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:

Antibody 10-1074 suppresses viremia in HIV-1-infected individuals

Abstract

Monoclonal antibody 10-1074 targets the V3 glycan supersite on the HIV-1 envelope (Env) protein. It is among the most potent anti-HIV-1 neutralizing antibodies isolated so far. Here we report on its safety and activity in 33 individuals who received a single intravenous infusion of the antibody. 10-1074 was well tolerated and had a half-life of 24.0 d in participants without HIV-1 infection and 12.8 d in individuals with HIV-1 infection. Thirteen individuals with viremia received the highest dose of 30 mg/kg 10-1074. Eleven of these participants were 10-1074-sensitive and showed a rapid decline in viremia by a mean of 1.52 log10 copies/ml. Virologic analysis revealed the emergence of multiple independent 10-1074-resistant viruses in the first weeks after infusion. Emerging escape variants were generally resistant to the related V3-specific antibody PGT121, but remained sensitive to antibodies targeting nonoverlapping epitopes, such as the anti-CD4-binding-site antibodies 3BNC117 and VRC01. The results demonstrate the safety and activity of 10-1074 in humans and support the idea that antibodies targeting the V3 glycan supersite might be useful for the treatment and prevention of HIV-1 infection.

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

Access options

Buy this article

Purchase on Springer Link

Instant access to full article PDF

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

Figure 1: Study design and pharmacokinetics of 10-1074 in HIV-1-negative participants and individuals with HIV-1 infection.
Figure 2: Viral load dynamics after 10-1074 infusion in participants with HIV-1.
Figure 3: Viral evolution after 10-1074 infusion in participants with HIV-1 infection.
Figure 4: Temporal evolution of escape from 10-1074 over time in individuals 1HB1, 1HB3, 1HC1, 1HD1, 1HD6K and 1HD10K.
Figure 5: SMRT sequencing analysis.

Similar content being viewed by others

Accession codes

Primary accessions

NCBI Reference Sequence

References

  1. Klein, F. et al. Antibodies in HIV-1 vaccine development and therapy. Science 341, 1199–1204 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Hraber, P. et al. Prevalence of broadly neutralizing antibody responses during chronic HIV-1 infection. AIDS 28, 163–169 (2014).

    Article  CAS  PubMed  Google Scholar 

  3. West, A.P. Jr. et al. Structural insights on the role of antibodies in HIV-1 vaccine and therapy. Cell 156, 633–648 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Mikell, I. et al. Characteristics of the earliest cross-neutralizing antibody response to HIV-1. PLoS Pathog. 7, e1001251 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Burton, D.R. & Mascola, J.R. Antibody responses to envelope glycoproteins in HIV-1 infection. Nat. Immunol. 16, 571–576 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Haynes, B.F. et al. HIV-host interactions: implications for vaccine design. Cell Host Microbe 19, 292–303 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Moore, P.L., Williamson, C. & Morris, L. Virological features associated with the development of broadly neutralizing antibodies to HIV-1. Trends Microbiol. 23, 204–211 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Buchacher, A. et al. Generation of human monoclonal antibodies against HIV-1 proteins; electrofusion and Epstein-Barr virus transformation for peripheral blood lymphocyte immortalization. AIDS Res. Hum. Retroviruses 10, 359–369 (1994).

    Article  CAS  PubMed  Google Scholar 

  9. Muster, T. et al. A conserved neutralizing epitope on gp41 of human immunodeficiency virus type 1. J. Virol. 67, 6642–6647 (1993).

    CAS  PubMed  PubMed Central  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  CAS  PubMed  PubMed Central  Google Scholar 

  11. Mouquet, H. et al. Complex-type N-glycan recognition by potent broadly neutralizing HIV antibodies. Proc. Natl. Acad. Sci. USA 109, E3268–E3277 (2012).

    Article  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Sok, D. et al. Promiscuous glycan site recognition by antibodies to the high-mannose patch of gp120 broadens neutralization of HIV. Sci. Transl. Med. 6, 236ra63 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Garces, F. et al. Structural evolution of glycan recognition by a family of potent HIV antibodies. Cell 159, 69–79 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. 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  CAS  PubMed  PubMed Central  Google Scholar 

  16. Sok, D. et al. Recombinant HIV envelope trimer selects for quaternary-dependent antibodies targeting the trimer apex. Proc. Natl. Acad. Sci. USA 111, 17624–17629 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Scheid, J.F. et al. Sequence and structural convergence of broad and potent HIV antibodies that mimic CD4 binding. Science 333, 1633–1637 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Liao, H.X. et al. Co-evolution of a broadly neutralizing HIV-1 antibody and founder virus. Nature 496, 469–476 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Scharf, L. et al. Antibody 8ANC195 reveals a site of broad vulnerability on the HIV-1 envelope spike. Cell Rep. 7, 785–795 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Huang, J. et al. Broad and potent HIV-1 neutralization by a human antibody that binds the gp41-gp120 interface. Nature 515, 138–142 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Pietzsch, J. et al. A mouse model for HIV-1 entry. Proc. Natl. Acad. Sci. USA 109, 15859–15864 (2012).

