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:

Systemic HIV and SIV latency reversal via non-canonical NF-κB signalling in vivo

Abstract

Long-lasting, latently infected resting CD4+ T cells are the greatest obstacle to obtaining a cure for HIV infection, as these cells can persist despite decades of treatment with antiretroviral therapy (ART). Estimates indicate that more than 70 years of continuous, fully suppressive ART are needed to eliminate the HIV reservoir1. Alternatively, induction of HIV from its latent state could accelerate the decrease in the reservoir, thus reducing the time to eradication. Previous attempts to reactivate latent HIV in preclinical animal models and in clinical trials have measured HIV induction in the peripheral blood with minimal focus on tissue reservoirs and have had limited effect2,3,4,5,6,7,8,9. Here we show that activation of the non-canonical NF-κB signalling pathway by AZD5582 results in the induction of HIV and SIV RNA expression in the blood and tissues of ART-suppressed bone-marrow–liver–thymus (BLT) humanized mice and rhesus macaques infected with HIV and SIV, respectively. Analysis of resting CD4+ T cells from tissues after AZD5582 treatment revealed increased SIV RNA expression in the lymph nodes of macaques and robust induction of HIV in almost all tissues analysed in humanized mice, including the lymph nodes, thymus, bone marrow, liver and lung. This promising approach to latency reversal—in combination with appropriate tools for systemic clearance of persistent HIV infection—greatly increases opportunities for HIV eradication.

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: Efficient in vitro AZD5582 target engagement and induction of HIV transcription.
Fig. 2: AZD5582 induces HIV RNA expression in resting CD4+ T cells from tissues of HIV-infected ART-suppressed BLT mice.
Fig. 3: AZD5582 induces SIV RNA expression in the plasma and lymph nodes of ART-suppressed SIV-infected rhesus macaques.
Fig. 4: AZD5582 specifically activates the ncNF-κB pathway in SIV-infected ART-suppressed rhesus macaques without generalized T cell activation.

Similar content being viewed by others

Data availability

Source Data for Figs. 14, Extended Data Figs. 1, 2, 4, 610 and Supplementary Tables 46 are provided with the paper. Gene-expression data are available at the Gene Expression Omnibus (GEO) repository (accession number GSE141546 and GSE142774). Any other data are available from corresponding authors on reasonable request.

References

  1. 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).

    CAS  PubMed  Google Scholar 

  2. Archin, N. M. et al. Interval dosing with the HDAC inhibitor vorinostat effectively reverses HIV latency. J. Clin. Invest. 127, 3126–3135 (2017).

    PubMed  PubMed Central  Google Scholar 

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

    ADS  CAS  PubMed  PubMed Central  Google Scholar 

  4. Elliott, J. H. et al. Activation of HIV transcription with short-course vorinostat in HIV-infected patients on suppressive antiretroviral therapy. PLoS Pathog. 10, e1004473 (2014).

    PubMed  PubMed Central  Google Scholar 

  5. Gutiérrez, C. et al. Bryostatin-1 for latent virus reactivation in HIV-infected patients on antiretroviral therapy. AIDS 30, 1385–1392 (2016).

    PubMed  Google Scholar 

  6. Kulkosky, J. et al. Intensification and stimulation therapy for human immunodeficiency virus type 1 reservoirs in infected persons receiving virally suppressive highly active antiretroviral therapy. J. Infect. Dis. 186, 1403–1411 (2002).

    CAS  PubMed  Google Scholar 

  7. Prins, J. M. et al. Immuno-activation with anti-CD3 and recombinant human IL-2 in HIV-1-infected patients on potent antiretroviral therapy. AIDS 13, 2405–2410 (1999).

    CAS  PubMed  Google Scholar 

  8. Rasmussen, T. A. et al. Panobinostat, a histone deacetylase inhibitor, for latent-virus reactivation in HIV-infected patients on suppressive antiretroviral therapy: a phase 1/2, single group, clinical trial. Lancet HIV 1, e13–e21 (2014).

