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:

CTLA-4 and PD-1 dual blockade induces SIV reactivation without control of rebound after antiretroviral therapy interruption

Abstract

The primary human immunodeficiency virus (HIV) reservoir is composed of resting memory CD4+ T cells, which often express the immune checkpoint receptors programmed cell death protein 1 (PD-1) and cytotoxic T lymphocyte-associated protein 4 (CTLA-4), which limit T cell activation via synergistic mechanisms. Using simian immunodeficiency virus (SIV)-infected, long-term antiretroviral therapy (ART)-treated rhesus macaques, we demonstrate that PD-1, CTLA-4 and dual CTLA-4/PD-1 immune checkpoint blockade using monoclonal antibodies is well tolerated, with evidence of bioactivity in blood and lymph nodes. Dual blockade was remarkably more effective than PD-1 blockade alone in enhancing T cell cycling and differentiation, expanding effector-memory T cells and inducing robust viral reactivation in plasma and peripheral blood mononuclear cells. In lymph nodes, dual CTLA-4/PD-1 blockade, but not PD-1 alone, decreased the total and intact SIV-DNA in CD4+ T cells, and SIV-DNA and SIV-RNA in B cell follicles, a major site of viral persistence during ART. None of the tested interventions enhanced SIV-specific CD8+ T cell responses during ART or viral control after ART interruption. Thus, despite CTLA-4/PD-1 blockade inducing robust latency reversal and reducing total levels of integrated virus, the degree of reservoir clearance was still insufficient to achieve viral control. These results suggest that immune checkpoint blockade regimens targeting PD-1 and/or CTLA-4, if performed in people living with HIV with sustained aviremia, are unlikely to induce HIV remission in the absence of additional interventions.

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: CTLA-4 and PD-1 blockade is biologically active in SIV-infected, ART-treated RMs.
Fig. 2: T cell proliferative and effector responses are synergistically improved by dual CTLA-4/PD-1 blockade.
Fig. 3: CTLA-4 and PD-1 combined blockade induces robust viral reactivation in ART-treated, SIV-infected RMs.
Fig. 4: CTLA-4 and dual blockade, but not PD-1 blockade, reduces the total and intact SIV-DNA in LN.

Similar content being viewed by others

Data availability

The data that support the findings of this study are available from the corresponding author on reasonable request. Sequences of gp160 env are available in GenBank (accession nos. MN856887MN857133).

References

  1. McGary, C. S. et al. CTLA-4+PD-1 memory CD4+ T cells critically contribute to viral persistence in antiretroviral therapy-suppressed, SIV-infected rhesus macaques. Immunity 47, 776–788 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  2. 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).

    CAS  PubMed  Google Scholar 

  3. Fromentin, R. et al. CD4+ T cells expressing PD-1, TIGIT and LAG-3 contribute to HIV persistence during ART. PLoS Pathog. 12, e1005761 (2016).

    PubMed  PubMed Central  Google Scholar 

  4. Perez, V. L. et al. Induction of peripheral T cell tolerance in vivo requires CTLA-4 engagement. Immunity 6, 411–417 (1997).

    CAS  PubMed  Google Scholar 

  5. Fife, B. T. & Bluestone, J. A. Control of peripheral T-cell tolerance and autoimmunity via the CTLA-4 and PD-1 pathways. Immunol. Rev. 224, 166–182 (2008).

    CAS  PubMed  Google Scholar 

  6. Perreau, M. et al. Follicular helper T cells serve as the major CD4 T cell compartment for HIV-1 infection, replication and production. J. Exp. Med. 210, 143–156 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  7. Day, C. L. et al. PD-1 expression on HIV-specific T cells is associated with T-cell exhaustion and disease progression. Nature 443, 350–354 (2006).

    CAS  PubMed  Google Scholar 

  8. Trautmann, L. et al. Upregulation of PD-1 expression on HIV-specific CD8+ T cells leads to reversible immune dysfunction. Nat. Med. 12, 1198–1202 (2006).

