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.

  • Letter
  • Published:

Enhanced neonatal Fc receptor function improves protection against primate SHIV infection

Subjects

Abstract

To protect against human immunodeficiency virus (HIV-1) infection, broadly neutralizing antibodies (bnAbs) must be active at the portals of viral entry in the gastrointestinal or cervicovaginal tracts. The localization and persistence of antibodies at these sites is influenced by the neonatal Fc receptor (FcRn)1,2, whose role in protecting against infection in vivo has not been defined. Here, we show that a bnAb with enhanced FcRn binding has increased gut mucosal tissue localization, which improves protection against lentiviral infection in non-human primates. A bnAb directed to the CD4-binding site of the HIV-1 envelope (Env) protein (denoted VRC01)3 was modified by site-directed mutagenesis to increase its binding affinity for FcRn. This enhanced FcRn-binding mutant bnAb, denoted VRC01-LS, displayed increased transcytosis across human FcRn-expressing cellular monolayers in vitro while retaining FcγRIIIa binding and function, including antibody-dependent cell-mediated cytotoxicity (ADCC) activity, at levels similar to VRC01 (the wild type). VRC01-LS had a threefold longer serum half-life than VRC01 in non-human primates and persisted in the rectal mucosa even when it was no longer detectable in the serum. Notably, VRC01-LS mediated protection superior to that afforded by VRC01 against intrarectal infection with simian–human immunodeficiency virus (SHIV). These findings suggest that modification of FcRn binding provides a mechanism not only to increase serum half-life but also to enhance mucosal localization that confers immune protection. Mutations that enhance FcRn function could therefore increase the potency and durability of passive immunization strategies to prevent HIV-1 infection.

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: In vitro evaluation of VRC01 and its FcRn-binding mutants.
Figure 2: Pharmacokinetic study in Indian rhesus macaques.
Figure 3: VRC01-LS affords greater protection against intrarectal SHIV BaLP4 challenge than VRC01.

Similar content being viewed by others

References

  1. Roopenian, D. C. & Akilesh, S. FcRn: the neonatal Fc receptor comes of age. Nature Rev. Immunol. 7, 715–725 (2007)

    Article  CAS  Google Scholar 

  2. Ward, E. S. & Ober, R. J. Multitasking by exploitation of intracellular transport functions: the many faces of FcRn. Adv. Immunol. 103, 77–115 (2009)

    Article  CAS  Google Scholar 

  3. Wu, X. et al. Rational design of envelope identifies broadly neutralizing human monoclonal antibodies to HIV-1. Science 329, 856–861 (2010)

    Article  ADS  CAS  Google Scholar 

  4. Hessell, A. J. et al. Fc receptor but not complement binding is important in antibody protection against HIV. Nature 449, 101–104 (2007)

    Article  ADS  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

  6. Hessell, A. J. et al. Broadly neutralizing human anti-HIV antibody 2G12 is effective in protection against mucosal SHIV challenge even at low serum neutralizing titers. PLoS Pathogens 5, e1000433 (2009)

    Article  Google Scholar 

  7. Walker, L. M. et al. Broad and potent neutralizing antibodies from an African donor reveal a new HIV-1 vaccine target. Science 326, 285–289 (2009)

    Article  ADS  CAS  Google Scholar 

  8. Hessell, A. J. et al. Broadly neutralizing monoclonal antibodies 2F5 and 4E10 directed against the human immunodeficiency virus type 1 gp41 membrane-proximal external region protect against mucosal challenge by simian–human immunodeficiency virus SHIVBa-L. J. Virol. 84, 1302–1313 (2010)

    Article  CAS  Google Scholar 

  9. Walker, L. M. et al. Broad neutralization coverage of HIV by multiple highly potent antibodies. Nature 477, 466–470 (2011)

    Article  ADS  CAS  Google Scholar 

  10. Huang, J. et al. Broad and potent neutralization of HIV-1 by a gp41-specific human antibody. Nature 491, 406–412 (2012)

    Article  ADS  CAS  Google Scholar 

  11. Moldt, B. et al. Highly potent HIV-specific antibody neutralization in vitro translates into effective protection against mucosal SHIV challenge in vivo. Proc. Natl Acad. Sci. USA 109, 18921–18925 (2012)

