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:

Broadly neutralizing hemagglutinin stalk–specific antibodies require FcγR interactions for protection against influenza virus in vivo

Subjects

Abstract

Neutralizing antibodies against influenza viruses have traditionally been thought to provide protection exclusively through their variable region; the contributions of mechanisms conferred by the Fc domain remain controversial. We investigated the in vivo contributions of Fc interactions with their cognate receptors for a collection of neutralizing anti-influenza antibodies. Whereas five broadly neutralizing monoclonal antibodies (bNAbs) targeting the conserved stalk region of hemagglutinin (HA) required interactions between the antibody Fc and Fc receptors for IgG (FcγRs) to confer protection from lethal H1N1 challenge, three strain-specific monoclonal Abs (mAbs) against the variable head domain of HA were equally protective in the presence or absence of FcγR interactions. Although all antibodies blocked infection, only anti-stalk bNAbs were capable of mediating cytotoxicity of infected cells, which accounts for their FcγR dependence. Immune complexes generated with anti–HA stalk mAb efficiently interacted with FcγRs, but anti–HA head immune complexes did not. These results suggest that FcγR binding capacity by anti-HA antibodies was dependent on the interaction of the cognate Fab with antigen. We exploited these disparate mechanisms of mAb-mediated protection to reengineer an anti-stalk bNAb to selectively enhance FcγR engagement to augment its protective activity. These findings reveal a previously uncharacterized property of bNAbs and guide an approach toward enhancing mAb-mediated antiviral therapeutics.

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

Figure 1: Fc-FcγR interactions are required for protection from viral infection by an anti–HA stalk bNAb in vivo.
Figure 2: Activating FcγRs are required for bNAb-mediated protection from viral infection in vivo.
Figure 3: Two strain-specific anti–H1 head mAbs do not require FcγR contributions during protection from viral infection in vivo.
Figure 4: Anti–HA stalk mAbs function through FcγRs after viral entry and induce superior ADCC compared to anti-head mAb.
Figure 5: Selectively enhancing huFc-huFcγR interactions augments bNAb-mediated protection from viral infection in vivo.

Similar content being viewed by others

References

  1. World Health Organization. Influenza (Seasonal) Fact sheet 211 http://www.who.int/mediacentre/factsheets/fs211/en/ (2009).

  2. Ahmed, R., Oldstone, M.B. & Palese, P. Protective immunity and susceptibility to infectious diseases: lessons from the 1918 influenza pandemic. Nat. Immunol. 8, 1188–1193 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Martinez, O., Tsibane, T. & Basler, C.F. Neutralizing anti-influenza virus monoclonal antibodies: therapeutics and tools for discovery. Int. Rev. Immunol. 28, 69–92 (2009).

    Article  CAS  PubMed  Google Scholar 

  4. Couch, R.B. & Kasel, J.A. Immunity to influenza in man. Annu. Rev. Microbiol. 37, 529–549 (1983).

    Article  CAS  PubMed  Google Scholar 

  5. Wang, T.T. & Palese, P. Biochemistry. Catching a moving target. Science 333, 834–835 (2011).

    Article  CAS  PubMed  Google Scholar 

  6. Skehel, J.J. & Wiley, D.C. Receptor binding and membrane fusion in virus entry: the influenza hemagglutinin. Annu. Rev. Biochem. 69, 531–569 (2000).

    Article  CAS  PubMed  Google Scholar 

  7. Knossow, M. et al. Mechanism of neutralization of influenza virus infectivity by antibodies. Virology 302, 294–298 (2002).

    Article  CAS  PubMed  Google Scholar 

  8. Wiley, D.C., Wilson, I.A. & Skehel, J.J. Structural identification of the antibody-binding sites of Hong Kong influenza haemagglutinin and their involvement in antigenic variation. Nature 289, 373–378 (1981).

    Article  CAS  PubMed  Google Scholar 

  9. Dreyfus, C. et al. Highly conserved protective epitopes on influenza B viruses. Science 337, 1343–1348 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Barbey-Martin, C. et al. An antibody that prevents the hemagglutinin low pH fusogenic transition. Virology 294, 70–74 (2002).