    CAS  PubMed  Google Scholar 

  9. Wightman, F. et al. Effect of ipilimumab on the HIV reservoir in an HIV-infected individual with metastatic melanoma. AIDS 29, 504–506 (2015).

    PubMed  PubMed Central  Google Scholar 

  10. Evans, V. A. et al. Programmed cell death-1 contributes to the establishment and maintenance of HIV-1 latency. AIDS 32, 1491–1497 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  11. Guihot, A. et al. Drastic decrease of the HIV reservoir in a patient treated with nivolumab for lung cancer. Ann. Oncol. 29, 517–518 (2018).

    CAS  PubMed  Google Scholar 

  12. Scully, E. P. et al. Inconsistent HIV reservoir dynamics and immune responses following anti-PD-1 therapy in cancer patients with HIV infection. Ann. Oncol. 29, 2141–2142 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  13. Le Garff, G. et al. Transient HIV-specific T cells increase and inflammation in an HIV-infected patient treated with nivolumab. AIDS 31, 1048–1051 (2017).

    PubMed  Google Scholar 

  14. Cecchinato, V. et al. Immune activation driven by CTLA-4 blockade augments viral replication at mucosal sites in simian immunodeficiency virus infection. J. Immunol. 180, 5439–5447 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  15. Hryniewicz, A. et al. CTLA-4 blockade decreases TGF-beta, IDO and viral RNA expression in tissues of SIVmac251-infected macaques. Blood 108, 3834–3842 (2006).

    CAS  PubMed  PubMed Central  Google Scholar 

  16. Velu, V. et al. Enhancing SIV-specific immunity in vivo by PD-1 blockade. Nature 458, 206–210 (2009).

    CAS  PubMed  Google Scholar 

  17. Mylvaganam, G. H. et al. Combination anti-PD-1 and antiretroviral therapy provides therapeutic benefit against SIV. JCI Insight 3, 122940 (2018).

    PubMed  Google Scholar 

  18. Bekerman, E. et al. PD-1 blockade and TLR7 activation lack therapeutic benefit in chronic simian immunodeficiency virus-infected macaques on antiretroviral therapy. Antimicrob. Agents Chemother. 63, e01163-19 (2019).

    PubMed  PubMed Central  Google Scholar 

  19. Wang, C. et al. In vitro characterization of the anti-PD-1 antibody nivolumab, BMS-936558 and in vivo toxicology in non-human primates. Cancer Immunol. Res. 2, 846–856 (2014).

    CAS  PubMed  Google Scholar 

  20. Hardtke, S., Ohl, L. & Forster, R. Balanced expression of CXCR5 and CCR7 on follicular T helper cells determines their transient positioning to lymph node follicles and is essential for efficient B-cell help. Blood 106, 1924–1931 (2005).

    CAS  PubMed  Google Scholar 

  21. Velu, V. et al. Induction of Th1-biased T follicular helper (Tfh) cells in lymphoid tissues during chronic simian immunodeficiency virus infection defines functionally distinct germinal center Tfh cells. J. Immunol. 197, 1832–1842 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  22. Pearce, E. L. et al. Control of effector CD8+ T cell function by the transcription factor eomesodermin. Science 302, 1041–1043 (2003).

    CAS  PubMed  Google Scholar 

  23. Chen, Z. et al. TCF-1-centered transcriptional network drives an effector versus exhausted CD8 T cell-fate decision. Immunity 51, 840–855 (2019).

    CAS  PubMed  Google Scholar 

  24. Fromentin, R. et al. PD-1 blockade potentiates HIV latency reversal ex vivo in CD4+ T cells from ART-suppressed individuals. Nat. Commun. 10, 814 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  25. Estes, J. D. et al. Defining total-body AIDS-virus burden with implications for curative strategies. Nat. Med. 23, 1271–1276 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  26. Bender, A. M. et al. The landscape of persistent viral genomes in ART-treated SIV, SHIV and HIV-2 infections. Cell Host Microbe 26, 73–85 (2019).