    Article  ADS  CAS  Google Scholar 

  12. Bitonti, A. J. et al. Pulmonary delivery of an erythropoietin Fc fusion protein in non-human primates through an immunoglobulin transport pathway. Proc. Natl Acad. Sci. USA 101, 9763–9768 (2004)

    Article  ADS  CAS  Google Scholar 

  13. Dall’ Acqua, W. F., Kiener, P. A. & Wu, H. Properties of human IgG1s engineered for enhanced binding to the neonatal Fc receptor (FcRn). J. Biol. Chem. 281, 23514–23524 (2006)

    Article  Google Scholar 

  14. Hinton, P. R. et al. An engineered human IgG1 antibody with longer serum half-life. J. Immunol. 176, 346–356 (2006)

    Article  CAS  Google Scholar 

  15. Petkova, S. B. et al. Enhanced half-life of genetically engineered human IgG1 antibodies in a humanized FcRn mouse model: potential application in humorally mediated autoimmune disease. Int. Immunol. 18, 1759–1769 (2006)

    Article  CAS  Google Scholar 

  16. Zalevsky, J. et al. Enhanced antibody half-life improves in vivo activity. Nature Biotechnol. 28, 157–159 (2010)

    Article  CAS  Google Scholar 

  17. Dall’ Acqua, W. F. et al. Increasing the affinity of a human IgG1 for the neonatal Fc receptor: biological consequences. J. Immunol. 169, 5171–5180 (2002)

    Article  Google Scholar 

  18. Roopenian, D. C. et al. The MHC class I-like IgG receptor controls perinatal IgG transport, IgG homeostasis, and fate of IgG–Fc-coupled drugs. J. Immunol. 170, 3528–3533 (2003)

    Article  CAS  Google Scholar 

  19. Smalls-Mantey, A. et al. Antibody-dependent cellular cytotoxicity against primary HIV-infected CD4+ T cells is directly associated with the magnitude of surface IgG binding. J. Virol. 86, 8672–8680 (2012)

    Article  CAS  Google Scholar 

  20. Gómez-Román, V. R. et al. A simplified method for the rapid fluorometric assessment of antibody-dependent cell-mediated cytotoxicity. J. Immunol. Methods 308, 53–67 (2006)

    Article  Google Scholar 

  21. Klein, K. et al. Neutralizing IgG at the portal of infection mediates protection against vaginal simian/human immunodeficiency virus challenge. J. Virol. 87, 11604–11616 (2013)

    Article  CAS  Google Scholar 

  22. Kwong, P. D., Mascola, J. R. & Nabel, G. J. Broadly neutralizing antibodies and the search for an HIV-1 vaccine: the end of the beginning. Nature Rev. Immunol. 13, 693–701 (2013)

    Article  CAS  Google Scholar 

  23. Onorato, I. M. et al. Mucosal immunity induced by enhance-potency inactivated and oral polio vaccines. J. Infect. Dis. 163, 1–6 (1991)

    Article  CAS  Google Scholar 

  24. Sicinski, P. et al. Poliovirus type 1 enters the human host through intestinal M cells. Gastroenterology 98, 56–58 (1990)

    Article  CAS  Google Scholar 

  25. Yoshida, M. et al. Human neonatal Fc receptor mediates transport of IgG into luminal secretions for delivery of antigens to mucosal dendritic cells. Immunity 20, 769–783 (2004)

    Article  CAS  Google Scholar 

  26. Keele, B. F. et al. Identification and characterization of transmitted and early founder virus envelopes in primary HIV-1 infection. Proc. Natl Acad. Sci. USA 105, 7552–7557 (2008)

    Article  ADS  CAS  Google Scholar 

  27. Li, Z. et al. Transfer of IgG in the female genital tract by MHC class I-related neonatal Fc receptor (FcRn) confers protective immunity to vaginal infection. Proc. Natl Acad. Sci. USA 108, 4388–4393 (2011)

    Article  ADS  CAS  Google Scholar 

  28. Robbie, G. J. et al. A novel investigational Fc-modified humanized monoclonal antibody, motavizumab-YTE, has an extended half-life in healthy adults. Antimicrob. Agents Chemother. 57, 6147–6153 (2013)