    PubMed  Google Scholar 

  9. Søgaard, O. S. et al. The depsipeptide romidepsin reverses HIV-1 latency in vivo. PLoS Pathog. 11, e1005142 (2015).

    PubMed  PubMed Central  Google Scholar 

  10. Ke, R., Conway, J. M., Margolis, D. M. & Perelson, A. S. Determinants of the efficacy of HIV latency-reversing agents and implications for drug and treatment design. JCI Insight 3, e123052 (2018).

    PubMed Central  Google Scholar 

  11. Sun, S. C. The noncanonical NF-κB pathway. Immunol. Rev. 246, 125–140 (2012).

    PubMed  PubMed Central  Google Scholar 

  12. Fulda, S. Molecular pathways: targeting death receptors and Smac mimetics. Clin. Cancer Res. 20, 3915–3920 (2014).

    ADS  CAS  PubMed  Google Scholar 

  13. Pache, L. et al. BIRC2/cIAP1 is a negative regulator of HIV-1 transcription and can be targeted by Smac mimetics to promote reversal of viral latency. Cell Host Microbe 18, 345–353 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  14. Hennessy, E. J. et al. Discovery of a novel class of dimeric Smac mimetics as potent IAP antagonists resulting in a clinical candidate for the treatment of cancer (AZD5582). J. Med. Chem. 56, 9897–9919 (2013).

    CAS  PubMed  Google Scholar 

  15. Honeycutt, J. B. et al. T cells establish and maintain CNS viral infection in HIV-infected humanized mice. J. Clin. Invest. 128, 2862–2876 (2018).

    PubMed  PubMed Central  Google Scholar 

  16. Kessing, C. F. et al. In vivo suppression of HIV rebound by didehydro-cortistatin A, a “block-and-lock” strategy for HIV-1 treatment. Cell Reports 21, 600–611 (2017).

    CAS  PubMed  Google Scholar 

  17. Tsai, P. et al. In vivo analysis of the effect of panobinostat on cell-associated HIV RNA and DNA levels and latent HIV infection. Retrovirology 13, 36 (2016).

    PubMed  PubMed Central  Google Scholar 

  18. Melkus, M. W. et al. Humanized mice mount specific adaptive and innate immune responses to EBV and TSST-1. Nat. Med. 12, 1316–1322 (2006).

    CAS  PubMed  Google Scholar 

  19. Choudhary, S. K. et al. Latent HIV-1 infection of resting CD4+ T cells in the humanized Rag2−/− γc −/− mouse. J. Virol. 86, 114–120 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  20. Denton, P. W. et al. Generation of HIV latency in humanized BLT mice. J. Virol. 86, 630–634 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  21. Wahl, A. et al. Precision mouse models with expanded tropism for human pathogens. Nat. Biotechnol. 37, 1163–1173 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  22. Mavigner, M. et al. Simian immunodeficiency virus persistence in cellular and anatomic reservoirs in antiretroviral therapy-suppressed infant rhesus macaques. J. Virol. 92, e00562-18 (2018).

  23. Mavigner, M. et al. Pharmacological modulation of the Wnt/β-catenin pathway inhibits proliferation and promotes differentiation of long-lived memory CD4 T cells in antiretroviral therapy-suppressed simian immunodeficiency virus-infected macaques. J. Virol. 94, e01094-19 (2019)

  24. Abrahams, M. R. et al. The replication-competent HIV-1 latent reservoir is primarily established near the time of therapy initiation. Sci. Transl. Med. 11, eaaw5589 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  25. Anderson, E. M. & Maldarelli, F. The role of integration and clonal expansion in HIV infection: live long and prosper. Retrovirology 15, 71 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  26. Ferris, A. L. et al. Clonal expansion of SIV-infected cells in macaques on antiretroviral therapy is similar to that of HIV-infected cells in humans. PLoS Pathog. 15, e1007869 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  27. Kuo, H. H. & Lichterfeld, M. Recent progress in understanding HIV reservoirs. Curr. Opin. HIV AIDS 13, 137–142 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  28. Clutton, G. T. & Jones, R. B. Diverse impacts of HIV latency-reversing agents on CD8+ T-cell function: implications for HIV cure. Front. Immunol. 9, 1452 (2018).