    CAS  PubMed  Google Scholar 

  27. Deleage, C. et al. Defining HIV and SIV reservoirs in lymphoid tissues. Pathog. Immun. 1, 68–106 (2016).

    PubMed  PubMed Central  Google Scholar 

  28. Bar, K. J. et al. Effect of HIV antibody VRC01 on viral rebound after treatment interruption. N. Engl. J. Med. 375, 2037–2050 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  29. Amancha, P. K. et al. In vivo blockade of the programmed cell death-1 pathway using soluble recombinant PD-1-Fc enhances CD4+ and CD8+ T cell responses but has limited clinical benefit. J. Immunol. 191, 6060–6070 (2013).

    CAS  PubMed  Google Scholar 

  30. Dyavar Shetty, R. et al. PD-1 blockade during chronic SIV infection reduces hyperimmune activation and microbial translocation in rhesus macaques. J. Clin. Invest. 122, 1712–1716 (2012).

    PubMed  PubMed Central  Google Scholar 

  31. Finnefrock, A. C. et al. PD-1 blockade in rhesus macaques: impact on chronic infection and prophylactic vaccination. J. Immunol. 182, 980–987 (2009).

    CAS  PubMed  Google Scholar 

  32. Okoye, A. A. et al. Early antiretroviral therapy limits SIV reservoir establishment to delay or prevent post-treatment viral rebound. Nat. Med. 24, 1430–1440 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  33. Borducchi, E. N. et al. Publisher correction: antibody and TLR7 agonist delay viral rebound in SHIV-infected monkeys. Nature 564, E8 (2018).

    CAS  PubMed  Google Scholar 

  34. Weiss, L. et al. Human immunodeficiency virus-driven expansion of CD4+CD25+ regulatory T cells, which suppress HIV-specific CD4 T-cell responses in HIV-infected patients. Blood 104, 3249–3256 (2004).

    CAS  PubMed  Google Scholar 

  35. Nilsson, J. et al. HIV-1-driven regulatory T-cell accumulation in lymphoid tissues is associated with disease progression in HIV/AIDS. Blood 108, 3808–3817 (2006).

    CAS  PubMed  PubMed Central  Google Scholar 

  36. Larkin, J. et al. Combined nivolumab and ipilimumab or monotherapy in untreated melanoma. N. Engl. J. Med. 373, 23–34 (2015).

    PubMed  PubMed Central  Google Scholar 

  37. Hao, C. et al. Efficacy and safety of anti-PD-1 and anti-PD-1 combined with anti-CTLA-4 immunotherapy to advanced melanoma: a systematic review and meta-analysis of randomized controlled trials. Medicine (Baltimore) 96, e7325 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  38. Koelzer, V. H. et al. Systemic inflammation in a melanoma patient treated with immune checkpoint inhibitors—an autopsy study. J. Immunother. Cancer 4, 13 (2016).

    PubMed  PubMed Central  Google Scholar 

  39. Cook, M. R. & Kim, C. Safety and efficacy of immune checkpoint inhibitor therapy in patients with HIV infection and advanced-stage cancer: a systematic review. JAMA Oncol. 5, 1049–1054 (2019).

    PubMed  Google Scholar 

  40. Hansen, S. G. et al. Profound early control of highly pathogenic SIV by an effector memory T-cell vaccine. Nature 473, 523–527 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  41. Del Prete, G. Q. et al. Short communication: comparative evaluation of coformulated injectable combination antiretroviral therapy regimens in simian immunodeficiency virus-infected rhesus macaques. AIDS Res. Hum. Retroviruses 32, 163–168 (2016).

    PubMed  PubMed Central  Google Scholar 

  42. Scott, M. J. et al. ‘In-format’ screening of a novel bispecific antibody format reveals significant potency improvements relative to unformatted molecules. MAbs 9, 85–93 (2017).

