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.

Clinical targeting of HIV capsid protein with a long-acting small molecule

Abstract

Oral antiretroviral agents provide life-saving treatments for millions of people living with HIV, and can prevent new infections via pre-exposure prophylaxis1,2,3,4,5. However, some people living with HIV who are heavily treatment-experienced have limited or no treatment options, owing to multidrug resistance6. In addition, suboptimal adherence to oral daily regimens can negatively affect the outcome of treatment—which contributes to virologic failure, resistance generation and viral transmission—as well as of pre-exposure prophylaxis, leading to new infections1,2,4,7,8,9. Long-acting agents from new antiretroviral classes can provide much-needed treatment options for people living with HIV who are heavily treatment-experienced, and additionally can improve adherence10. Here we describe GS-6207, a small molecule that disrupts the functions of HIV capsid protein and is amenable to long-acting therapy owing to its high potency, low in vivo systemic clearance and slow release kinetics from the subcutaneous injection site. Drawing on X-ray crystallographic information, we designed GS-6207 to bind tightly at a conserved interface between capsid protein monomers, where it interferes with capsid-protein-mediated interactions between proteins that are essential for multiple phases of the viral replication cycle. GS-6207 exhibits antiviral activity at picomolar concentrations against all subtypes of HIV-1 that we tested, and shows high synergy and no cross-resistance with approved antiretroviral drugs. In phase-1 clinical studies, monotherapy with a single subcutaneous dose of GS-6207 (450 mg) resulted in a mean log10-transformed reduction of plasma viral load of 2.2 after 9 days, and showed sustained plasma exposure at antivirally active concentrations for more than 6 months. These results provide clinical validation for therapies that target the functions of HIV capsid protein, and demonstrate the potential of GS-6207 as a long-acting agent to treat or prevent infection with HIV.

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

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

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

Fig. 1: GS-6207 is a potent CA-targeting inhibitor of HIV replication.
Fig. 2: GS-6207 inhibits multiple CA-dependent steps of HIV-1 replication.
Fig. 3: Resistance to GS-6207 maps to CA.
Fig. 4: Clinical testing of GS-6207 in humans.

Data availability

All data to understand and assess the conclusions of this research are available in the Article and Supplementary Information. Raw gel source data for Fig. 2f are available in Supplementary Fig. 1. Small-molecule X-ray crystallographic coordinates and structure factor files have been deposited in the Protein Data Bank (PDB) with accession number 6V2F. Study GS-US-200-4072 was registered with ClinicalTrials.gov, NCT03739866. The datasets generated during and/or analysed during the current study are available from the corresponding author on reasonable request.

References

  1. Grant, R. M. et al. Preexposure chemoprophylaxis for HIV prevention in men who have sex with men. N. Engl. J. Med. 363, 2587–2599 (2010).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  2. Baeten, J. M. et al. Antiretroviral prophylaxis for HIV prevention in heterosexual men and women. N. Engl. J. Med. 367, 399–410 (2012).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  3. Molina, J. M. et al. On-demand preexposure prophylaxis in men at high risk for HIV-1 infection. N. Engl. J. Med. 373, 2237–2246 (2015).

    Article  PubMed  CAS  Google Scholar 

  4. Thigpen, M. C. et al. Antiretroviral preexposure prophylaxis for heterosexual HIV transmission in Botswana. N. Engl. J. Med. 367, 423–434 (2012).

    Article  PubMed  CAS  Google Scholar 

  5. WHO. HIV/AIDS Fact Sheets, 15 November 2019. https://www.who.int/news-room/fact-sheets/detail/hiv-aids (WHO, 2019).

  6. Emu, B. et al. Phase 3 study of ibalizumab for multidrug-resistant HIV-1. N. Engl. J. Med. 379, 645–654 (2018).

    Article  PubMed  CAS  Google Scholar 

  7. Bangsberg, D. R. et al. Non-adherence to highly active antiretroviral therapy predicts progression to AIDS. AIDS 15, 1181–1183 (2001).

    Article  PubMed  CAS  Google Scholar 

  8. Marrazzo, J. M. et al. Tenofovir-based preexposure prophylaxis for HIV infection among African women. N. Engl. J. Med. 372, 509–518 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  9. Anderson, P. L. et al. Emtricitabine–tenofovir concentrations and pre-exposure prophylaxis efficacy in men who have sex with men. Sci. Transl. Med. 4, 151ra125 (2012).