    Article  CAS  Google Scholar 

  29. Pegu, A. et al. Neutralizing antibodies to HIV-1 envelope protect more effectively in vivo than those to the CD4 receptor. Sci. Transl. Med. 6, 243ra88 (2014)

    Article  Google Scholar 

  30. Edelman, G. M. et al. The covalent structure of an entire γG immunoglobulin molecule. Proc. Natl Acad. Sci. USA 63, 78–85 (1969)

    Article  ADS  CAS  Google Scholar 

  31. Gastinel, L. N., Simister, N. E. & Bjorkman, P. J. Expression and crystallization of a soluble and functional form of an Fc receptor related to class I histocompatibility molecules. Proc. Natl Acad. Sci. USA 89, 638–642 (1992)

    Article  ADS  CAS  Google Scholar 

  32. Li, M. et al. Human immunodeficiency virus type 1 env clones from acute and early subtype B infections for standardized assessments of vaccine-elicited neutralizing antibodies. J. Virol. 79, 10108–10125 (2005)

    Article  CAS  Google Scholar 

  33. Shu, Y. et al. Efficient protein boosting after plasmid DNA or recombinant adenovirus immunization with HIV-1 vaccine constructs. Vaccine 25, 1398–1408 (2007)

    Article  CAS  Google Scholar 

  34. Seaman, M. S. et al. Tiered categorization of a diverse panel of HIV-1 Env pseudoviruses for assessment of neutralizing antibodies. J. Virol. 84, 1439–1452 (2010)

    Article  CAS  Google Scholar 

  35. Claypool, S. M., Dickinson, B. L., Yoshida, M., Lencer, W. I. & Blumberg, R. S. Functional reconstitution of human FcRn in Madin–Darby canine kidney cells requires co-expressed human β2-microglobulin. J. Biol. Chem. 277, 28038–28050 (2002)

    Article  CAS  Google Scholar 

  36. Zeng, M. et al. Cumulative mechanisms of lymphoid tissue fibrosis and T cell depletion in HIV-1 and SIV infections. J. Clin. Invest. 121, 998–1008 (2011)

    Article  CAS  Google Scholar 

  37. Baker, K. et al. Neonatal Fc receptor expression in dendritic cells mediates protective immunity against colorectal cancer. Immunity 39, 1095–1107 (2013)

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We thank M. Roederer for advice on the design and statistical analysis of animal studies. This research was supported by the Intramural Research Program of the Vaccine Research Center, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), and in part by a grant from the Foundation for the National Institutes of Health with support from the Collaboration for AIDS Vaccine Discovery (CAVD), award OPP1039775, from the Bill & Melinda Gates Foundation. R.S.B. is supported by the NIH (DK044319, DK051362, DK053056 and DK088199) and the Harvard Digestive Diseases Center (DK0034854). T.R. is supported by the German research foundation (DFG; RA 2040/1-1). The findings and conclusions in this report are those of the authors and do not necessarily reflect the views of the funding agencies.

Author information

Authors and Affiliations

Authors

Contributions

S.-Y.K., Z.-Y.Y., J.R.M. and G.J.N. designed the study, analysed the data and prepared the manuscript. A.P. analysed the data, set up the ADCC assay and provided the material for the ADCC assays. R.S.R. and K.O.S. helped to prepare the manuscript. M.G.J. performed the surface plasmon resonance analysis. X.C. performed DNA cloning and protein purification. T.D.K., T.R. and R.S.B. performed the transcytosis assay. S.B., M.Z. and A.T.H. performed immunohistochemical staining. S.D.S. and J.R.M. performed the neutralization assays. K.W., J.-P.T. and S.S.R. performed the pharmacokinetics and challenge study. S.R.P. analysed the pharmacokinetic data. M.C.N. conducted statistical analyses.

Corresponding authors

Correspondence to John R. Mascola or Gary J. Nabel.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Extended data figures and tables

Extended Data Figure 1 Enhanced FcRn-binding mutants of VRC01 are released from human FcRn at pH 7.4.

VRC01 or the indicated FcRn-binding mutants were injected in PBS (pH 6.0) at a concentration of 100 nM over human-FcRn-immobilized Biacore CM5 sensor chips (500 response units (RU)) and were dissociated using PBS at pH 6.0 followed by PBS at pH 7.4.

Extended Data Figure 2 Mutants with enhanced human FcRn binding persist at higher serum concentrations than VRC01 in the human-FcRn(276)-transgenic mouse.