    PubMed  PubMed Central  Google Scholar 

  29. Gupta, R. K. et al. HIV-1 remission following CCR5Δ32/Δ32 haematopoietic stem-cell transplantation. Nature 568, 244–248 (2019).

    ADS  CAS  PubMed  PubMed Central  Google Scholar 

  30. Hütter, G. et al. Long-term control of HIV by CCR5 Delta32/Delta32 stem-cell transplantation. N. Engl. J. Med. 360, 692–698 (2009).

    PubMed  Google Scholar 

  31. Nixon, C, C., Mavigner, M., Silvestri G. & Garcia, J. V. In vivo models of human immunodeficiency virus persistence and cure strategies. J. Infect. Dis. 215, S142–S151 (2017).

  32. 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).

    CAS  PubMed  PubMed Central  Google Scholar 

  33. Keedy, K. S. et al. A limited group of class I histone deacetylases acts to repress human immunodeficiency virus type 1 expression. J. Virol. 83, 4749–4756 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  34. Trumble, I. M. et al. SLDAssay: a software package and web tool for analyzing limiting dilution assays. J. Immunol. Methods 450, 10–16 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  35. Dobin, A. et al. STAR: ultrafast universal RNA-seq aligner. Bioinformatics 29, 15–21 (2013).

    CAS  PubMed  Google Scholar 

  36. Patro, R., Duggal, G., Love, M. I., Irizarry, R. A. & Kingsford, C. Salmon provides fast and bias-aware quantification of transcript expression. Nat. Methods 14, 417–419 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  37. Love, M. I., Huber, W. & Anders, S. Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2. Genome Biol. 15, 550 (2014).

    PubMed  PubMed Central  Google Scholar 

  38. Benjamini, Y. & Hochberg, Y. Controlling the false discovery rate: a practical and powerful approach to multiple testing. J. R. Stat. Soc. B 57, 289–300 (1995).

    MathSciNet  MATH  Google Scholar 

  39. Subramanian, A. et al. Gene set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profiles. Proc. Natl Acad. Sci. USA 102, 15545–15550 (2005).

    ADS  CAS  PubMed  PubMed Central  Google Scholar 

  40. Denton, P. W. et al. Antiretroviral pre-exposure prophylaxis prevents vaginal transmission of HIV-1 in humanized BLT mice. PLoS Med. 5, e16 (2008).

    PubMed  PubMed Central  Google Scholar 

  41. Denton, P. W. et al. One percent tenofovir applied topically to humanized BLT mice and used according to the CAPRISA 004 experimental design demonstrates partial protection from vaginal HIV infection, validating the BLT model for evaluation of new microbicide candidates. J. Virol. 85, 7582–7593 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  42. Reed, L. J. & Muench, H. A simple method of estimating fifty per cent endpoints. Am. J. Epidemiol. 27, 493–497 (1938).

    Google Scholar 

  43. Palesch, D. et al. Short-term pegylated interferon α2a treatment does not significantly reduce the viral reservoir of simian immunodeficiency virus-infected, antiretroviral therapy-treated rhesus macaques. J. Virol. 92, e00279-18 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

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

    ADS  CAS  PubMed  Google Scholar 

  45. 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).

