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 and TLR7 agonist delay viral rebound in SHIV-infected monkeys

A Publisher Correction to this article was published on 05 November 2018

This article has been updated

Abstract

The latent viral reservoir is the critical barrier for the development of a cure for HIV-1 infection. Previous studies have shown direct antiviral activity of potent HIV-1 Env-specific broadly neutralizing antibodies (bNAbs) administered when antiretroviral therapy (ART) was discontinued, but it remains unclear whether bNAbs can target the viral reservoir during ART. Here we show that administration of the V3 glycan-dependent bNAb PGT121 together with the Toll-like receptor 7 (TLR7) agonist vesatolimod (GS-9620) during ART delayed viral rebound following discontinuation of ART in simian–human immunodeficiency virus (SHIV)-SF162P3-infected rhesus monkeys in which ART was initiated during early acute infection. Moreover, in the subset of monkeys that were treated with both PGT121 and GS-9620 and that did not show viral rebound after discontinuation of ART, adoptive transfer studies and CD8-depletion studies also did not reveal virus. These data demonstrate the potential of bNAb administration together with innate immune stimulation as a possible strategy for targeting the viral reservoir.

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: Plasma viral loads after infection with SHIV-SF162P3 and before discontinuation of ART.
Fig. 2: Cellular immune activation following administration of GS-9620 and before discontinuation of ART.
Fig. 3: Viral DNA before discontinuation of ART.
Fig. 4: Viral loads following ART discontinuation.
Fig. 5: Analysis and correlations of viral rebound.
Fig. 6: Adoptive transfer and CD8 depletion studies.

Similar content being viewed by others

Data availability

All data generated and analysed in this study are available from the corresponding author upon reasonable request. Source data for figures from individual animals are available online.

Change history

  • 05 November 2018

    In Fig. 4b of this Article, the x-axis labels ‘PGT121’ and ‘GS-9620’ were inadvertently swapped in both graphs. In Fig. 5a, b, ‘TLR7’ should have been ‘GS-9620’. These figures have been corrected online.

References

  1. Finzi, D. et al. Identification of a reservoir for HIV-1 in patients on highly active antiretroviral therapy. Science 278, 1295–1300 (1997).

    Article  ADS  CAS  Google Scholar 

  2. Persaud, D., Zhou, Y., Siliciano, J. M. & Siliciano, R. F. Latency in human immunodeficiency virus type 1 infection: no easy answers. J. Virol. 77, 1659–1665 (2003).

    Article  CAS  Google Scholar 

  3. Chun, T. W. et al. Presence of an inducible HIV-1 latent reservoir during highly active antiretroviral therapy. Proc. Natl Acad. Sci. USA 94, 13193–13197 (1997).

    Article  ADS  CAS  Google Scholar 

  4. Ho, Y. C. et al. Replication-competent noninduced proviruses in the latent reservoir increase barrier to HIV-1 cure. Cell 155, 540–551 (2013).

    Article  CAS  Google Scholar 

  5. Finzi, D. et al. Latent infection of CD4+ T cells provides a mechanism for lifelong persistence of HIV-1, even in patients on effective combination therapy. Nat. Med. 5, 512–517 (1999).

    Article  CAS  Google Scholar 

  6. Chun, T. W., Davey, R. T., Jr, Engel, D., Lane, H. C. & Fauci, A. S. Re-emergence of HIV after stopping therapy. Nature 401, 874–875 (1999).

    Article  ADS  CAS  Google Scholar 

  7. Barouch, D. H. & Deeks, S. G. Immunologic strategies for HIV-1 remission and eradication. Science 345, 169–174 (2014).

    Article  ADS  CAS  Google Scholar 

  8. Deeks, S. G. et al. International AIDS Society global scientific strategy: towards an HIV cure 2016. Nat. Med. 22, 839–850 (2016).

    Article  CAS  Google Scholar 

  9. Shan, L. et al. Stimulation of HIV-1-specific cytolytic T lymphocytes facilitates elimination of latent viral reservoir after virus reactivation. Immunity 36, 491–501 (2012).

    Article  CAS  Google Scholar 

  10. 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  ADS  CAS  Google Scholar 

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

    Article  ADS  CAS  Google Scholar 

  12. Julg, B. et al. Virological control by the CD4-binding site antibody N6 in simian-human immunodeficiency virus-infected rhesus monkeys. J. Virol. 91, e00498-17 (2017).

    Article  Google Scholar 

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

    Article  ADS  CAS  Google Scholar 

  14. Caskey, M. et al. Antibody 10-1074 suppresses viremia in HIV-1-infected individuals. Nat. Med. 23, 185–191 (2017).

    Article  CAS  Google Scholar 

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

    Article  ADS  CAS  Google Scholar 

  16. 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  Google Scholar 

  17. 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 

  18. Liu, J. et al. Antibody-mediated protection against SHIV challenge includes systemic clearance of distal virus. Science 353, 1045–1049 (2016).