    Article  PubMed  PubMed Central  Google Scholar 

  10. Gulick, R. M. & Flexner, C. Long-acting HIV drugs for treatment and prevention. Annu. Rev. Med. 70, 137–150 (2019).

    Article  PubMed  CAS  Google Scholar 

  11. Thenin-Houssier, S. & Valente, S. T. HIV-1 capsid inhibitors as antiretroviral agents. Curr. HIV Res. 14, 270–282 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  12. Carnes, S. K., Sheehan, J. H. & Aiken, C. Inhibitors of the HIV-1 capsid, a target of opportunity. Curr. Opin. HIV AIDS 13, 359–365 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  13. Scott, D. E., Bayly, A. R., Abell, C. & Skidmore, J. Small molecules, big targets: drug discovery faces the protein–protein interaction challenge. Nat. Rev. Drug Discov. 15, 533–550 (2016).

    Article  PubMed  CAS  Google Scholar 

  14. Freed, E. O. HIV-1 assembly, release and maturation. Nat. Rev. Microbiol. 13, 484–496 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  15. Ganser, B. K., Li, S., Klishko, V. Y., Finch, J. T. & Sundquist, W. I. Assembly and analysis of conical models for the HIV-1 core. Science 283, 80–83 (1999).

    Article  ADS  PubMed  CAS  Google Scholar 

  16. Yamashita, M. & Engelman, A. N. Capsid-dependent host factors in HIV-1 infection. Trends Microbiol. 25, 741–755 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  17. Huang, P. T. et al. FEZ1 is recruited to a conserved cofactor site on capsid to promote HIV-1 trafficking. Cell Rep. 28, 2373–2385 (2019).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  18. Fernandez, J. et al. Transportin-1 binds to the HIV-1 capsid via a nuclear localization signal and triggers uncoating. Nat. Microbiol. 4, 1840–1850 (2019).

    Article  PubMed  CAS  Google Scholar 

  19. Carlson, L. A. et al. Three-dimensional analysis of budding sites and released virus suggests a revised model for HIV-1 morphogenesis. Cell Host Microbe 4, 592–599 (2008).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  20. Briggs, J. A., Wilk, T., Welker, R., Kräusslich, H. G. & Fuller, S. D. Structural organization of authentic, mature HIV-1 virions and cores. EMBO J. 22, 1707–1715 (2003).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  21. Yant, S. R. et al. A highly potent long-acting small-molecule HIV-1 capsid inhibitor with efficacy in a humanized mouse model. Nat. Med. 25, 1377–1384 (2019).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  22. Graupe, M. et al. Therapeutic compounds. US patent 10,071,985 B2 (2018).

    Google Scholar 

  23. Matreyek, K. A., Yücel, S. S., Li, X. & Engelman, A. Nucleoporin NUP153 phenylalanine-glycine motifs engage a common binding pocket within the HIV-1 capsid protein to mediate lentiviral infectivity. PLoS Pathog. 9, e1003693 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  24. Price, A. J. et al. CPSF6 defines a conserved capsid interface that modulates HIV-1 replication. PLoS Pathog. 8, e1002896 (2012).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  25. Price, A. J. et al. Host cofactors and pharmacologic ligands share an essential interface in HIV-1 capsid that is lost upon disassembly. PLoS Pathog. 10, e1004459 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  26. Bhattacharya, A. et al. Structural basis of HIV-1 capsid recognition by PF74 and CPSF6. Proc. Natl Acad. Sci. USA 111, 18625–18630 (2014).

    Article  ADS  PubMed  CAS  Google Scholar 

  27. Lee, K. et al. Flexible use of nuclear import pathways by HIV-1. Cell Host Microbe 7, 221–233 (2010).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  28. Perrier, M. et al. Prevalence of gag mutations associated with in vitro resistance to capsid inhibitor GS-CA1 in HIV-1 antiretroviral-naive patients. J. Antimicrob. Chemother. 72, 2954–2955 (2017).

    Article  PubMed  CAS  Google Scholar 

  29. Li, G. et al. Functional conservation of HIV-1 Gag: implications for rational drug design. Retrovirology 10, 126 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  30. Yant, S. R. et al. In vitro resistance profile of GS-6207, a first-in-class picomolar HIV capsid inhibitor in clinical development as a novel long-acting antiretroviral agent. In 10th IAS Conference on HIV Science http://programme.ias2019.org/Abstract/Abstract/683 (IAS, 2019).

  31. Tsiang, M. et al. Antiviral activity of bictegravir (GS-9883), a novel potent HIV-1 integrase strand transfer inhibitor with an improved resistance profile. Antimicrob. Agents Chemother. 60, 7086–7097 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  32. Margot, N. A., Gibbs, C. S. & Miller, M. D. Phenotypic susceptibility to bevirimat in isolates from HIV-1-infected patients without prior exposure to bevirimat. Antimicrob. Agents Chemother. 54, 2345–2353 (2010).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  33. Nakabayashi, H., Taketa, K., Miyano, K., Yamane, T. & Sato, J. Growth of human hepatoma cells lines with differentiated functions in chemically defined medium. Cancer Res. 42, 3858–3863 (1982).

