Program from the National Natural Science Foundation of China (81622031), the Excellent Young Scientist Program of CAS and the Youth Innovation Promotion Association CAS (2015078).

Author information

Authors and Affiliations

Authors

Contributions

K.-Y.A.H., G.F.G., Y.S. and A.R.T. conceived and designed the study. K.-Y.A.H., P.R., H.J., B.W., L.S., T.D., Y.-M.L., P.C., M.I., M.-C.W., Z.C., R.S., C.-C.H., J.-H.Y., J.Q., T.-Y.L., A.L., T.J.P., J.-T.J., C.M., G.F.G., Y.S. and A.R.T. carried out the experiments. K.-Y.A.H., P.R., H.J., B.W., L.S., T.D., Y.-M.L., J.-T.J., C.M., G.F.G., Y.S. and A.R.T. performed data analysis and figure/table preparation. K.-Y.A.H., P.R., G.F.G., Y.S. and A.R.T. wrote the manuscript.

Corresponding authors

Correspondence to Kuan-Ying A. Huang, George F. Gao, Yi Shi or Alain R. Townsend.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Publisher’s note: Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Supplementary information

Supplementary Information

Supplementary Figures 1–8, Supplementary Tables 1–6.

Reporting Summary

Supplementary Table 1

Binding and neutralizing activities of of 73 H7-reactive monoclonal antibodies isolated from donors.

Supplementary table 2

Details of 73 H7-reactive monoclonal antibodies variable domain sequences isolated from donors.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Huang, KY.A., Rijal, P., Jiang, H. et al. Structure–function analysis of neutralizing antibodies to H7N9 influenza from naturally infected humans. Nat Microbiol 4, 306–315 (2019). https://doi.org/10.1038/s41564-018-0303-7

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41564-018-0303-7

This article is cited by

Search

Quick links

Nature Briefing Microbiology

Sign up for the Nature Briefing: Microbiology newsletter — what matters in microbiology research, free to your inbox weekly.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing: Microbiology