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:

Broad CTL response is required to clear latent HIV-1 due to dominance of escape mutations

Subjects

Abstract

Despite antiretroviral therapy (ART), human immunodeficiency virus (HIV)-1 persists in a stable latent reservoir1,2, primarily in resting memory CD4+ T cells3,4. This reservoir presents a major barrier to the cure of HIV-1 infection. To purge the reservoir, pharmacological reactivation of latent HIV-1 has been proposed5 and tested both in vitro and in vivo6,7,8. A key remaining question is whether virus-specific immune mechanisms, including cytotoxic T lymphocytes (CTLs), can clear infected cells in ART-treated patients after latency is reversed. Here we show that there is a striking all or none pattern for CTL escape mutations in HIV-1 Gag epitopes. Unless ART is started early, the vast majority (>98%) of latent viruses carry CTL escape mutations that render infected cells insensitive to CTLs directed at common epitopes. To solve this problem, we identified CTLs that could recognize epitopes from latent HIV-1 that were unmutated in every chronically infected patient tested. Upon stimulation, these CTLs eliminated target cells infected with autologous virus derived from the latent reservoir, both in vitro and in patient-derived humanized mice. The predominance of CTL-resistant viruses in the latent reservoir poses a major challenge to viral eradication. Our results demonstrate that chronically infected patients retain a broad-spectrum viral-specific CTL response and that appropriate boosting of this response may be required for the elimination of the latent reservoir.

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: CTL escape variants dominate the latent reservoir of CP-treated but not AP-treated patients.
Figure 2: CD8+ T cells pre-stimulated with a mixture of Gag peptides eliminate autologous CD4+ T cells infected with autologous HIV-1 from resting CD4+ T cells.
Figure 3: CD8+ T cells targeting unmutated epitopes, not epitopes with identified escape mutations, eliminate CTL escape variants.
Figure 4: Broad-spectrum CTLs suppress in vivo replication of HIV-1 from the latent reservoir of the same patients in patient-derived humanized mice.

Similar content being viewed by others

References

  1. Siliciano, J. D. et al. Long-term follow-up studies confirm the stability of the latent reservoir for HIV-1 in resting CD4+ T cells. Nature Med. 9, 727–728 (2003)

    Article  CAS  Google Scholar 

  2. Strain, M. C. et al. Heterogeneous clearance rates of long-lived lymphocytes infected with HIV: intrinsic stability predicts lifelong persistence. Proc. Natl Acad. Sci. USA 100, 4819–4824 (2003)

    Article  ADS  CAS  Google Scholar 

  3. 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 

  4. Chun, T. W. et al. Quantification of latent tissue reservoirs and total body viral load in HIV-1 infection. Nature 387, 183–188 (1997)

    Article  ADS  CAS  Google Scholar 

  5. Richman, D. D. et al. The challenge of finding a cure for HIV infection. Science 323, 1304–1307 (2009)

    Article  ADS  CAS  Google Scholar 

  6. Archin, N. M. et al. Expression of latent HIV induced by the potent HDAC inhibitor suberoylanilide hydroxamic acid. AIDS Res. Hum. Retroviruses 25, 207–212 (2009)

    Article  CAS  Google Scholar 

  7. Contreras, X. et al. Suberoylanilide hydroxamic acid reactivates HIV from latently infected cells. J. Biol. Chem. 284, 6782–6789 (2009)

    Article  CAS  Google Scholar 

  8. Archin, N. M. et al. Administration of vorinostat disrupts HIV-1 latency in patients on antiretroviral therapy. Nature 487, 482–485 (2012)

    Article  ADS  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. Walker, B. D. et al. HIV-specific cytotoxic T lymphocytes in seropositive individuals. Nature 328, 345–348 (1987)

    Article  ADS  CAS  Google Scholar 

  11. Koup, R. A. et al. Temporal association of cellular immune responses with the initial control of viremia in primary human immunodeficiency virus type 1 syndrome. J. Virol. 68, 4650–4655 (1994)

    Article  CAS  Google Scholar 

  12. Borrow, P. et al. Antiviral pressure exerted by HIV-1-specific cytotoxic T lymphocytes (CTLs) during primary infection demonstrated by rapid selection of CTL escape virus. Nature Med. 3, 205–211 (1997)