    PubMed  CAS  Google Scholar 

  34. Balakrishnan, M. et al. Non-catalytic site HIV-1 integrase inhibitors disrupt core maturation and induce a reverse transcription block in target cells. PLoS ONE 8, e74163 (2013).

    Article  ADS  PubMed  PubMed Central  CAS  Google Scholar 

  35. Prichard, M. N. & Shipman, C. Jr. Analysis of combinations of antiviral drugs and design of effective multidrug therapies. Antivir. Ther. 1, 9–20 (1996).

    PubMed  CAS  Google Scholar 

  36. Bam, R. A. et al. TLR7 agonist GS-9620 is a potent inhibitor of acute HIV-1 infection in human peripheral blood mononuclear cells. Antimicrob. Agents Chemother. 61, e01369-16 (2016).

    PubMed  PubMed Central  Google Scholar 

  37. Warren, T. K. et al. Therapeutic efficacy of the small molecule GS-5734 against Ebola virus in rhesus monkeys. Nature 531, 381–385 (2016).

    Article  ADS  PubMed  PubMed Central  CAS  Google Scholar 

  38. Hung, M. et al. Large-scale functional purification of recombinant HIV-1 capsid. PLoS ONE 8, e58035 (2013).

    Article  ADS  PubMed  PubMed Central  CAS  Google Scholar 

  39. Pornillos, O. et al. X-ray structures of the hexameric building block of the HIV capsid. Cell 137, 1282–1292 (2009).

    Article  PubMed  PubMed Central  Google Scholar 

  40. Pornillos, O., Ganser-Pornillos, B. K. & Yeager, M. Atomic-level modelling of the HIV capsid. Nature 469, 424–427 (2011).

    Article  ADS  PubMed  PubMed Central  CAS  Google Scholar 

  41. Kissinger, C. R., Gehlhaar, D. K. & Fogel, D. B. Rapid automated molecular replacement by evolutionary search. Acta Crystallogr. D 55, 484–491 (1999).

    Article  PubMed  CAS  Google Scholar 

  42. Adams, P. D. et al. PHENIX: a comprehensive Python-based system for macromolecular structure solution. Acta Crystallogr. D 66, 213–221 (2010).

    PubMed  PubMed Central  CAS  Google Scholar 

  43. Emsley, P., Lohkamp, B., Scott, W. G. & Cowtan, K. Features and development of Coot. Acta Crystallogr. D 66, 486–501 (2010).

    PubMed  PubMed Central  CAS  Google Scholar 

Download references

Acknowledgements

We thank D. Cowfer, K. Brendza, G. Czerwieniec, M. Tsiang, K. Wang, G. Lane, M. Kenney, M. Ceo, S. Kazerani, T. Lane, L. Meng, T. Rainey, A. Vandehey, A. Wagner, M. O’Keefe, J. Yoon, S. Neville, W. Lew, B. Ross, Q. Wang, J. Cha, M. Tran and K. Nguyen for their support and contributions, and all the people who participated in the phase-I clinical trials, including the study participants, their families, and the principal investigators and their staff.