    Article  PubMed  PubMed Central  Google Scholar 

  23. 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  PubMed  PubMed Central  Google Scholar 

  24. Gautam, R. et al. A single injection of anti-HIV-1 antibodies protects against repeated SHIV challenges. Nature 533, 105–109 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Shingai, M. et al. Passive transfer of modest titers of potent and broadly neutralizing anti-HIV monoclonal antibodies block SHIV infection in macaques. J. Exp. Med. 211, 2061–2074 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Klein, F. et al. HIV therapy by a combination of broadly neutralizing antibodies in humanized mice. Nature 492, 118–122 (2012).

    Article  CAS  PubMed  Google Scholar 

  27. Horwitz, J.A. et al. HIV-1 suppression and durable control by combining single broadly neutralizing antibodies and antiretroviral drugs in humanized mice. Proc. Natl. Acad. Sci. USA 110, 16538–16543 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  28. Barouch, D.H. et al. Therapeutic efficacy of potent neutralizing HIV-1-specific monoclonal antibodies in SHIV-infected rhesus monkeys. Nature 503, 224–228 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Shingai, M. et al. Antibody-mediated immunotherapy of macaques chronically infected with SHIV suppresses viraemia. Nature 503, 277–280 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Halper-Stromberg, A. et al. Broadly neutralizing antibodies and viral inducers decrease rebound from HIV-1 latent reservoirs in humanized mice. Cell 158, 989–999 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  32. Bournazos, S. et al. Broadly neutralizing anti-HIV-1 antibodies require Fc effector functions for in vivo activity. Cell 158, 1243–1253 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Lu, C.L. et al. Enhanced clearance of HIV-1-infected cells by broadly neutralizing antibodies against HIV-1 in vivo. Science 352, 1001–1004 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Caskey, M. et al. Viraemia suppressed in HIV-1-infected humans by broadly neutralizing antibody 3BNC117. Nature 522, 487–491 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Lynch, R.M. et al. Virologic effects of broadly neutralizing antibody VRC01 administration during chronic HIV-1 infection. Sci. Transl. Med. 7, 319ra206 (2015).

    Article  CAS  PubMed  Google Scholar 

  36. Ledgerwood, J.E. et al. Safety, pharmacokinetics and neutralization of the broadly neutralizing HIV-1 human monoclonal antibody VRC01 in healthy adults. Clin. Exp. Immunol. 182, 289–301 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Schoofs, T. et al. HIV-1 therapy with monoclonal antibody 3BNC117 elicits host immune responses against HIV-1. Science 352, 997–1001 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Scheid, J.F. et al. HIV-1 antibody 3BNC117 suppresses viral rebound in humans during treatment interruption. Nature 535, 556–560 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Yoon, H. et al. CATNAP: a tool to compile, analyze and tally neutralizing antibody panels. Nucleic Acids Res. 43, W213–W219 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. 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  PubMed  PubMed Central  Google Scholar 

  41. 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  PubMed  Google Scholar 

  42. Sarzotti-Kelsoe, M. et al. Optimization and validation of the TZM-bl assay for standardized assessments of neutralizing antibodies against HIV-1. J. Immunol. Methods 409, 131–146 (2014).

    Article  CAS  PubMed  Google Scholar 

  43. Keizer, R.J., Huitema, A.D., Schellens, J.H. & Beijnen, J.H. Clinical pharmacokinetics of therapeutic monoclonal antibodies. Clin. Pharmacokinet. 49, 493–507 (2010).