    CAS  PubMed  Google Scholar 

  43. Chhabra, M. et al. Binding proteins. Patent WO/2018/083087 A2 (2018).

  44. Micci, L. et al. Interleukin-21 combined with ART reduces inflammation and viral reservoir in SIV-infected macaques. J. Clin. Invest. 125, 4497–4513 (2015).

    PubMed  PubMed Central  Google Scholar 

  45. Pallikkuth, S. et al. Maintenance of intestinal Th17 cells and reduced microbial translocation in SIV-infected rhesus macaques treated with interleukin (IL)-21. PLoS Pathog. 9, e1003471 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  46. Li, H. & Pauza, C. D. CD25+ Bcl6low T follicular helper cells provide help to maturing B cells in germinal centers of human tonsil. Eur. J. Immunol. 45, 298–308 (2015).

    PubMed  Google Scholar 

  47. Li, H. et al. Envelope residue 375 substitutions in simian-human immunodeficiency viruses enhance CD4 binding and replication in rhesus macaques. Proc. Natl Acad. Sci. USA 113, E3413–E3422 (2016).

    CAS  PubMed  Google Scholar 

  48. Amara, R. R. et al. Control of a mucosal challenge and prevention of AIDS by a multiprotein DNA/MVA vaccine. Science 292, 69–74 (2001).

    CAS  PubMed  Google Scholar 

  49. Hansen, S. G. et al. Addendum: immune clearance of highly pathogenic SIV infection. Nature 547, 123–124 (2017).

    CAS  PubMed  Google Scholar 

  50. Roederer, M., Nozzi, J. L. & Nason, M. C. SPICE: exploration and analysis of post-cytometric complex multivariate datasets. Cytometry A 79, 167–174 (2011).

    PubMed  PubMed Central  Google Scholar 

  51. Bruner, K. M. et al. A quantitative approach for measuring the reservoir of latent HIV-1 proviruses. Nature 566, 120–125 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  52. Policicchio, B. B. et al. Dynamics of simian immunodeficiency virus two-long-terminal-repeat circles in the presence and absence of CD8+ cells. J. Virol. 92, e02100-17 (2018).

    PubMed  PubMed Central  Google Scholar 

  53. Salantes, D. B. et al. HIV-1 latent reservoir size and diversity are stable following brief treatment interruption. J. Clin. Invest. 128, 3102–3115 (2018).

    PubMed  PubMed Central  Google Scholar 

  54. 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).

    CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank S. Ehnert, C. Souder and E. Strobert (Research Resources and Veterinary Medicine) at YNPRC for providing animal and veterinary care, as well as the Emory Flow Cytometry Core. We thank C. Ashman, G. Jones, L. Anderson and A. Barnard from GSK for assisting with the preparation and quality control of the therapeutic antibodies, as well as K. Fraley, A. Mayer and G. Page from GSK for measuring antibody and anti-drug antibody levels. We also thank R. Shoemaker, K. Oswald and W. Bosche at Leidos Biomedical Research for technical assistance. The SIVmac239 strain used to infect the RMs was provided by K. Van Rompay of UC-Davis, anti-Gag tetramers were provided by D. Long at the NIH Tetramer Core Facility at Emory, and ART was supplied through ViiV Healthcare and GSK. This work was supported by the NIAID, NIH, under awards R01AI116379 and R21/R33AI116171 to M.P. and award UM1AI124436 (Emory CIAR). Support for this work was also provided by GlaxoSmithKline and Qura Therapeutics under subcontract 5105399 from the Collaboratory of AIDS Researchers for Eradication (CARE, 1UM1AI126619-01) to D.M., the Virology & Drug Discovery Core of Emory CFAR (P30AI050409), ORIP/OD awards P51OD011132 (YNPRC) and P51OD011092 (ONPRC), and in part with federal funds from the National Cancer Institute, National Institutes of Health, under contract no. HHSN261200800001E. The content of this publication does not necessarily reflect the views or policies of the Department of Health and Human Services, nor does mention of trade names, commercial products or organizations imply endorsement by the US Government.