    Article  CAS  PubMed  Google Scholar 

  11. Ekiert, D.C. et al. Antibody recognition of a highly conserved influenza virus epitope. Science 324, 246–251 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Sui, J. et al. Structural and functional bases for broad-spectrum neutralization of avian and human influenza A viruses. Nat. Struct. Mol. Biol. 16, 265–273 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Corti, D. et al. A neutralizing antibody selected from plasma cells that binds to group 1 and group 2 influenza A hemagglutinins. Science 333, 850–856 (2011).

    Article  CAS  PubMed  Google Scholar 

  14. Abboud, N. et al. A requirement for FcgγR in antibody-mediated bacterial toxin neutralization. J. Exp. Med. 207, 2395–2405 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Bournazos, S., Chow, S.K., Abboud, N., Casadevall, A. & Ravetch, J.V. Human IgG Fc domain engineering enhances anti-toxin neutralizing antibody activity. J. Clin. Invest. (in the press).

  16. Schmitz, N. et al. Universal vaccine against influenza virus: linking TLR signaling to anti-viral protection. Eur. J. Immunol. 42, 863–869 (2012).

    Article  CAS  PubMed  Google Scholar 

  17. Huber, V.C., Lynch, J.M., Bucher, D.J., Le, J. & Metzger, D.W. Fc receptor–mediated phagocytosis makes a significant contribution to clearance of influenza virus infections. J. Immunol. 166, 7381–7388 (2001).

    Article  CAS  PubMed  Google Scholar 

  18. Nimmerjahn, F. & Ravetch, J.V. Fcγ receptors as regulators of immune responses. Nat. Rev. Immunol. 8, 34–47 (2008).

    Article  CAS  PubMed  Google Scholar 

  19. Nimmerjahn, F. & Ravetch, J.V. Divergent immunoglobulin g subclass activity through selective Fc receptor binding. Science 310, 1510–1512 (2005).

    Article  CAS  PubMed  Google Scholar 

  20. Tan, G.S. et al. A pan-H1 anti-hemagglutinin monoclonal antibody with potent broad-spectrum efficacy in vivo. J. Virol. 86, 6179–6188 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Li, G.M. et al. Pandemic H1N1 influenza vaccine induces a recall response in humans that favors broadly cross-reactive memory B cells. Proc. Natl. Acad. Sci. USA 109, 9047–9052 (2012).

    Article  PubMed  PubMed Central  Google Scholar 

  22. Reale, M.A. et al. Characterization of monoclonal antibodies specific for sequential influenza A/PR/8/34 virus variants. J. Immunol. 137, 1352–1358 (1986).

    CAS  PubMed  Google Scholar 

  23. Wrammert, J. et al. Broadly cross-reactive antibodies dominate the human B cell response against 2009 pandemic H1N1 influenza virus infection. J. Exp. Med. 208, 181–193 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Smith, P., DiLillo, D.J., Bournazos, S., Li, F. & Ravetch, J.V. Mouse model recapitulating human Fcγ receptor structural and functional diversity. Proc. Natl. Acad. Sci. USA 109, 6181–6186 (2012).

    Article  PubMed  PubMed Central  Google Scholar 

  25. Manicassamy, B. et al. Protection of mice against lethal challenge with 2009 H1N1 influenza A virus by 1918-like and classical swine H1N1 based vaccines. PLoS Pathog. 6, e1000745 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Jegaskanda, S. et al. Cross-reactive influenza-specific antibody-dependent cellular cytotoxicity antibodies in the absence of neutralizing antibodies. J. Immunol. 190, 1837–1848 (2013).

    Article  CAS  PubMed  Google Scholar 

  27. Jegaskanda, S., Weinfurter, J.T., Friedrich, T.C. & Kent, S.J. Antibody-dependent cellular cytotoxicity (ADCC) is associated with control of pandemic H1N1 influenza virus infection of macaques. J. Virol. 87, 5512–5522 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Srivastava, V. et al. Identification of dominant ADCC epitopes on hemagglutinin antigen of pandemic H1N1 influenza virus. J. Virol. 87, 5831–5840 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Alter, G., Malenfant, J.M. & Altfeld, M. CD107a as a functional marker for the identification of natural killer cell activity. J. Immunol. Methods 294, 15–22 (2004).