    Article  CAS  PubMed  Google Scholar 

  44. Moir, S. & Fauci, A.S. B cells in HIV infection and disease. Nat. Rev. Immunol. 9, 235–245 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Gristick, H.B. et al. Natively glycosylated HIV-1 Env structure reveals new mode for antibody recognition of the CD4-binding site. Nat. Struct. Mol. Biol. 23, 906–915 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Abram, M.E., Ferris, A.L., Shao, W., Alvord, W.G. & Hughes, S.H. Nature, position, and frequency of mutations made in a single cycle of HIV-1 replication. J. Virol. 84, 9864–9878 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Coffin, J.M. HIV population dynamics in vivo: implications for genetic variation, pathogenesis, and therapy. Science 267, 483–489 (1995).

    Article  CAS  PubMed  Google Scholar 

  48. Maldarelli, F. et al. HIV populations are large and accumulate high genetic diversity in a nonlinear fashion. J. Virol. 87, 10313–10323 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Zhou, S., Jones, C., Mieczkowski, P. & Swanstrom, R. Primer ID validates template sampling depth and greatly reduces the error rate of next-generation sequencing of HIV-1 genomic RNA populations. J. Virol. 89, 8540–8555 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Bhiman, J.N. et al. Viral variants that initiate and drive maturation of V1V2-directed HIV-1 broadly neutralizing antibodies. Nat. Med. 21, 1332–1336 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Laird Smith, M. et al. Rapid Sequencing of Complete env Genes from Primary HIV-1 Samples. in Virus Evolution (Oxford University Press, 2016).

  52. Bar, K.J. et al. Effect of HIV Antibody VRC01 on Viral Rebound after Treatment Interruption. N. Engl. J. Med. 375, 2037–2050 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Lynch, R.M. et al. HIV-1 fitness cost associated with escape from the VRC01 class of CD4 binding site neutralizing antibodies. J. Virol. 89, 4201–4213 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. West, A.P. Jr. et al. Computational analysis of anti-HIV-1 antibody neutralization panel data to identify potential functional epitope residues. Proc. Natl. Acad. Sci. USA 110, 10598–10603 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  55. Trkola, A. et al. Delay of HIV-1 rebound after cessation of antiretroviral therapy through passive transfer of human neutralizing antibodies. Nat. Med. 11, 615–622 (2005).

    Article  CAS  PubMed  Google Scholar 

  56. Simek, M.D. et al. Human immunodeficiency virus type 1 elite neutralizers: individuals with broad and potent neutralizing activity identified by using a high-throughput neutralization assay together with an analytical selection algorithm. J. Virol. 83, 7337–7348 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Ratcliffe, S.J. & Shults, J. GEEQBOX: A MATLAB toolbox for generalized estimating equations and quasi-least squares. J. Stat. Softw. 25, 1–14 (2008).

    Article  Google Scholar 

  58. Salazar-Gonzalez, J.F. et al. Deciphering human immunodeficiency virus type 1 transmission and early envelope diversification by single-genome amplification and sequencing. J. Virol. 82, 3952–3970 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Larkin, M.A. et al. Clustal W and Clustal X version 2.0. Bioinformatics 23, 2947–2948 (2007).

    Article  CAS  PubMed  Google Scholar 

  60. Guindon, S. et al. New algorithms and methods to estimate maximum-likelihood phylogenies: assessing the performance of PhyML 3.0. Syst. Biol. 59, 307–321 (2010).

    Article  CAS  PubMed  Google Scholar 

  61. Darriba, D., Taboada, G.L., Doallo, R. & Posada, D. jModelTest 2: more models, new heuristics and parallel computing. Nat. Methods 9, 772 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Stamatakis, A. RAxML version 8: a tool for phylogenetic analysis and post-analysis of large phylogenies. Bioinformatics 30, 1312–1313 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Kirchherr, J.L. et al. High throughput functional analysis of HIV-1 env genes without cloning. J. Virol. Methods 143, 104–111 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. 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  PubMed  PubMed Central  Google Scholar 