    ADS  CAS  PubMed  PubMed Central  Google Scholar 

  46. Krisko, J. F., Martinez-Torres, F., Foster, J. L. & Garcia, J. V. HIV restriction by APOBEC3 in humanized mice. PLoS Pathog. 9, e1003242 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  47. Cartwright, E. K. et al. CD8+ lymphocytes are required for maintaining viral suppression in SIV-infected macaques treated with short-term antiretroviral therapy. Immunity 45, 656–668 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  48. Rosenbloom, D. I. S., Hill, A. L., Laskey, S. B. & Siliciano, R. F. Re-evaluating evolution in the HIV reservoir. Nature 551, E6–E9 (2017).

    ADS  CAS  PubMed  PubMed Central  Google Scholar 

  49. Zimin, A. V. et al. A new rhesus macaque assembly and annotation for next-generation sequencing analyses. Biol. Direct 9, 20 (2014).

    PubMed  PubMed Central  Google Scholar 

  50. Huang, W., Sherman, B. T. & Lempicki, R. A. Systematic and integrative analysis of large gene lists using DAVID bioinformatics resources. Nat. Protocols 4, 44–57 (2009).

    CAS  Google Scholar 

  51. Li, J. et al. Isolation and transcriptome analyses of human erythroid progenitors: BFU-E and CFU-E. Blood 124, 3636–3645 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  52. Roederer, M., Nozzi, J. L. & Nason, M. C. SPICE: Exploration and analysis of post cytometric complex multivariate datasets. Cytometry 79A, 167–174 (2014).

    Google Scholar 

Download references

Acknowledgements

We thank Garcia and Chahroudi laboratory members, the Animal Histopathology & Laboratory Medicine Core at the University of North Carolina-Chapel Hill (UNC-CH), which is supported in part by an NCI Center Core Support Grant (5P30CA016086-41) to the UNC Lineberger Comprehensive Cancer Center and technicians from the Department of Comparative Medicine at UNC-CH; the HIV/STD Laboratory Core and the Clinical Pharmacology and Analytical Chemistry Core of the UNC Center for AIDS Research (CFAR) (P30 AI050410); D. Hazuda, B. Howell and S. Barrett (Merck & Co.) for assistance with ART-containing chow; Yerkes Animal and Research Resources; the Children’s Healthcare of Atlanta and Emory University Pediatric Flow Cytometry Core, Emory CFAR Translational Virology and Reservoir Cores, the Quantitative Molecular Diagnostics Core of the AIDS and Cancer Virus Program, Frederick National Laboratory, as well as GSK for tenofovir disoproxil fumarate, emtricitabine and dolutegravir. This work was supported by the National Institutes of Allergy and Infectious Diseases (NIAID)(AI123010, AI096113, AI111899, AI117851 and P30 AI050410), and Mental Health (NIMH) (MH108179). This work was supported by the Emory Consortium for Innovative AIDS Research in Nonhuman Primates (UM1 AI124436), amfAR (109353-59-RGRL), the Yerkes National Primate Research Center (P51 OD011132) and the Translational Virology and Reservoir Cores of the Center for AIDS Research at Emory University (P30 AI050409). Research was also supported by Qura Therapeutics and by CARE, a Martin Delaney Collaboratory (1UM1AI126619-01) of the NIAID, NINDS, NIDA and NIMH. By the Natural Science Foundation of Guangdong Province, China (2016A030310108), UNC-South China STD Research Training Center (1D43TW009532) and Chinese National Key Technologies R&D Program for the 13th Five-year Plan (2017ZX10202101003). Federal funds were used for this research from the National Cancer Institute, NIH (contracts HHSN261200800001E and 75N91019D0002). 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.V.G., A.C., G.S., R.M.D., A.W. and D.M.M. conceived and designed the studies. G.C.S., D.M.I., S.R.W., S.D.F., C.G., E.P.B. and R.G.F. assessed in vitro AZD5582 activity. M.K., Z.W., J.H.B., D.F. and C.D.J. performed the in vitro transcriptomic study. D.F. and J.-P.R. contributed clinical specimens. C.C.N., P.T.H., C.D., N.J.S. and B.L. performed mouse experiments. R.A.S. and C.G.C. performed the qPCR analysis of HIV RNA levels in mouse samples. R.A.C. performed the immunohistochemical analysis. N.M.A. isolated human resting CD4+ T cells. C.C.N. analysed mouse data. M.M., A.D.B. and S.J. performed the monkey experiments. G.C.S., J.D., S.M. and R.M.D. conducted pharmacokinetic studies in NHP. C.M. and N.S. isolated resting CD4+ T cells from macaques. T.H.V. supervised the qPCR analyses of SIV RNA and DNA levels in monkey samples. M.M. and A.D.B. performed flow cytometry and analysed monkey data. G.K.T., A.A.U. and H.W. performed the macaque RNA-seq analyses and S.E.B. analysed these data. C.M.F. and B.F.K. performed env sequencing analyses. J.D.L. supervised the ultrasensitive plasma viral loads. G.C.S. and C.G. analysed AZD5582 target engagement in human, mouse and macaque samples and J.D. and R.M.D. analysed the data. C.G. performed the ELISPOTs. C.C.N., M.M., A.W., A.C. and J.V.G. wrote the manuscript with input from all authors.

