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.

Ad26/MVA therapeutic vaccination with TLR7 stimulation in SIV-infected rhesus monkeys

Abstract

The development of immunologic interventions that can target the viral reservoir in HIV-1-infected individuals is a major goal of HIV-1 research1,2. However, little evidence exists that the viral reservoir can be sufficiently targeted to improve virologic control following discontinuation of antiretroviral therapy. Here we show that therapeutic vaccination with Ad26/MVA (recombinant adenovirus serotype 26 (Ad26) prime, modified vaccinia Ankara (MVA) boost)3,4 and stimulation of TLR7 (Toll-like receptor 7) improves virologic control and delays viral rebound following discontinuation of antiretroviral therapy in SIV-infected rhesus monkeys that began antiretroviral therapy during acute infection. Therapeutic vaccination with Ad26/MVA resulted in a marked increase in the magnitude and breadth of SIV-specific cellular immune responses in virologically suppressed, SIV-infected monkeys. TLR7 agonist administration led to innate immune stimulation and cellular immune activation. The combination of Ad26/MVA vaccination and TLR7 stimulation resulted in decreased levels of viral DNA in lymph nodes and peripheral blood, and improved virologic control and delayed viral rebound following discontinuation of antiretroviral therapy. The breadth of cellular immune responses correlated inversely with set point viral loads and correlated directly with time to viral rebound. These data demonstrate the potential of therapeutic vaccination combined with innate immune stimulation as a strategy aimed at a functional cure for HIV-1 infection.

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

Figure 1: SIV RNA and vaccine immunogenicity before ART discontinuation.
Figure 2: SIV DNA before ART discontinuation.
Figure 3: SIV RNA following ART discontinuation.
Figure 4: Correlations of cellular immune breadth with set point viral loads and time to viral rebound.

References

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

    Article  CAS  ADS  Google Scholar 

  2. 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 

  3. 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 

  4. Barouch, D. H. et al. Vaccine protection against acquisition of neutralization-resistant SIV challenges in rhesus monkeys. Nature 482, 89–93 (2012)

    Article  CAS  ADS  Google Scholar 

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

    Article  CAS  ADS  Google Scholar 

  6. 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 

  7. 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  CAS  ADS  Google Scholar 

  8. 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 

  9. 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 

  10. 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  CAS  ADS  Google Scholar 

  11. 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 

  12. Deng, K. et al. Broad CTL response is required to clear latent HIV-1 due to dominance of escape mutations. Nature 517, 381–385 (2015)

    Article  CAS  ADS  Google Scholar 

  13. Liu, J. et al. Immune control of an SIV challenge by a T-cell-based vaccine in rhesus monkeys. Nature 457, 87–91 (2009)

    Article  CAS  ADS  Google Scholar 

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

    Article  CAS  ADS  Google Scholar 

  15. Abbink, P. et al. Comparative seroprevalence and immunogenicity of six rare serotype recombinant adenovirus vaccine vectors from subgroups B and D. J. Virol. 81, 4654–4663 (2007)

    Article  CAS  Google Scholar 

  16. 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 

  17. 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 

  18. 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 

  19. 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 

  20. Banga, R. et al. PD-1+ and follicular helper T cells are responsible for persistent HIV-1 transcription in treated aviremic individuals. Nat. Med. 22, 754–761 (2016)

    Article  CAS  Google Scholar 

  21. Hel, Z. et al. Viremia control following antiretroviral treatment and therapeutic immunization during primary SIV251 infection of macaques. Nat. Med. 6, 1140–1146 (2000)

    Article  CAS  Google Scholar 

  22. Rosenberg, E. S. et al. Safety and immunogenicity of therapeutic DNA vaccination in individuals treated with antiretroviral therapy during acute/early HIV-1 infection. PLoS One 5, e10555 (2010)

    Article  ADS  Google Scholar 

  23. Li, J. Z. et al. Factors associated with viral rebound in HIV-1-infected individuals enrolled in a therapeutic HIV-1 gag vaccine trial. J. Infect. Dis. 203, 976–983 (2011)

    Article  Google Scholar 

  24. Liu, J. et al. Low-dose mucosal simian immunodeficiency virus infection restricts early replication kinetics and transmitted virus variants in rhesus monkeys. J. Virol. 84, 10406–10412 (2010)