  65. Katoh, K. & Standley, D.M. MAFFT multiple sequence alignment software version 7: improvements in performance and usability. Mol. Biol. Evol. 30, 772–780 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Price, M.N., Dehal, P.S. & Arkin, A.P. FastTree 2—approximately maximum-likelihood trees for large alignments. PLoS One 5, e9490 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank all study participants for devoting their time to support our research. We thank the Clinical Research Support Teams of the Rockefeller University Hospital and the Infectious Disease Division at the University Hospital Cologne, in particular, C. Golder, G. Kremer, S. Margane and E. Thomas. We thank L. Burke, S. Durant, M. Platten,I. Suárez and the nursing staff for patient care and recruitment, and all members of the laboratories of M.C.N. and F.K. for helpful discussions. We thank P. Fast and H. Park for clinical monitoring, A. Louie, D. Jordan, C. Conrad and D. Adzic for regulatory support, C. Anthony and S. Zhou for help in establishing Primer-ID sequencing, C. Ruping, K. Jain, M. Ercanoglu, R. Patel and J. Dizon for sample processing, U. Kerkweg, R. Macarthur and A. Johnson for pharmacy services, A. Germann and H. von Briesen for HIV culture analyses, R. Kaiser for p24 measurements and D. Sok for providing PGT121 and PGDM1400 for neutralization assays. Amplification and library preparation for SMRT sequencing was performed with the support of the Translational Virology Core at the UC San Diego Center for AIDS Research (P30 AI036214). SMRT sequencing was conducted at the IGM Genomics Center, University of California, San Diego, La Jolla, California. Computational analysis of sequence data was performed, in part, on a cluster, which was supported by U01 GM110749 (NIH/NIGMS). This work was supported in part by the Bill and Melinda Gates Foundation Collaboration for AIDS Vaccine Discovery (CAVD) Grants OPP1032144 (M.S.S.), OPP1092074 and OPP1124068 (M.C.N.), National Institute of Allergy and Infectious Diseases of the National Institutes of Health Grant HIVRAD P01 AI100148 (P.J.B.), a BEAT-HIV Delaney grant UM1 AI126620 (B.H.H.), the Robertson Foundation to M.C.N., and the NIH Center for HIV/AIDS Vaccine Immunology and Immunogen Discovery (CHAVI-ID) 1UM1 AI100663-01 (M.C.N.). T.S. was supported by a German Research Foundation postdoctoral fellowship (SCHO 1612/1-1) and is currently supported in part by grant #UL1 TR001866 from the National Center for Advancing Translational Sciences (NCATS), National Institutes of Health (NIH) Clinical and Translational Science Award (CTSA) program. H.G. is supported by a fellowship from the German Center for Infection Research (DZIF). T. Karagounis is an HHMI Medical Research Fellow. E.F.K. is supported by a Ruth L. Kirschstein National Research Service Award (F30 AI112426). B.M. was supported by grant number R00 AI120851 from the National Institute of Allergy and Infectious Diseases. K.E. was supported by T15 LM007092 from the National Library of Medicine. F.K. is supported by the Heisenberg Program of the DFG (KL 2389/2-1), the European Research Council (ERC-StG639961) and the German Center for Infection Research (DZIF), partner site Bonn–Cologne, Cologne, Germany. M.C.N. is a Howard Hughes Medical Institute Investigator. Aspects of this work are encompassed by patent application PCT/US2013/065696.

Author information

Authors and Affiliations

Authors

Contributions

M.C. (principal investigator, US), M.C.N. and F.K. (principal investigator, Germany) designed the trial; M.C., T.S., H.G., M.C.N. and F.K. analyzed the data and wrote the manuscript; R.M.G., G.F. and S.J.S. contributed to study design and implementation. M.C., H.G., A.S., Y.Z.C., R.L., M.W.-P. and F.K. implemented the study. C.L., D.G., T. Kümmerle., C.W., S.K., B.S.Z. and G.F. contributed to participant recruitment and clinical assessments. I.S., C.U.-O. and D.W. coordinated sample processing. T.S., T. Karagounis and L.N., performed viral culture, SGS and Primer-ID sequencing work. T.Y.O. performed Primer-ID analyses and bioinformatics processing of SGS data. A.R. and M.S.S. performed TZM.bl neutralization assays. B.M., K.E. and C.I. carried out SMRT sequencing and analysis. E.F.K., G.H.L. and B.H.H. analyzed SGS data. N.P. performed statistical analysis. H.M., A.P.W. and P.J.B. contributed to data analysis. T. Keler was responsible for 10-1074 manufacture and provided regulatory guidance. All authors read and contributed to the writing of the manuscript.

Corresponding authors

Correspondence to Marina Caskey, Michel C Nussenzweig or Florian Klein.

Ethics declarations

Competing interests

T. Keler is employed by Celldex Therapeutics. 10-1074 was manufactured by Celldex Therapeutics.

Supplementary information

Supplementary Figures and Tables

Supplementary Figures 1–9 and Supplementary Tables 1–8 (PDF 6076 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Caskey, M., Schoofs, T., Gruell, H. et al. Antibody 10-1074 suppresses viremia in HIV-1-infected individuals. Nat Med 23, 185–191 (2017). https://doi.org/10.1038/nm.4268

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nm.4268

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