    Article  ADS  CAS  Google Scholar 

  19. Tsai, A. et al. Toll-like receptor 7 agonist GS-9620 induces HIV expression and HIV-specific immunity in cells from HIV-infected individuals on suppressive antiretroviral therapy. J. Virol. 91, e02166-16 (2017).

    Article  Google Scholar 

  20. Borducchi, E. N. et al. Ad26/MVA therapeutic vaccination with TLR7 stimulation in SIV-infected rhesus monkeys. Nature 540, 284–287 (2016).

    Article  ADS  CAS  Google Scholar 

  21. Barouch, D. H. et al. Protective efficacy of a global HIV-1 mosaic vaccine against heterologous SHIV challenges in rhesus monkeys. Cell 155, 531–539 (2013).

    Article  CAS  Google Scholar 

  22. Kawai, T. et al. Interferon-α induction through Toll-like receptors involves a direct interaction of IRF7 with MyD88 and TRAF6. Nat. Immunol. 5, 1061–1068 (2004).

    Article  CAS  Google Scholar 

  23. Hemmi, H. et al. Small anti-viral compounds activate immune cells via the TLR7 MyD88-dependent signaling pathway. Nat. Immunol. 3, 196–200 (2002).

    Article  CAS  Google Scholar 

  24. Whitney, J. B. et al. Rapid seeding of the viral reservoir prior to SIV viraemia in rhesus monkeys. Nature 512, 74–77 (2014).

    Article  ADS  CAS  Google Scholar 

  25. Henrich, T. J. et al. Antiretroviral-free HIV-1 remission and viral rebound after allogeneic stem cell transplantation: report of 2 cases. Ann. Intern. Med. 161, 319–327 (2014).

    Article  Google Scholar 

  26. Persaud, D. et al. Absence of detectable HIV-1 viremia after treatment cessation in an infant. N. Engl. J. Med. 369, 1828–1835 (2013).

    Article  CAS  Google Scholar 

  27. Nishimura, Y. et al. Early antibody therapy can induce long-lasting immunity to SHIV. Nature 543, 559–563 (2017).

    Article  ADS  CAS  Google Scholar 

Download references

Acknowledgements

We thank K. Reimann and A. Hill for advice, assistance, and reagents. We acknowledge support from the Bill & Melinda Gates Foundation (OPP1107669), the American Foundation for AIDS Research (109219-58-RGRL), the National Institutes of Health (AI096040, AI124377, AI126603, AI129797, AI128751, OD024917), and the Ragon Institute of MGH, MIT, and Harvard.

Author information

Authors and Affiliations

Authors

Contributions

D.H.B. and R.G. designed the study. J.H. and R.G. developed the ART formulation and TLR7 agonist. E.B. conducted the cytokine analyses. E.N.B., J.L., J.P.N., A.M.C., P.A., N.B.M., A.C., D.J., L.P., K.M., and E.T.M. performed the immunologic and virologic assays. W.-H.Y., S.F., T.B., and G.A. led the computational modelling. W.L. led the statistical analysis. M.G.L. led the clinical care of the rhesus monkeys. D.H.B. led the study and wrote the paper with all co-authors.

Corresponding author

Correspondence to Dan H. Barouch.

Ethics declarations

Competing interests

E.B., J.H., and R.G. are employees of Gilead Sciences. The other 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.

Extended data figures and tables

Extended Data Fig. 1 Study design.

Forty-four rhesus monkeys (n = 11 monkeys per group) were infected with SHIV-SF162P3 at week 0 and ART was initiated at week 1 (day 7). GS-9620 administrations and PGT121 infusions are shown from weeks 96 to 114. ART was discontinued at week 130.

Extended Data Fig. 2 Immune activation following GS-9620 administration and before ART discontinuation.

a, Activation of CD4+ T cells was assessed by CD38 expression on days 0 and 1 following GS-9620 administration, supplementing the data shown in Fig. 2a (n = 11 monkeys per group). Representative data are shown following the fifth GS-9620 dose, which was comparable to the other doses. Red horizontal bars indicate median values. P values reflect two-sided Mann–Whitney tests. b, Plasma levels of IFNα, IL-1RA, I-TAC, eotaxin, MIG, MCP-1, IL-1β, IL-6, IP-10 are shown on day 1 following GS-9620 administration (n = 11 monkeys per group). Red bars represent mean values. Combined data from all GS-9620 administrations with pre-dose levels subtracted are shown.

Extended Data Fig. 3 Anti-drug antibody (ADA) assay before ART discontinuation.

ADA responses were assessed in the PGT121+GS-9620 and PGT121-alone groups every 2 weeks from weeks 106 to 124 using an electrochemoluminescence (ECL) assay with an anti-PGT121 idiotypic mAb (n = 11 monkeys per group). No ADA was detected. One monkey in the PGT121+GS-9620 group had background reactivity in this assay at week 106 before PGT121 exposure (green bars).