    Article  CAS  PubMed  Google Scholar 

  30. Nimmerjahn, F. & Ravetch, J.V. Analyzing antibody–Fc-receptor interactions. Methods Mol. Biol. 415, 151–162 (2008).

    CAS  PubMed  Google Scholar 

  31. El Bakkouri, K. et al. Universal vaccine based on ectodomain of matrix protein 2 of influenza A: Fc receptors and alveolar macrophages mediate protection. J. Immunol. 186, 1022–1031 (2011).

    Article  CAS  PubMed  Google Scholar 

  32. Jegerlehner, A., Schmitz, N., Storni, T. & Bachmann, M.F. Influenza A vaccine based on the extracellular domain of M2: weak protection mediated via antibody-dependent NK cell activity. J. Immunol. 172, 5598–5605 (2004).

    Article  CAS  PubMed  Google Scholar 

  33. Tedder, T.F., Baras, A. & Xiu, Y. Fcγ receptor–dependent effector mechanisms regulate CD19 and CD20 antibody immunotherapies for B lymphocyte malignancies and autoimmunity. Springer Semin. Immunopathol. 28, 351–364 (2006).

    Article  CAS  PubMed  Google Scholar 

  34. Nordstrom, J.L. et al. Anti-tumor activity and toxicokinetics analysis of MGAH22, an anti-HER2 monoclonal antibody with enhanced Fcγ receptor binding properties. Breast Cancer Res. 13, R123 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Clynes, R.A., Towers, T.L., Presta, L.G. & Ravetch, J.V. Inhibitory Fc receptors modulate in vivo cytotoxicity against tumor targets. Nat. Med. 6, 443–446 (2000).

    Article  CAS  PubMed  Google Scholar 

  36. Yang, X. et al. Cetuximab-mediated tumor regression depends on innate and adaptive immune responses. Mol. Ther. 21, 91–100 (2013).

    Article  CAS  PubMed  Google Scholar 

  37. Belser, J.A., Katz, J.M. & Tumpey, T.M. The ferret as a model organism to study influenza A virus infection. Dis. Model. Mech. 4, 575–579 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Nimmerjahn, F. & Ravetch, J.V. Antibodies, Fc receptors and cancer. Curr. Opin. Immunol. 19, 239–245 (2007).

    Article  CAS  PubMed  Google Scholar 

  39. Nimmerjahn, F. & Ravetch, J.V. FcγRs in health and disease. Curr. Top. Microbiol. Immunol. 350, 105–125 (2011).

    CAS  PubMed  Google Scholar 

  40. Barkas, T. & Watson, C.M. Induction of an Fc conformational change by binding of antigen: the generation of protein A–reactive sites in chicken immunoglobulin. Immunology 36, 557–561 (1979).

    CAS  PubMed  PubMed Central  Google Scholar 

  41. Kilàr, F. & Zavodszky, P. Non-covalent interactions between Fab and Fc regions in immunoglobulin G molecules. Hydrogen-deuterium exchange studies. Eur. J. Biochem. 162, 57–61 (1987).

    Article  PubMed  Google Scholar 

  42. Torres, M., Fernandez-Fuentes, N., Fiser, A. & Casadevall, A. The immunoglobulin heavy chain constant region affects kinetic and thermodynamic parameters of antibody variable region interactions with antigen. J. Biol. Chem. 282, 13917–13927 (2007).

    Article  CAS  PubMed  Google Scholar 

  43. Oda, M., Kozono, H., Morii, H. & Azuma, T. Evidence of allosteric conformational changes in the antibody constant region upon antigen binding. Int. Immunol. 15, 417–426 (2003).