Author information

Authors and Affiliations

Authors

Contributions

J.H. contributed to conceptualization, methodology, formal analysis, investigation, writing (original draft, review and editing) and visualization. S.G. contributed to conceptualization, formal analysis, investigation and writing the original draft. C.N.C. contributed to formal analysis, investigation and visualization. H.W., C.G., S.L.M.R., J.L.R., K.N., M.N., K.B.-S., C.K., M.P., L.M., B.C., S.J., A.S. and A.A.K. contributed to investigation. E.L. and J.S. contributed to formal analysis and investigation. S.D.F. contributed to methodology and investigation. C.S.M. contributed to conceptualization and investigation. B.J. contributed to resources. H.A.-M., J.L. and J.D.E. contributed to methodology and writing (review and editing). D.M.M. contributed to methodology, resources, writing (review and editing) and funding acquisition. G.S. contributed to writing (review and editing) and funding acquisition. K.J.B. contributed to methodology, formal analysis, and writing (review and editing). D.F. contributed to conceptualization, methodology, resources, writing (review and editing), supervision and funding acquisition. M.P. contributed to conceptualization, methodology, resources, writing (original draft, review and editing), supervision and funding acquisition.

Corresponding author

Correspondence to Mirko Paiardini.

Ethics declarations

Competing interests

S.G., C.G., J.S., A.S., B.J., A.A.K., H.A.-M. and D.F. are employed by and/or have financial interests in GlaxoSmithKline or ViiV Healthcare.

Additional information

Peer review information Alison Farrell is the primary editor on this article and managed its editorial process and peer review in collaboration with the rest of the editorial team.

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

Extended data

Extended Data Fig. 1 Plasma viral loads during acute SIV infection and subsequent decay during ART.

Viral loads (SIVmac239 RNA copies/mL) were quantified in plasma utilizing RT-qPCR and represented on a logarithmic scale. The terminal measurement (indicated in light red) for each animal indicates the start of ICB therapy (d0 p.t.). Initiation of ICB therapy was dependent on duration of undetectable viremia in plasma. Data sets are grouped based on ICB treatment as follows: (a) VHDUM (control; n=6), black; (b) anti-CTLA-4 mAb (αCTLA-4; n=6), blue; (c) anti-PD-1 mAb (αPD-1; n=6), pink; (d) anti-CTLA-4 plus anti-PD-1 mAb (combined blockade; n=7), red; and (e) anti-CTLA-4/PD-1 mAbdAb (BsAb; n=8), purple. Individual RMs are indicated by shape with closed data points indicating viral reactivation in plasma during subsequent ICB. The vertical dotted line indicates ART initiation (d60 p.i.) and the shaded gray area represents ongoing ART administration. The horizontal dotted line indicates the assay’s limit of detection (≤60 copies/mL) with undetectable events represented as 30 copies/mL.

Extended Data Fig. 2 Macaque characteristics and ICB antibodies.

aAge in months at SIV infection (d0 p.i.; post-infection) and at ICB (d0 p.t.; post-treatment). bIndicates the number of CD4+ T cells per μL of PB at chronic infection prior to ART initiation (d52 p.i.) as determined by complete blood counts combined with flow cytometry. cSIV-RNA viral loads per mL of plasma was measured by quantitative RT-qPCR with a limit of detection of ≤60 copies/mL at acute (d14 p.i) and chronic infection (d52 p.i.). dDays of undetectable (UD) viral load prior to ICB treatment were calculated from the first UD observation following ART initiation and does not account for subsequent blips. eIndicates whether Fc gamma receptor has been disabled by a LAGA mutation (L235A and G237A) and is capable of antibody-dependent cellular cytotoxicity (ADCC), antibody-dependent cellular phagocytosis (ADCP), ect. fIndicates the pharmacological generic equivalent of the variable domain and epitope specificity.

Extended Data Fig. 3 Blockade pharmacokinetics and emergence of anti-drug antibodies.