Corresponding authors

Correspondence to Richard M. Dunham, Ann Chahroudi or J. Victor Garcia.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Peer review information Nature thanks Mathias Lichterfeld 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 Fig. 1 Short-duration exposure to AZD5582 activates the ncNF-κB pathway.

ad, Isolated total human CD4+ T cells treated with 100 nM AZD5582 and then either not washed (a) or washed three times with PBS 8 h (b), 4 h (c) or 1 h (d) after treatment. Whole-cell lysates were then analysed by immunoblot for components of the ncNF-κB pathway. e, Densitometry analysis of the ratio of p52 to p100 from the pulse-wash assay immunoblots. Points represent values for the densitometric ratio from one western blot, representative of several independent experiments. ae, Entire set representative of 1 experiment with 5 replicates of the 1-h wash condition conducted. f, Fold induction of ncNF-κB target gene expression in isolated CD4+ T cells from an uninfected donor treated with the indicated concentrations of AZD5582 and either washed after 1 h or not washed, and subsequently cultured for 24 h as measured by RT–qPCR. Points represent two technical replicates and lines represent the mean. The data presented are representative of three independent experiments. g, DMSO-normalized induction of luciferase activity from the Jurkat reporter model after exposure to AZD5582 (10, 100 or 1,000 nM) for 30 min (blue), 1 h (red), 2 h (green) or continued exposure (purple). Points represent three replicates in one assay run, representative of two independent experiments. Lines represent the mean of the three replicates. For gel source data, see Supplementary Fig. 1.

Source data

Extended Data Fig. 2 AZD5582 induces HIV RNA expression in resting CD4+ T cells from tissues of HIV-infected, ART-suppressed BLT mice.

HIV RNA levels in resting CD4+ T cells isolated from the bone marrow, lymph nodes, liver, lung and spleen of control (Cntrl, blue circles) or AZD5582-treated (AZD, red circles) mice (cells pooled from n = 6 mice per group for each tissue) were analysed in triplicate. Statistical significance was determined with a two-sided Student’s t-test. The mean fold increase in viral RNA levels in resting CD4+ T cells from tissues in the experiment shown in Fig. 2 was 12.1 (±3.7), whereas in this figure it was 8.4 (±6.0). These values were not statistically different (P = 0.4286, two-sided Mann–Whitney U-test). Data are mean ± s.e.m.