    Article  CAS  Google Scholar 

  13. Goonetilleke, N. et al. The first T cell response to transmitted/founder virus contributes to the control of acute viremia in HIV-1 infection. J. Exp. Med. 206, 1253–1272 (2009)

    Article  CAS  Google Scholar 

  14. McMichael, A. J., Borrow, P., Tomaras, G. D., Goonetilleke, N. & Haynes, B. F. The immune response during acute HIV-1 infection: clues for vaccine development. Nature Rev. Immunol. 10, 11–23 (2010)

    Article  CAS  Google Scholar 

  15. Phillips, R. E. et al. Human immunodeficiency virus genetic variation that can escape cytotoxic T cell recognition. Nature 354, 453–459 (1991)

    Article  ADS  CAS  Google Scholar 

  16. Koup, R. A. Virus escape from CTL recognition. J. Exp. Med. 180, 779–782 (1994)

    Article  CAS  Google Scholar 

  17. Goulder, P. J. et al. Late escape from an immunodominant cytotoxic T-lymphocyte response associated with progression to AIDS. Nature Med. 3, 212–217 (1997)

    Article  CAS  Google Scholar 

  18. Goulder, P. J. & Watkins, D. I. HIV and SIV CTL escape: implications for vaccine design. Nature Rev. Immunol. 4, 630–640 (2004)

    Article  CAS  Google Scholar 

  19. Henn, M. R. et al. Whole genome deep sequencing of HIV-1 reveals the impact of early minor variants upon immune recognition during acute infection. PLoS Pathog. 8, e1002529 (2012)

    Article  CAS  Google Scholar 

  20. Liu, M. K. et al. Vertical T cell immunodominance and epitope entropy determine HIV-1 escape. J. Clin. Invest. 123, 380–393 (2013)

    CAS  PubMed  Google Scholar 

  21. Rolland, M., Nickle, D. C. & Mullins, J. I. HIV-1 group M conserved elements vaccine. PLoS Pathog. 3, e157 (2007)

    Article  Google Scholar 

  22. Létourneau, S. et al. Design and pre-clinical evaluation of a universal HIV-1 vaccine. PLoS ONE 2, e984 (2007)

    Article  ADS  Google Scholar 

  23. Yu, X. G. et al. Consistent patterns in the development and immunodominance of human immunodeficiency virus type 1 (HIV-1)-specific CD8+ T-cell responses following acute HIV-1 infection. J. Virol. 76, 8690–8701 (2002)

    Article  CAS  Google Scholar 

  24. 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 

  25. Rongvaux, A. et al. Development and function of human innate immune cells in a humanized mouse model. Nature Biotechnol. 32, 364–372 (2014)

    Article  CAS  Google Scholar 

  26. 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 

  27. Ananworanich, J. et al. Impact of multi-targeted antiretroviral treatment on gut T cell depletion and HIV reservoir seeding during acute HIV infection. PLoS ONE 7, e33948 (2012)

    Article  ADS  CAS  Google Scholar 

  28. Goulder, P. J. et al. Evolution and transmission of stable CTL escape mutations in HIV infection. Nature 412, 334–338 (2001)

    Article  ADS  CAS  Google Scholar 

  29. Le Gall, S., Stamegna, P. & Walker, B. D. Portable flanking sequences modulate CTL epitope processing. J. Clin. Invest. 117, 3563–3575 (2007)

    Article  CAS  Google Scholar 

  30. Hansen, S. G. et al. Cytomegalovirus vectors violate CD8+ T cell epitope recognition paradigms. Science 340, 1237874 (2013)

    Article  Google Scholar 

  31. Magoč, T. & Salzberg, S. L. FLASH: fast length adjustment of short reads to improve genome assemblies. Bioinformatics 27, 2957–2963 (2011)

    Article  Google Scholar 

  32. Langmead, B. & Salzberg, S. L. Fast gapped-read alignment with Bowtie 2. Nature Methods 9, 357–359 (2012)

    Article  CAS  Google Scholar 

  33. Streeck, H., Frahm, N. & Walker, B. D. The role of IFN-γ Elispot assay in HIV vaccine research. Nature Protocols 4, 461–469 (2009)