(a) Plasma concentrations (ng/mL) of Co-IR blocking monoclonal antibodies (mAb) were quantified with an antibody capture assay; stratified by treatment group; and represented on a logarithmic scale. Blood was drawn immediately prior to- and 5 mins following ICB administration. (b) Using only blood drawn prior to ICB infusion, the levels of anti-drug antibodies (ECL, electrochemiluminescence) were quantified by MSD assay, which were utilized to calculate the percentage of inhibition. Vertical arrows and dotted lines represent either an ICB infusion or ART interruption, as indicated, with the gray shaded area representing ongoing ART. Individual RMs are indicated by shape with closed data points indicating animals with viral reactivation in plasma.

Extended Data Fig. 4 Combined blockade leads to a limited restoration of cytolytic functionality in ICR-expressing CD8+ T cell subsets.

By flow cytometry, the frequencies of granzyme B+ cells were determined within (a) CTLA-4+PD-1+ and (b) CTLA-4+PD-1- memory CD8+ T cells in LN following the fourth ICB infusion (d28 p.t.). All RMs are color-coded and grouped based on ICB therapy with population sizes as indicated for all analyses: controls (n=6), black; αCTLA-4 (n=6), blue; αPD-1 (n=6), pink; combined blockade (n=7), red; and BsAb (n=8), purple. Averaged data are presented as the mean ± s.e.m. and were analyzed with a two-sided Kruskal-Wallis test with Dunn’s correction for multiple comparisons relative to controls.

Extended Data Fig. 5 SIV-specific CD8+ T cells display an exhausted phenotype that is not rescued by ICB during suppressive ART, but by viral rebound following ATI.

Within memory CD8+ T cells at the on-ART, pretreatment baseline (d-29 p.t.; n=5 ICB-treated, Mamu-A*01+ RMs) the frequencies of exhaustion, proliferative and activation biomarkers (as indicated above) were quantified via flow cytometry in SIV-specific and non-specific subsets, as assessed by anti-Gag-CM9 staining (as indicated below) in (a) LN and (b) PBMCs. Likewise, within LN SIV-specific memory CD8+ T cells the frequencies of (c) Ki-67+, (d) HLA-DR+CD38+, (e) T-bet+, and (f) Granzyme B+ cells were measured at the pre-ICB baseline and following the fourth ICB infusion (d28 p.t.). Likewise, the frequencies of (g) Ki-67+, (h) HLA-DR+CD38+, (i) T-bet+, and (j) Granzyme B+ cells within SIV-specific memory CD8+ T cells in PBMCs were quantified at the pre-ICB baseline, following the third ICB treatment (d21 p.t.), and 15 days following ART interruption (d50 p.t.). Analyses were conducted in up to 6 Mamu-A*01+ RMs (g,h,i,j), of which 5 were ICB-treated (a,b,c,d,e,f). Each data point represents an individual animal, as indicated by shape, and those with ICB-related viral reactivation in plasma are represented as closed data points. Averaged data are presented as the mean ± s.e.m. and were analyzed with (a,b) a one-way, pair-wise ANOVA using a Bonferroni correction for multiple corrections.

Extended Data Fig. 6 SIV-specific CD8+ T cell responses are not enhanced by blockade of PD-1 and/or CTLA-4 during long-term ART.