Source data

Extended Data Fig. 3 AZD5582 ex vivo target engagement.

a, Western blot analysis of p100, p52 and cIAP1 protein levels in cells isolated from the bone marrow, lymph node and spleen of BLT mice before and 20 h after ex vivo treatment with AZD5582. Loading control, β-actin. Representative of two experiments. b, cIAP expression in resting CD4+ T cells isolated from the thymus, spleen, lymph nodes, liver, lung and bone marrow. Loading control, β-actin. Representative of two experiments. c, cIAP expression in the thymic organoid of HIV-infected ART-suppressed BLT mice 48 h after the administration of vehicle control or AZD5582 (three control and one AZD5582-treated analysed). Positive cells, brown. Imaged at 4× and 40× magnifications; scale bars, 100 μm (4×) and 50 μm (40×). Boxes in the 4× images indicate regions corresponding to images at 40× magnification. For gel source data, see Supplementary Fig. 1.

Extended Data Fig. 4 Pharmacokinetic and pharmacodynamic assessment of AZD5582 in rhesus macaques.

a, AZD5582 (0.1 mg kg−1) was administered to healthy rhesus macaques (n = 3) by intravenous infusion. Plasma concentrations of AZD5582 (left y axis) are shown for the indicated time points. Flow cytometry was used to measure intracellular p100 levels, shown as the geometric mean fluorescence intensity (gMFI) in CD4+ T cells and plotted as the percentage of baseline of p100 gMFI (right y axis). b, Plasma concentrations of AZD5582 after one (dark red), three (red), six (pink) or ten (orange) doses in six SIV-infected ART-treated rhesus macaques (RM) and after one dose in three uninfected control rhesus macaques (grey). Individual values are shown as symbols. c, Western blot analyses of inactive p100 and active p52 forms of NF-κB2 in lymph-node mononuclear cells collected 48 h after the third or tenth dose of AZD5582 in SIV-infected ART-suppressed rhesus macaques (red; n = 3 for both the 3-dose and 10-dose groups) or at equivalent time points for placebo controls (blue; n = 2 for both the 3-dose and 10-dose groups). Immunoblots are shown in the top panels and densitometry analyses of the p52:p100 ratios are shown in the bottom panels. The line represents the median. d, Cryopreserved control rhesus macaque splenocytes were treated with the indicated concentrations of AZD5582 for 48 h, then p100/p52 levels were analysed by western blotting to measure engagement of the ncNF-κB pathway. e, DMSO-normalized densitometric p52:p100 ratio versus the AZD5582 concentration. For d, e, the experiments were performed in duplicate. f, Cryopreserved rhesus macaque splenocytes were exposed to DMSO alone (Untx), 100 nM AZD5582 washed off after 1 h and cultured for 47 h (Pulsed) or continuous 100 nM AZD5582 for 48 h (Cont.), after which the cells were studied by western blot for p100 and p52 levels. g, Densitometric p52:p100 ratio. For f, g, data represent a single experiment. For gel source data, see Supplementary Fig. 1.

Source data

Extended Data Fig. 5 Phylogenetic trees based on full env sequencing.

Plasma virus was sequenced from four time points per macaque (n = 5): near peak viraemia (2 weeks after infection; red), immediately before ART (8 weeks after infection; orange) and two time points of on-ART viraemia during AZD5582 treatment (green and blue). All sequences per macaque were phylogenetically analysed and the resulting phylogenetic trees are shown for each macaque. The horizontal bar indicates the genetic distance. nt, nucleotide.

Extended Data Fig. 6 SIV DNA levels in total CD4+ T cells and replication-competent reservoir size in ART-suppressed SIV-infected rhesus macaques.