    Article  CAS  Google Scholar 

  34. Siliciano, J. D. & Siliciano, R. F. Enhanced culture assay for detection and quantitation of latently infected, resting CD4+ T-cells carrying replication-competent virus in HIV-1-infected individuals. Methods Mol. Biol. 304, 3–15 (2005)

    PubMed  Google Scholar 

Download references

Acknowledgements

We thank all study participants. We thank J. Blankson for critical advice to the project; L. Alston and R. Hoh for coordinating patient recruitment; J. Alderman, C. Weibel and E. Henchey for technical assistance in the animal study. We thank the National Institutes of Health (NIH) AIDS Reagent Program for providing HIV-1 consensus B peptides. R.F.S. is supported by the Howard Hughes Medical Institute, by the Martin Delaney CARE and DARE Collaboratories (NIH grants AI096113 and 1U19AI096109), by an ARCHE Collaborative Research Grant from the Foundation for AIDS Research (amFAR 108165-50-RGRL), by the Johns Hopkins Center for AIDS Research (P30AI094189), and by NIH grant 43222. L.S. is supported by NIH grant T32 AI07019. R.A.F. is supported by the Bill and Melinda Gates Foundation and the Howard Hughes Medical Institute.

Author information

Authors and Affiliations

Authors

Contributions

K.D., L.S., R.A.F. and R.F.S. conceived and designed the research studies; K.D., J.L., H.Z., J.B.M. and L.S. performed the in vitro experiments; K.D., A.R., L.W., C.G., A.J.M., D.M.V., G.D.Y., T.S., P. K. and L.S. performed animal experiments; C.M.D., G.G., H.L.M. and S.G.D. provided patient samples; K.D., M.P., L.W., H.H., J.D.S., S.L.S., L.S. and R.F.S. analysed data; K.D., L.S. and R.F.S. wrote the manuscript.

Corresponding authors

Correspondence to Richard A. Flavell, Liang Shan or Robert F. Siliciano.

Ethics declarations

Competing interests

C.G., A.J.M., D.M.V. and G.D.Y. are employees and shareholders of Regeneron Pharmaceuticals, Inc.

Extended data figures and tables

Extended Data Figure 1 CTL escape variants dominate the latent reservoir of CP-treated HIV-1-positive individuals, but not AP-treated individuals.

Frequency of variants in Gag CTL epitopes in proviruses from resting CD4+ T cells. Results of all 25 patients tested are shown. Only optimal CTL epitopes relevant to each patient’s HLA type are listed in linear positional order on the x-axis. Results from both PacBio (left bar) and MiSeq (right bar) sequencing platforms are shown for each epitope. The absence of bars above a listed epitope indicates that only wild-type sequences were detected. For each mutation in a CTL epitope, information regarding the effect of the mutation on CTL recognition from the Los Alamos National Laboratory (LANL) HIV Molecular Immunology Database or from ELISpot assays described in Methods was used to assign the mutation to one of the categories indicated at the bottom. See Methods for definitions of these categories.

Extended Data Figure 2 Characterization of CTL responses against HIV-1 Gag epitopes by interferon-γ ELISpot.

Results of seven patients tested are shown. The peptides tested are listed for each patient in each graph. Error bars represent s.e.m., n = 3.

Extended Data Figure 3 Partial Gag sequences from proviral DNA and outgrowth virus from resting CD4+ T cells from eight CP-treated patients.

CTL epitopes with no observed variation are highlighted in blue. Documented escape mutations (red shading), inferred escape mutations (yellow shading), diminished response (pink shading), susceptible form (green shading) or undetermined variations (grey shading) in relevant optimal epitopes are indicated. See Methods for definitions of these types of mutations. This figure supplements Fig. 1e, as a total of nine CP-treated patients were tested.

Extended Data Figure 4 CD8+ T cells pre-stimulated with a mixture of consensus B Gag peptides eliminate autologous CD4+ T cells infected with autologous HIV-1 from resting CD4+ T cells.

a, HIV-1 isolated from ART-treated individuals replicates as well as the laboratory strain virus BaL. p24 values represent mean of three replicates. Error bars represent s.e.m., n = 3. b, CD8+ T cells are not stimulated after co-culture with PHA-activated CD4+ T cells. c, A representative flow cytometric analysis of CTL-mediated killing after co-culture of infected CD4+ T cells with autologous CD8+ T cells. CTL activity is measured by the percentage of Gag-positive, CD8-negative cells after 3 days of co-culture relative to cultures without CD8+ T cells. d, Pre-stimulated CD8+ T cells eliminate autologous infected CD4+ T cells more efficiently than non-stimulated CD8+ T cells. All results were normalized to the CD4 only control group. Error bars represent s.e.m., n = 3. *P < 0.05, **P < 0.01, ***P < 0.001, NS, not significant (P > 0.05), paired t-test.