Author information

Authors and Affiliations

Authors

Contributions

T.C. conceived the project. D.J., M.H. and N.N. conducted protein purifications, and X.L. and R.S. oversaw the analysis. S.R.Y., N.P., T.Z.C., D.K. and L.L designed, conducted and analysed small-molecule library screens. T.C., J.O.L., S.R.Y., R.L.H., W.C.T., M.S.R. and A.E.C. provided project leadership. J.O.L., W.C.T., S.D.S., C.-H.C., E.C., G.B., J.R.Z., J. Li, M.G., P.M., Q.L., Q.W., R.L.H., R.D.S., S.D.S., S.E.L. and S.B. were responsible for the design, synthesis, characterization and scaling-up of small molecules. A.N.-M. conducted and analysed in vitro CA assembly assays. S.R.Y. and A.N.-M. conducted and analysed virion morphology by electron microscopy. G.J.S., S.A. and H.Y. designed, conducted and analysed high-throughput antiviral measurements. G.J.S, S.A., A.M. and Y.X. conducted cell-based assays for cytotoxicity. G.J.S. conducted and analysed in vitro drug combination studies. A.M. and R.R.R. conducted and analysed in vitro antiviral testing against HIV mutants with resistance to existing agents. A.G.V. and R.L.A. conducted protein crystallization studies, J.R.S. collected and analysed X-ray crystallographic data, and T.C.A. prepared the refinement table. C.E.C. and E.Y.H. conducted structural modelling studies to guide small-molecule development. G.A.P., M.H.W., S.A.L., S.C. and L.L. conducted and analysed biosensor binding studies. S.R.Y. A.M., E.S. and L.K.T. conducted and analysed cell-based mechanism-of-action studies. A.L. conducted and analysed biochemical protease assays. A.M., D.H., R.A.B. and S.R.Y. conducted resistance selection assays and characterized emergent HIV-1 CA variants. J.Z., B.L. and J.M. designed and executed preclinical pharmacokinetics and metabolism studies, and summarized results. A.E.C. oversaw all anatomical pathology examinations and analyses of preclinical animal species. W.R., S. Sellers and A.C. designed and tested drug formulations. A.C. and S.A.W. oversaw GS-6207 chemistry, manufacture and control for clinical studies. M.S.R., R.H., R.B. and D.M.B. designed and supervised the clinical studies, and G.I.S., P.J.R., G.E.C., C.K.M. and E.S.D conducted them. R.B., J. Ling, Y.-P. L., N.M. and C.C. conducted and coordinated clinical sample and statistical analyses. W.I.S. provided project guidance during the early discovery phase, and S. Swaminathan and W.E.L. provided long-term project oversight. S.R.Y., M.S.R., J.O.L. and T.C. wrote the manuscript, with input from all authors.

Corresponding author

Correspondence to Stephen R. Yant.

Ethics declarations

Competing interests

All authors are current or previous employees of Gilead Sciences (except for G.I.S., P.J.R., G.E.C., C.K.M., W.I.S. and E.S.D.) and received salary and stock ownership as compensation for their employment. M.G., J.O.L., W.R., R.D.S., S.D.S., W.C.T. and J.R.Z. are inventors on granted US patent no. 10,071,985B2 covering GS-6207 composition of matter and methods of use. G.I.S. receives research support from Gilead Sciences, Janssen Pharmaceutica, GlaxoSmithKline, Abbvie and Cepheid, and is on the speaker’s bureau and advisory board for Janssen, ViiV Healthcare and Merck. G.E.C. receives grants (investigator research payments) from Gilead Sciences, ViiV Healthcare, Merck and Janssen Pharmaceutica. C.K.M. receives research support from Gilead Sciences, Merck, ViiV Healthcare and Janssen Pharmaceutica, is on the speaker’s bureau for Gilead Sciences, Merck and Insmed, and is on an advisory board for Gilead Sciences. E.S.D. receives research support from Gilead Sciences, Merck and ViiV Healthcare, and has served as a consultant for Gilead Sciences.

Additional information

Peer review information Nature thanks Daniel R. Kuritzkes, Kevan Shokat and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.

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

Extended data figures and tables

Extended Data Table 1 Cytotoxicity of GS-6207 in human cell lines and primary cells
Extended Data Table 2 In vitro combination studies with GS-6207
Extended Data Table 3 GS-6207 activity against HIV-1 isolates that are resistant to existing antiretroviral inhibitors
Extended Data Table 4 Data collection and refinement statistics (molecular replacement)
Extended Data Table 5 Biosensor surface-plasmon-resonance assays of GS-6207 binding to recombinant CA
Extended Data Table 6 Resistance profile of HIV-1 CA mutants identified in viruses selected by GS-6207
Extended Data Table 7 Baseline characteristics and clinical adverse events in healthy participants (that affected >1 participant overall) in study GS-US-200-4070
Extended Data Table 8 Baseline characteristics and clinical serious adverse events (that affected any participant) and adverse events (that affected >1 participant overall) in participants living with HIV in study GS-US-200-4072
Extended Data Table 9 Clinical pharmacokinetic parameters in healthy volunteers and participants living with HIV
Extended Data Table 10 Plasma HIV-1 RNA levels and genotype in participants living with HIV (GS-US-200-4072)

Supplementary information

Supplementary Information

This file contains Supplementary Figures 1-5 and the Chemistry Experimental details.

Reporting Summary

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Link, J.O., Rhee, M.S., Tse, W.C. et al. Clinical targeting of HIV capsid protein with a long-acting small molecule. Nature 584, 614–618 (2020). https://doi.org/10.1038/s41586-020-2443-1

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41586-020-2443-1

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: Translational Research

Sign up for the Nature Briefing: Translational Research newsletter — top stories in biotechnology, drug discovery and pharma.

Get what matters in translational research, free to your inbox weekly. Sign up for Nature Briefing: Translational Research