    Article  CAS  Google Scholar 

  25. Burg, D., Rong, L., Neumann, A. U. & Dahari, H. Mathematical modeling of viral kinetics under immune control during primary HIV-1 infection. J. Theor. Biol. 259, 751–759 (2009)

    Article  MathSciNet  CAS  Google Scholar 

  26. Wodarz, D. et al. A new theory of cytotoxic T-lymphocyte memory: implications for HIV treatment. Philos. Trans. R. Soc. B Biol. Sci. 355, 329–343 (2000)

    Article  CAS  Google Scholar 

  27. Luzyanina, T., Engelborghs, K., Ehl, S., Klenerman, P. & Bocharov, G. Low level viral persistence after infection with LCMV: a quantitative insight through numerical bifurcation analysis. Math. Biosci. 173, 1–23 (2001)

    Article  MathSciNet  CAS  Google Scholar 

  28. Althaus, C. L. & De Boer, R. J. Dynamics of immune escape during HIV/SIV infection. PLOS Comput. Biol. 4, e1000103 (2008)

    Article  MathSciNet  ADS  Google Scholar 

  29. Elemans, M. et al. Why don’t CD8+ T cells reduce the lifespan of SIV-infected cells in vivo? PLOS Comput. Biol. 7, e1002200 (2011)

    Article  CAS  Google Scholar 

  30. Gadhamsetty, S., Coorens, T. & Boer, R. J. de. Notwithstanding circumstantial alibis, cytotoxic T cells can be major killers of HIV-1 infected cells. J. Virol. 306–316 (2016)

  31. Chen, H. Y., Di Mascio, M., Perelson, A. S., Ho, D. D. & Zhang, L. Determination of virus burst size in vivo using a single-cycle SIV in rhesus macaques. Proc. Natl Acad. Sci. USA 104, 19079–19084 (2007)

    Article  CAS  ADS  Google Scholar 

  32. De Boer, R. J., Ribeiro, R. M. & Perelson, A. S. Current estimates for HIV-1 production imply rapid viral clearance in lymphoid tissues. PLOS Comput. Biol. 6, e1000906 (2010)

    Article  MathSciNet  ADS  Google Scholar 

  33. Ramratnam, B. et al. Rapid production and clearance of HIV-1 and hepatitis C virus assessed by large volume plasma apheresis. Lancet 354, 1782–1785 (1999)

    Article  CAS  Google Scholar 

  34. Chivers, C. MHadaptive: General Markov Chain Monte Carlo for Bayesian Inference using adaptive Metropolis-Hastings sampling. https://cran.r-project.org/web/packages/MHadaptive/MHadaptive.pdf (2012)

  35. Soetaert, K., Petzoldt, T. & Setzer, R. W. deSolve: Solvers for Initial Value Problems of Differential Equations (ODE, DAE, DDE). https://cran.r-project.org/web/packages/deSolve/vignettes/deSolve.pdf (2016)

Download references

Acknowledgements

We thank C. Linde, T. Broge, T. Barnes, D. van Manen, F. Wegmann, C. Shaver, W. Wagner, M. Boyd, R. Nityanandam, K. Smith, S. Blackmore, L. Parenteau, P. Giglio, M. Shetty, S. Levin, J. Shields, G. Neubauer, and F. Stephens for generous advice, assistance, and reagents. We acknowledge support from the US Army Medical Research and Materiel Command and the Military HIV Research Program, Walter Reed Army Institute of Research through its cooperative agreement with the Henry M. Jackson Foundation (W81XWH-11-2-0174); the National Institutes of Health (AI096040, AI124377, AI126603, OD019851); the Ragon Institute of MGH, MIT, and Harvard. Mathematical model fitting was performed on the Orchestra High Performance Compute Cluster at Harvard Medical School. The views expressed in this manuscript are those of the authors and do not represent the official views of the Department of the Army or the Department of Defense.

Author information

Authors and Affiliations

Authors

Contributions

D.H.B, N.L.M., J.H.K., M.L.R., M.G.P., H.S., and R.G. designed the studies. J.H. and R.G. developed the ART formulation and TLR7 agonist. E.N.B., C.C., K.E.S., J.L., J.P.N., A.L.B., L.P., B.C.L., J.J., D.J., J.M., S.M., A.C., K.M., and G.A. performed the immunologic assays. P.A. and D.N. conducted the virologic assays. J.M.G. and A.L.H. performed the viral dynamics modelling. M.G.L. led the clinical care of the rhesus monkeys. D.H.B. wrote the paper with all co-authors.