a, Longitudinal assessment of cell-associated SIV DNA levels in total CD4+ T cells isolated from peripheral blood and lymph nodes of AZD5582-treated (n = 12, red) and control (n = 4, blue) ART-suppressed SIV-infected rhesus macaques. Grey shading represents the period of ART administration. b, Comparison of cell-associated SIV DNA levels in total CD4+ T cells isolated from the lymph nodes and peripheral blood of AZD5582-treated and control ART-suppressed SIV-infected rhesus macaques. Total CD4+ T cells were analysed from AZD5582-treated rhesus macaques 48 h after receiving 3 doses (lymph nodes and peripheral blood, n = 3) or 10 doses (lymph nodes and peripheral blood, n = 9) of AZD5582. Total CD4+ T cells were analysed from placebo-control rhesus macaques (lymph nodes and peripheral blood, n = 4) at equivalent time points. Open symbols indicate AZD5582-treated rhesus macaques with on-ART viraemia above 60 copies per ml of plasma. Statistical significance was determined using a two-sided Mann–Whitney U-test. c, Quantitative viral outgrowth assays were performed for AZD5582-treated rhesus macaques 48 h after receiving 3 doses (lymph nodes, n = 2; spleen, n = 3) or 10 doses (lymph nodes, n = 3; spleen, n = 2) of AZD5582. Quantitative viral outgrowth assays were performed from control rhesus macaques (lymph nodes and spleen, n = 4) at equivalent time points. Open symbols indicate AZD5582-treated rhesus macaques with on-ART viraemia. Statistical significance was determined with a two-sided Mann–Whitney U-test. Horizontal lines represent the median (b, c).

Source data

Extended Data Fig. 7 AZD5582-induced gene pathways and genes in SIV-infected ART-suppressed rhesus macaques.

a, DAVID analysis showing pathways significantly enriched in genes differentially expressed after AZD5582 treatment relative to baseline in CD4+ T cells isolated from lymph nodes (black bars) and peripheral blood (grey bars). n = 6 for both lymph nodes and peripheral blood; for each, n = 3 for 3 doses of AZD5582 and n = 3 for 10 doses of AZD5582. b, Leading edge genes from the ‘hallmark TNF signalling via NF-κB’ pathway from MSigDB. Genes were identified in the leading edge of CD4+ T cell samples from the lymph nodes before and after treatment with AZD5582 (shown in Fig. 4b). The contrast depicted is the fold change of each gene for each rhesus macaque’s post-treatment sample relative to the pre-treatment values for CD4+ T cells from the lymph nodes (top) and the peripheral blood (bottom). n = 6 for both lymph nodes and peripheral blood; for each, n = 3 for 3 doses of AZD5582 and n = 3 for 10 doses of AZD5582. c, Heat map of cNF-κB (top) and ncNF-κB (bottom) pathway gene expression in rhesus macaques with (left, n = 5) or without (right, n = 6) on-ART viraemia of >60 copies per ml plasma. One rhesus macaque without on-ART viraemia was excluded from this analysis because of technical issues (higher than expected unmapped and multi-mapped reads, and lower than expected unique identified reads compared to the means). Colour scale, log2-transformed fold changes of post-treatment compared with pre-treatment values.

Source data

Extended Data Fig. 8 Relationship between virological status before intervention and virological response to AZD5582 treatment in SIV-infected ART-suppressed rhesus macaques.

a, Individual representations of plasma SIV RNA levels measured by ultrasensitive assay (limit of detection 3 copies per ml of plasma) before and during AZD5582 treatment in SIV-infected ART-suppressed rhesus macaques (n = 12). ‘Baseline’ shows two or three plasma viral loads before AZD5582 treatment and ‘AZD5582’ shows three or four plasma viral loads during AZD5582 treatment. Open symbols indicate the five AZD5582-treated rhesus macaques with on-ART viraemia of >60 copies of SIV RNA per ml of plasma using the standard viral load assay (confirmed with ultrasensitive assay). The orange box highlights an additional 3 rhesus macaques who did not experience on-ART viraemia of >60 copies per ml of plasma but had ≥2 SIV RNA measurements above baseline values using the ultrasensitive quantification method. b, Comparison of the medians of plasma SIV RNA levels at baseline and during AZD5582 treatment measured by ultrasensitive assay for ART-suppressed SIV-infected rhesus macaques (n = 12). Statistical significance was determined with a Wilcoxon matched-pairs signed-rank test. c, Comparison of the levels of plasma SIV RNA at peak, plasma SIV RNA before ART initiation, SIV DNA in peripheral-blood CD4+ T cells before AZD5582 treatment (pre-LRA), SIV DNA in lymph-node CD4+ T cells pre-LRA, SIV RNA in peripheral blood CD4+ T cells pre-LRA and SIV RNA in lymph-node CD4+ T cells pre-LRA in rhesus macaques with increased plasma viral loads (PVL) by standard and/or ultrasensitive assay during AZD5582 treatment (increased plasma viral loads, n = 8) compared with rhesus macaques that did not demonstrate increased viral loads (stable plasma viral loads, n = 4). Statistical significance was determined using a two-sided Mann–Whitney U-test. Horizontal lines represent the median (a, c).