    Article  CAS  PubMed  Google Scholar 

  44. Schlessinger, J., Steinberg, I.Z., Givol, D., Hochman, J. & Pecht, I. Antigen-induced conformational changes in antibodies and their Fab fragments studied by circular polarization of fluorescence. Proc. Natl. Acad. Sci. USA 72, 2775–2779 (1975).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Tong, H. et al. Peptide-conjugation induced conformational changes in human IgG1 observed by optimized negative-staining and individual-particle electron tomography. Sci. Rep. 3, 1089 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Takai, T., Li, M., Sylvestre, D., Clynes, R. & Ravetch, J.V. FcR γ chain deletion results in pleiotrophic effector cell defects. Cell 76, 519–529 (1994).

    Article  CAS  PubMed  Google Scholar 

  47. Takai, T., Ono, M., Hikida, M., Ohmori, H. & Ravetch, J.V. Augmented humoral and anaphylactic responses in FcγRII-deficient mice. Nature 379, 346–349 (1996).

    Article  CAS  PubMed  Google Scholar 

  48. Ioan-Facsinay, A. et al. FcγRI (CD64) contributes substantially to severity of arthritis, hypersensitivity responses, and protection from bacterial infection. Immunity 16, 391–402 (2002).

    Article  CAS  PubMed  Google Scholar 

  49. Hazenbos, W.L. et al. Impaired IgG-dependent anaphylaxis and Arthus reaction in Fc γ RIII (CD16) deficient mice. Immunity 5, 181–188 (1996).

    Article  CAS  PubMed  Google Scholar 

  50. Nimmerjahn, F. et al. FcγRIV deletion reveals its central role for IgG2a and IgG2b activity in vivo. Proc. Natl. Acad. Sci. USA 107, 19396–19401 (2010).

    Article  PubMed  PubMed Central  Google Scholar 

  51. Li, F. & Ravetch, J.V. Inhibitory Fcγ receptor engagement drives adjuvant and anti-tumor activities of agonistic CD40 antibodies. Science 333, 1030–1034 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Nimmerjahn, F., Bruhns, P., Horiuchi, K. & Ravetch, J.V. FcγRIV: a novel FcR with distinct IgG subclass specificity. Immunity 23, 41–51 (2005).

    Article  CAS  PubMed  Google Scholar 

  53. Wang, T.T. et al. Broadly protective monoclonal antibodies against H3 influenza viruses following sequential immunization with different hemagglutinins. PLoS Pathog. 6, e1000796 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank P. Wilson (University of Chicago), J. Wrammert (Emory University) and R. Ahmed (Emory University) for providing mAb constructs, F. Krammer (Mt. Sinai Medical Center) for providing recombinant soluble HA protein, S. Bournazos for assistance with surface plasmon resonance studies and J. Carroll and P. Smith for technical assistance. We also thank the Biodefense and Emerging Infections Research Resources Repository for supplying recombinant soluble HA protein. Research reported in this publication was partially supported by the National Institute of Allergy and Infectious Disease of the US National Institutes of Health (NIH) under award numbers P01AI081677, R01AI035875 and U54AI057158 to J.V.R. and AI097092 to P.P. Research support was also provided by the Bill & Melinda Gates Foundation grant OPP1033115 to J.V.R. This work was also supported in part by the US Army Research Laboratory and the US Army Research Office under contract number W911NF-13-2-0036 (to J.V.R.). Partial support for P.P. was provided by the Program for Appropriate Technology in Health and by the NIH-funded Center for Research on Influenza Pathogenesis (HHSN266200700010C). D.J.D. received fellowship support from the Leukemia and Lymphoma Society and received funding support from the New York Community Trust.

Author information

Authors and Affiliations

Authors

Contributions

D.J.D., G.S.T., P.P. and J.V.R. designed experiments, D.J.D. and G.S.T. performed the experiments, D.J.D., G.S.T., P.P. and J.V.R. analyzed data and D.J.D. and J.V.R. wrote the paper.

Corresponding author

Correspondence to Jeffrey V Ravetch.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–15 and Supplementary Table 1 (PDF 1290 kb)

Rights and permissions

Reprints and permissions

About this article

Cite this article

DiLillo, D., Tan, G., Palese, P. et al. Broadly neutralizing hemagglutinin stalk–specific antibodies require FcγR interactions for protection against influenza virus in vivo. Nat Med 20, 143–151 (2014). https://doi.org/10.1038/nm.3443

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nm.3443

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

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