The indicated VRC01-derived monoclonal antibodies (2 mg kg−1) were injected intravenously into 6–8-week-old human-FcRn(276)-transgenic mice (n = 4, male and female mice evenly distributed). Serum concentrations were assessed by indirect ELISA against RSC3 over time. The percentage of the monoclonal antibodies remaining in the serum is shown compared with the percentage on day 1 (set at 100%).

Extended Data Figure 3 Binding of VRC01, VRC01-LS and other FcRn-binding mutants to FcγRIIa and FcγRIIb.

Binding to the indicated human Fc receptors was evaluated by ELISA as described in Methods.

Extended Data Figure 4 Serum pharmacokinetics, rectal tissue accumulation and mucosal secretion of VRC01-derived monoclonal antibodies in cynomolgus macaques.

VRC01 or VRC01-LS (10 mg kg−1) were injected intravenously into cynomolgus macaques. a, The serum levels (n = 4) were analysed over time. b, c, The amounts of monoclonal antibody per mg of total tissue protein in rectal biopsy samples (b) and per mg of total IgG in rectal secretions (c) from each monoclonal-antibody-injected cynomolgus macaque (n = 2 per group) were quantitated and are shown over time. The values for each macaque and the mean values for the groups are shown as dotted lines and heavier solid lines, respectively (b). Pharmacokinetic parameters were calculated with a two-compartment model (VRC01 versus VRC01-LS, half-life, 9.0 versus 30.3 days; clearance, 15.7 versus 3.7 ml day−1 kg−1; area under the curve (AUC), 896 versus 2,812 day × µg ml−1).

Extended Data Figure 5 Viral load measurements over time after passive antibody transfer and SHIV challenge of non-human primates.

The indicated monoclonal antibodies (0.3 mg kg−1) were administered intravenously to rhesus macaques (n = 12). Macaques were challenged with SHIV BaLP4 5 days later, and the plasma viral loads were measured over time. Normal human IgG was given to the control group. Five of 12 VRC01-LS-injected macaques and 10 of 12 VRC01-injected macaques were infected (P = 0.0447, one-tailed Fisher’s exact test).

Extended Data Figure 6 Accumulation and localization of VRC01 in vaginal tissue.

a, VRC01 or VRC01-LS (10 mg kg−1) were injected intravenously into female cynomolgus macaques (n = 2). The monoclonal antibody concentration per mg of total tissue protein in vaginal biopsy samples was quantitated at the indicated times. The values for each macaque and the mean values for the groups are shown as dotted and solid lines, respectively. b, VRC01 (20 mg kg−1) was injected intravenously into rhesus macaques (n = 2), and vaginal biopsy samples were taken and processed. Immunohistochemical staining was performed before and after the antibody dosing. The arrows indicate VRC01 (RSC3 staining, red) in basal and parabasal epithelial cells. The sections are representative of the sections assessed for two macaques. Scale bar, 50 µm.

Extended Data Figure 7 Similar antibody responses to VRC01 and VRC01-LS in non-human primates.

VRC01 or VRC01-LS (10 mg kg−1) was injected intravenously into Indian rhesus macaques (n = 4). a, Anti-VRC01 antibody responses (left) or anti-VRC01-LS antibody responses (right) were evaluated by ELISA. b, Sera from animals that were administered VRC01 or VRC01-LS were absorbed with 20 µg ml−1 VRC01, and binding to VRC01-LS was then evaluated by ELISA.

Extended Data Table 1 Comparative potency of HIV-1 neutralization by VRC01 and enhanced FcRn-binding mutants
Extended Data Table 2 EC50 values for the binding of VRC01 and VRC01-LS to human FcRn, as determined by ELISA
Extended Data Table 3 Relative abundance of N297 glycosylation in VRC01 and VRC01-LS*

PowerPoint slides

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Ko, SY., Pegu, A., Rudicell, R. et al. Enhanced neonatal Fc receptor function improves protection against primate SHIV infection. Nature 514, 642–645 (2014). https://doi.org/10.1038/nature13612

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nature13612

This article is cited by

Comments

By submitting a comment you agree to abide by our Terms and Community Guidelines. If you find something abusive or that does not comply with our terms or guidelines please flag it as inappropriate.

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