Extended Data Fig. 4 PGT121 pharmacokinetics in serum and tissues before ART discontinuation.

a, Peak serum PGT121 levels are shown (limit of detection 0.5 μg ml−1) 1 h following each of five infusions of PGT121 (weeks 106–114) and during the washout period (weeks 114–130) (n = 11 monkeys per group). b, PGT121 levels (limit of detection 0.5 μg ml−1) were assessed in cell lysates (L) and initial wash supernatants (S) from 106 lymph node (LN) and colorectal (CR) cells from week 120 (n = 11 monkeys per group). Positive controls (Ctrl) included lymph node samples from naive monkeys spiked with PGT121. Red bars represent median values.

Extended Data Fig. 5 IFNγ ELISPOT responses before ART discontinuation.

Gag-, Env-, and Pol-specific IFNγ ELISPOT responses in PBMCs are shown at week 4 (a), week 96 (b), and week 120 (c) (n = 11 monkeys per group). Spot-forming cells (SFCs) per million PBMCs are shown. Red horizontal bars indicate median values. The dotted line represents the assay limit of quantitation (55 SFCs per million PBMCs).

Extended Data Fig. 6 IFNγ intracellular cytokine staining (ICS) responses before ART discontinuation.

Gag-, Env-, and Pol-specific IFNγ ICS responses in PBMCs (a) and in LNMCs (b) are shown at week 120 (n = 11 monkeys per group). Per cent IFNγ-producing CD8+CD3+ T cells in PBMCs and per cent IFNγ-producing total CD8+CD3+ T cells (CD8) and follicular CXCR5+CD8+CD3+ T cells (fCD8) in LNMCs are shown. Red horizontal bars indicate median values. The dotted line represents the assay limit of quantitation (0.05% CD8+CD3+ T cells).

Extended Data Fig. 7 IFNγ ELISPOT responses following ART discontinuation and trends for viral rebound.

a, Gag-, Env-, and Pol-specific IFNγ ELISPOT responses in PBMCs are shown at day 140 following ART discontinuation (n = 11 monkeys per group). SFCs per million PBMCs are shown. Monkeys in each group that demonstrated viral rebound versus no rebound are shown separately. Red horizontal bars indicate median values. The dotted line represents the assay limit of quantitation (55 SFCs per million PBMCs). b, Trends for viral rebound are shown in the GS-9620 and PGT121 groups in relation to pre-ART week 1 viral loads, supplementing the data shown in Fig. 5c. Red horizontal bars indicate median values.

Extended Data Fig. 8 CD8 depletion efficiency.

CD8+ T cells per μl peripheral blood are shown in PGT121+GS-9620-treated monkeys before and after CD8 depletion in monkeys that exhibited viral rebound and post-rebound virologic control (n = 5, left, red lines) and in monkeys that exhibited no viral rebound following ART discontinuation (n = 5, right, black lines).

Extended Data Fig. 9 Computational model.

a, LASSO and PLSR model identifies the parameters that correlate with delayed viral rebound (n = 11 monkeys per group). Left, individual monkeys are shown distributed by latent variables 1 and 3 of the model. Timing of viral rebound is indicated by the colour gradient. R2 = 0.176, root mean square error (RMSE) = 0.917, P < 0.001 in two-sided permutation tests. Middle, the contribution of the selected features to model separation is displayed in variable importance in projection (VIP) scores, related to early (blue) or late (red) viral rebound. Right, correlation between viral rebound and latent variable 1. P value reflects a two-sided Spearman rank-correlation test. b, LASSO and PLSR model identifies the parameters that correlate with reduced total viral loads (n = 11 monkeys per group). Left, individual monkeys are showed distributed by latent variables 1 and 2. Total viral loads are indicated by the colour gradient. R2 = 0.282, root mean square error (RMSE) = 0.857, P < 0.001 in in two-sided permutation tests. Middle, the contribution of the selected features to model separation is displayed in VIP scores, related to high (blue) or low (red) total viral loads. Right, correlation between total viral loads and latent variable 1. P value reflects a two-sided Spearman rank-correlation test.

Extended Data Fig. 10 In vitro killing studies.

a, GS-9620 treatment led to CD69 upregulation of CD56+ NK cells and CD3+ T cells in vitro following incubation of human PBMCs with 1,000 nM GS-9620 for 5 days (n = 9). b, GS-9620 treatment augmented PGT121-mediated killing of PGT121 in vitro. Per cent p24 reduction in CD4+ T cells (n = 6) using an antibody-mediated killing assay (see Methods). Per cent killing was calculated as the per cent reduction in p24 in CD4+ T cells with PGT121 compared with no PGT121. P values reflect two-sided paired Student’s t-tests.

Supplementary information

Source data

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Borducchi, E.N., Liu, J., Nkolola, J.P. et al. Antibody and TLR7 agonist delay viral rebound in SHIV-infected monkeys. Nature 563, 360–364 (2018). https://doi.org/10.1038/s41586-018-0600-6

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41586-018-0600-6

Keywords

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