Source data

Extended Data Fig. 9 AZD5582 can be safely administered in SIV-infected ART-suppressed rhesus macaques.

ac, Longitudinal assessment of serum chemistries (a), complete blood counts (b) and weight (c) of SIV-infected ART-treated rhesus macaques treated with AZD5582 (red, n = 12) compared with controls (blue, n = 9). Grey shading represents the period of ART administration. AST, aspartate aminotransferase; ALT, alanine aminotransferase; BUN, blood urea nitrogen; GGT, γ-glutamyltransferase; HGB, haemoglobin; WBC, white blood cell.

Source data

Extended Data Fig. 10 Immunological effect of AZD5582 on SIV-infected ART-suppressed rhesus macaques.

a, b, Longitudinal assessment of the count (a) and the frequency and viability (b) of CD4+ T cells in SIV-infected ART-treated rhesus macaques during the period of AZD5582 treatment (n = 12). Lines represent median values. c, d, CD4+ T cell naive and memory subset frequencies (c) and their expression of Ki-67 (d) in SIV-infected ART-treated rhesus macaques during the period of AZD5582 treatment (n = 12). Lines represent median values. e, SIV gag-specific (left) and SIV env-specific (right) CD8+IFNγ+ T cell responses in SIV-infected, ART-suppressed, AZD5582-treated (red, n = 6) and control (blue, n = 4) rhesus macaques. SFU, spot-forming units. f, Pie charts depicting the ability of memory CD8+ T cells isolated from SIV-infected ART-suppressed control rhesus macaques (n = 5) to produce IFNγ, IL-2 and/or TNF in response to stimulation with PMA and ionomycin in the absence (left) or presence (right) of AZD5582 pre-treatment. g, Memory CD8+ T cell proliferative response to stimulation with PMA and ionomycin with or without AZD5582 pre-treatment. Top, representative flow cytometry dot plots gated on memory CD8+ T cells. Bottom, comparison between divided cells 5 days after stimulation in each group (n = 5 for each). Statistical significance was determined with a Wilcoxon matched-pairs signed-rank test. Horizontal lines represent the median. h, Longitudinal assessment of plasma levels of IP-10, MIP-1β, MCP-1, IL-6 and IL-10 by multiplex assay in SIV-infected ART-suppressed rhesus macaques treated with AZD5582 (red, n = 6) or control rhesus macaques (blue, n = 4). An additional four analytes (IFNγ, IL-8, IL-1β and IL-2) were undetectable in all macaques. ae, h, Grey shading represents the period of ART administration. Dashed lines represent AZD5582 or placebo infusions. LLOD, lower limit of detection.

Source data

Supplementary information

Supplementary Information

This file contains Supplementary Figures 1-2 and Supplementary Tables 1-7.

Reporting Summary

Supplementary Table 8

Gene expression in AZD5582-treated on-ART viremic and on-ART non-viremic RM.

Supplementary Data

This zipped file contains source data for Supplementary Tables 4-6.

Source data

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Nixon, C.C., Mavigner, M., Sampey, G.C. et al. Systemic HIV and SIV latency reversal via non-canonical NF-κB signalling in vivo. Nature 578, 160–165 (2020). https://doi.org/10.1038/s41586-020-1951-3

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41586-020-1951-3

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