Corresponding author

Correspondence to Dan H. Barouch.

Ethics declarations

Competing interests

M.G.P. and H.S. are employees of Janssen Infectious Diseases and Vaccines. J.H. and R.G. are employees of Gilead Sciences.

Additional information

Reviewer Information

Nature thanks S. Lewin and the other anonymous reviewer(s) for their contribution to the peer review of this work.

Extended data figures and tables

Extended Data Figure 1 Activation of CD8+ T cells following GS-986 administration.

Representative data on days 0, 1, and 2 following GS-986 administration. Activation was assessed by CD69 expression on CD3+CD8+ T cells. Red bars represent mean values for each group.

Extended Data Figure 2 Activation of CD4+ T cells following GS-986 administration.

Representative data on days 0, 1, and 2 following GS-986 administration. Activation was assessed by CD69 expression on CD3+CD4+ T cells.

Extended Data Figure 3 Innate immune stimulation following GS-986 administration.

Plasma IFN-α (pg ml−1) levels are shown on day 1 following GS-986 administration. Red bars represent mean values for each group. Data points reflect all animals following all GS-986 administrations combined with pre-dose levels subtracted.

Extended Data Figure 4 CD8+ T cells following Ad26/MVA vaccination.

SIVmac239 Gag-, Pol- and Env-specific, IFN-γ+CD3+CD8+ central memory T cells were assessed by multiparameter intracellular cytokine staining assays.

Extended Data Figure 5 CD4+ T cells following Ad26/MVA vaccination.

SIVmac239 Gag-, Pol- and Env-specific, IFN-γ+CD3+CD4+ central memory T cells were assessed by multiparameter intracellular cytokine staining assays.

Extended Data Figure 6 Cellular immune breadth.

Responses to subpools of 10 peptides spanning SIVmac239 Gag, Pol, and Env are shown before vaccination (week 20, blue), after Ad26 prime (week 29, red), and after MVA boost (week 50, green). Coloured squares indicate positive responses. ‘x’ indicates missing data as a result of insufficient PBMCs.

Extended Data Figure 7 Correlations of cellular immune breadth to day 7 SIV RNA.

Correlations are shown for the breadth of Gag, Pol, Env and total (Gag+Pol+Env) cellular immune responses at the time of ART discontinuation at week 72 and pre-ART day 7 log SIV RNA. P value indicates two-sided Spearman rank-correlation test. R value indicates correlation coefficient.

Extended Data Figure 8 Correlations of cellular immune breadth to set point viral loads.

Correlations are shown for the breadth of Gag, Pol, Env and total (Gag+Pol+Env) cellular immune responses at peak immunity at week 50 and set point log SIV RNA following ART discontinuation. P values indicate two-sided Spearman rank-correlation tests. R values indicate correlation coefficients.

Extended Data Figure 9 Correlations of cellular immune breadth to time to viral rebound.

Correlations are shown for the breadth of Gag, Pol, Env and total (Gag+Pol+Env) cellular immune responses at peak immunity at week 50 and time to viral rebound following ART discontinuation. P values indicate two-sided Spearman rank-correlation tests. R values indicate correlation coefficients.

Extended Data Figure 10 Rebound kinetic parameters estimated from viral dynamics modelling.

Viral load values following ART discontinuation in each animal were fit to a viral dynamics model using a Bayesian framework. ad, Plots show the median and 95% credible intervals for estimations of the rate of reactivation of cells from the latent reservoir (a), the initial exponential growth rate (b), the immune proliferation rate (c), and the time at which viral load reaches the detection threshold of 200 copies per ml for each treatment group (d). Monkeys treated with both the vaccine and TLR7 agonist exhibited slower viral growth rates and stronger immune responses than all other groups (P < 0.01 for each comparison). These monkeys and monkeys treated with only the vaccine exhibited a lower reservoir exit rate than untreated monkeys (P < 0.05 for each comparison).

PowerPoint slides

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Borducchi, E., Cabral, C., Stephenson, K. et al. Ad26/MVA therapeutic vaccination with TLR7 stimulation in SIV-infected rhesus monkeys. Nature 540, 284–287 (2016). https://doi.org/10.1038/nature20583

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nature20583

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