Extended Data Figure 5 The elimination of infected CD4+ T cells is mediated by direct killing by autologous CD8+ T cells.

a, Killing of infected CD4+ T cells is enhanced by increased effector to target ratios for both pre-stimulated and non-stimulated CD8+ T cells. b, Killing of the infected CD4+ T cells depends on direct cell–cell contact between CD4+ T cells and CTLs. All results were normalized to the CD4+ only control group. Error bars represent s.e.m., n = 3. *P < 0.05, **P < 0.01, ***P < 0.001, NS, not significant (P > 0.05), paired t-test.

Extended Data Figure 6 Viral dynamics and depletion of CD4+ T cells in humanized mice.

a, Viral dynamics in CP18-infected MIS(KI)TRG mice. CP18-derived MIS(KI)TRG mice were infected with autologous HIV-1. Plasma HIV-1 RNA levels were measured from day 0 to day 56. b, Depletion of CD4+ T cells in peripheral blood of HIV-1 BaL-infected mice. MIS(KI)TRG mice engrafted with fetal liver CD34+ cells were infected with HIV-1 BaL. The CD4 to CD8 ratio in peripheral blood was measured by fluorescence-activated cell sorting (FACS) from day 0 to day 29 after infection. Error bars represent s.e.m., n = 5. c, Depletion of CD4+ T cells in spleen of HIV-1 BaL-infected mice. MIS(KI)TRG mice engrafted with fetal liver CD34+ cells were infected with HIV-1 BaL. The CD4 to CD8 ratio in spleen was measured by FACS 20 days after infection. Medians and P values from Mann–Whitney test are shown. d, Detection of cell-associated HIV-1 RNA in T cells and macrophages/monocytes. CD3+ and CD14+ human cells from HIV-1-infected MIS(KI)TRG mice from spleen and lung were purified by FACS. CD3CD14 cells were also collected as controls. Cell-associated HIV-1 RNA was quantified by gag-specific quantitative polymerase chain reaction (qPCR). Error bars represent s.e.m., n = 3. *P < 0.05, unpaired t-test.

Extended Data Figure 7 HIV-1 infection occurs in peripheral blood and tissues in humanized mice.

a, Engraftment levels of MIS(KI)TRG mice with fetal liver or patient CD34+ cells. b, Memory CD4+ T cells are detected in MIS(KI)TRG mice after infection. MIS(KI)TRG mice were infected with HIV-1 BaL. Peripheral blood and indicated tissues from infected mice were collected at 20 days post-infection. Memory CD4+ T cells were determined by CD45RO staining. c, Total number of cell-associated HIV-1 DNA in blood and tissues. DNA from peripheral blood or indicated tissues was isolated for the measurement of total amount of cell-associated HIV-1 DNA by real-time PCR. b, c, Medians and P values from Mann–Whitney test are shown.

Extended Data Figure 8 Broad-spectrum CTLs suppress in vivo infection of patient-derived humanized mice with autologous latent HIV-1.

The generation of patient CP36-derived humanized mice is described in Fig. 4. Mice were infected with autologous viruses at 6 weeks old. CD8+ T cells from patient CP36 were pre-stimulated with the mixture of Gag peptides or left untreated for 6 days in vitro, and were injected into mice intravenously 9 days after infection. Plasma HIV-1 RNA (a) and HIV-1 DNA (b) in peripheral blood were measured by real-time PCR.

Extended Data Table 1 Characteristics of study subjects

Supplementary information

Supplementary Tables

This file contains Supplementary Table 1. (PDF 229 kb)

PowerPoint slides

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Deng, K., Pertea, M., Rongvaux, A. et al. Broad CTL response is required to clear latent HIV-1 due to dominance of escape mutations. Nature 517, 381–385 (2015). https://doi.org/10.1038/nature14053

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nature14053

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