In 5 Mamu-A*01+, ICB-treated RMs, the frequency of SIV-specific cells, as determined via anti-Gag-CM9 tetramer staining with flow cytometry, were measured in (a) LN and (b) PBMC memory CD8+ T cells at treatment baseline (d-29 p.t.) and following ICB (d21 or 28 p.t.). The number of interferon gamma (IFN-γ) spot forming units (SFU) per 106 mononuclear cells were quantified by ELISpot upon SIV-Gag stimulation in (c) PBMCs as a fold-change (d21 p.t. relative to d-29 p.t.) and (d) cross-sectionally in LN (d21 p.t). Points represent unique animals, as indicated by shape and fill, as an average of three replicates with DMSO background subtracted. All RMs are color-coded and grouped based on ICB therapy as follows with population sizes as indicated for all ELISpot analyses: controls (n=6), black; αCTLA-4 (n=6), blue; αPD-1 (n=5), pink; combined blockade (n=7), red; and BsAb (n=8), purple. From ex vivo SIV-Gag-stimulated PBMCs with unstimulated background subtracted, the distribution of cytokine co-expression was determined via flow cytometry in memory (e) CD4+ and (f) CD8+ T cells before (d-29 p.t.) and during (d21 p.t.; indicated at left) ICB administration (indicated above). Rainbow-colored inner pie wedges represent each Boolean combination of cytokine co-expression (annotated at far right); whereas, the stacked, outer concentric rings overlap with pie wedges that are positive for that cytokine (indicated at middle right): IFN-γ+, red; IL-2+, orange; TNFα+, green; and CD107a+, purple. Population sizes for all cytokine analyses are as follows: controls (n=6), black; αCTLA-4 (n=6), blue; αPD-1 (n=6), pink; combined blockade (n=7), red; and BsAb (n=8), purple. Averaged data are presented as the mean ± s.e.m., and were analyzed with a two-sided (c,d) Mann-Whitney U test or (e,f) a Permutation test.

Extended Data Fig. 7 Combined blockade increases the ratio of cell-associated SIV-RNA relative to SIV-DNA content in PBMCs and limit intact provirus in LN CD4+ T cells.

(a) The number of log-transformed cell-associated SIV-DNA copies per 106 PBMCs were quantified by 12 replicate reaction RT-qPCR prior to the first infusion (d0 p.t.), and 1 week following the first (d7 p.t.) and fourth infusions (d28 p.t.); from which (b) the ratio of SIV-RNA to -DNA copies was computed. All RMs are color-coded and grouped based on ICB therapy as follows with population sizes as indicated for all RT-qPCR analyses: controls (n=6), black; αCTLA-4 (n=6), blue; αPD-1 (n=6), pink; combined blockade (n=7), red; and BsAb (n=8), purple. By IPDA with ddPCR, (c) the DNA shearing index pre- and post-ICB, and (d) the fold change in intact provirus per 106 cells relative to pre-treatment baseline were quantified in LN CD4+ cells. For all IPDA analyses, population sizes are as follows: control (n=3), αCTLA-4 (n=6), αPD-1 (n=5), combined blockade (n=7), and BsAb (n=7). (e) An example IPDA plot is given for the amplification of the env relative response element (RRE) on VIC dye against pol on FAM dye in relative fluorescence units (RFU). A positivity cutoff of 1250 is used for env and 2000 for pol. Sample plots shown (as indicated in red) are as follows: negative control, merged analysis from no-template control and PBMCs at pre-infection from RLt16; env positive control, env gBlock (IDT); pol positive control, pol gBlock (IDT); and IPDA (shown are LN CD4+ cells at d-29 p.t. from RWs16; 1 of 58 image sets of 8 replicate reactions). Averaged data are presented as the mean ± s.e.m. Each data point represents an individual animal, as indicated by shape, and those with ICB-related viral reactivation in plasma are represented as closed data points. Data were analyzed with a two-sided (a,b) Wilcoxon matched-pairs signed rank test, (d) a Mann-Whitney U test, or (c) a two-way ANOVA with Bonferroni’s correction for multiple comparisons relative to baseline and controls.

Extended Data Fig. 8 ICB does not differentially impact viral reservoir content between lymphoid and myeloid lineages, and vDNA content correlates with intact proviral genomes.

(a) The number of LN vRNA+ cells per 105 cells were quantified by RNAscope before (d-29) and after ICB therapy (d28 p.t.). All RMs are color-coded and grouped based on ICB therapy as follows with population sizes as indicated for RNAscope analyses: controls (n=6), black; αCTLA-4 (n=6), blue; αPD-1 (n=6), pink; combined blockade (n=7), red; and BsAb (n=8), purple. In the LN B cell follicle (BCF) and T cell zone (TCZ), the number of (b) vDNA or (c) vRNA cells per 105 cells were measured based on their co-expression of CD3 (lymphoid) or CD68/CD163 (MØ; myeloid) in a subset of ICB-responsive RMs (n=19). (d) In situ immunofluorescence staining for CD3 (green), CD68/CD163 (blue), and DAPI (grey) combined with hybridization for SIV vDNA (red) in LN TCZ or BCF (annotated at upper right) prior to or following ICB in representative RMs (3 of 38 unique samples; up to two tissue sections were analyzed). vDNA positive cells are annotated with a white arrow and successive magnification within the indicated boxed area (white) are shown below. (e) From LN at d28 p.t., the log-transformed frequency of vDNA+ cells per 106 cells were correlated against the contemporaneous frequency of log-transformed intact proviral genomes per 106 CD4+ cells (n=29). Averaged data are presented as the mean ± s.e.m. and were analyzed with (a,b,c) a two-sided Wilcoxon matched-pairs signed rank test relative to baseline or (e) a Pearson correlation coefficient.

Extended Data Fig. 9 Dual CTLA-4/PD-1 blockade does not control or delay viral rebound after ATI.

The delay in viral rebound by ICB was analyzed as the average number of days until detectable viral loads were observed in plasma with RT-qPCR. Plasma viral loads following ATI are shown for individual RMs by ICB: (b) control, (c) αCTLA-4, (d) αPD-1, (e) combined blockade, and (f) BsAb. Mamu-A*01+ RMs are shown with a bold connecting line and animals with prior ICB-related viral reactivation in plasma are represented as closed data points. The dotted horizontal line indicates the PCR assay limit of detection, with undetectable events plotted as 30 copies/mL. (g) Plasma viral loads following ATI were averaged by ICB and are shown with the mean as the solid line in bold and the s.e.m. represented by the color-matched shaded region. (h) The attenuation in set-point plasma viremia was analyzed between chronic infection (d52 p.i.) and following rebound (d170 post-ATI, p.A.). Population sizes are as follows for all viral load and rebound delay analyses: controls (n=6), black; αCTLA-4 (n=6), blue; αPD-1 (n=6), pink; combined blockade (n=7), red; and BsAb (n=8), purple. (i) By 12 replicate reaction RT-qPCR, the number of cell-associated SIV-RNA copies per 106 cells was quantified in axillary LN at necropsy: controls (n=6), αCTLA-4 (n=6), αPD-1 (n=5), combined blockade (n=6), and BsAb (n=7). Averaged data are presented as the mean ± s.e.m., and were analyzed with a two-sided (g) two-way ANOVA with Dunnett’s correction for multiple comparisons or (a,h,i) a Mann-Whitney U test and/or Wilcoxon ranked sign test. Color-coded statistical asterisks indicate significance between control animals and the treatment group of that color. *, p<0.05; **, p<0.01; ***, p<0.001; ****, p<0.0001.

Extended Data Fig. 10 Phylogenetic trees of gp160 env sequences from plasma.

Single genome sequencing-derived gp160 env sequences from the plasma of 9 RMs are displayed in maximum-likelihood phylogenetic trees. Virus obtained from different experimental phases are as follows: blue, SIVmac239 inoculum; black, chronic infection prior to ART initiation (d52 p.i.); red, during on-ART ICB-mediated reactivation (d7-28 p.t.); and green, during acute rebound post-ATI. A scale bar indicating diversity for each phylogenetic tree is indicated below and lineage determinations are annotated for RUm16 (at right). Lineages were deemed distinct if they differed at 3 or more nucleotide positions over env.

Supplementary information

Supplementary Information

Supplementary Figs. 1–7 and Tables 1–3.

Reporting Summary

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Harper, J., Gordon, S., Chan, C.N. et al. CTLA-4 and PD-1 dual blockade induces SIV reactivation without control of rebound after antiretroviral therapy interruption. Nat Med 26, 519–528 (2020). https://doi.org/10.1038/s41591-020-0782-y

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41591-020-0782-y

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