Administration of vorinostat disrupts HIV-1 latency in patients on antiretroviral therapy

N M Archin, A L Liberty, A D Kashuba, S K Choudhary, J D Kuruc, A M Crooks, D C Parker, E M Anderson, M F Kearney, M C Strain, D D Richman, M G Hudgens, R J Bosch, J M Coffin, J J Eron, D J Hazuda, D M Margolis, N M Archin, A L Liberty, A D Kashuba, S K Choudhary, J D Kuruc, A M Crooks, D C Parker, E M Anderson, M F Kearney, M C Strain, D D Richman, M G Hudgens, R J Bosch, J M Coffin, J J Eron, D J Hazuda, D M Margolis

Abstract

Despite antiretroviral therapy, proviral latency of human immunodeficiency virus type 1 (HIV-1) remains a principal obstacle to curing the infection. Inducing the expression of latent genomes within resting CD4(+) T cells is the primary strategy to clear this reservoir. Although histone deacetylase inhibitors such as suberoylanilide hydroxamic acid (also known as vorinostat, VOR) can disrupt HIV-1 latency in vitro, the utility of this approach has never been directly proven in a translational clinical study of HIV-infected patients. Here we isolated the circulating resting CD4(+) T cells of patients in whom viraemia was fully suppressed by antiretroviral therapy, and directly studied the effect of VOR on this latent reservoir. In each of eight patients, a single dose of VOR increased both biomarkers of cellular acetylation, and simultaneously induced an increase in HIV RNA expression in resting CD4(+) cells (mean increase, 4.8-fold). This demonstrates that a molecular mechanism known to enforce HIV latency can be therapeutically targeted in humans, provides proof-of-concept for histone deacetylase inhibitors as a therapeutic class, and defines a precise approach to test novel strategies to attack and eradicate latent HIV infection directly.

Trial registration: ClinicalTrials.gov NCT01319383.

Figures

Figure 1. The relative HIV-1 RNA copy…
Figure 1. The relative HIV-1 RNA copy number in resting CD4+ T cells of 13 ART-treated HIV+ patients with plasma HIV RNA BDL
Values are calculated by cycle number, and limit of quantitation of cell-associated RNA is 10 copies. Cells were cultured alone (untreated), with VOR 335 nM (VOR), or activated with 3 µg/ml PHA and 60 U/ml IL2 for 6 hours. Data from 16 patients (a), and those who later received in vivo dosing [1 (Δ), 2 (□), 3 (◊), 4 (X), 5 (▽), 6 (❍), 7 (+), and 8 (⨀)] are shown in detail (b, mean and s.d.).
Figure 1. The relative HIV-1 RNA copy…
Figure 1. The relative HIV-1 RNA copy number in resting CD4+ T cells of 13 ART-treated HIV+ patients with plasma HIV RNA BDL
Values are calculated by cycle number, and limit of quantitation of cell-associated RNA is 10 copies. Cells were cultured alone (untreated), with VOR 335 nM (VOR), or activated with 3 µg/ml PHA and 60 U/ml IL2 for 6 hours. Data from 16 patients (a), and those who later received in vivo dosing [1 (Δ), 2 (□), 3 (◊), 4 (X), 5 (▽), 6 (❍), 7 (+), and 8 (⨀)] are shown in detail (b, mean and s.d.).
Figure 2. VOR exposure and histone acetylation
Figure 2. VOR exposure and histone acetylation
Median VOR plasma concentrations in patients 1–8 after a single 400 mg oral dose (circles with range shown) is shown in comparison to mean total cellular acetylated histone H3 in PBMCs (diamonds with s.d. shown), and relative levels of acetylated histone H3 at the human p21 gene promoter in resting CD4+ T cells (bars, mean and s.d.). A significant increase (†) in cellular acetylated histone H3 is seen at 8 hours, p

Figure 3. VOR upregulates HIV RNA expression

Figure 3. VOR upregulates HIV RNA expression

The relative HIV-1 RNA copy number (mean, s.d.)…

Figure 3. VOR upregulates HIV RNA expression
The relative HIV-1 RNA copy number (mean, s.d.) measured in the resting CD4+ T cells of eight HIV+ patients with plasma HIV RNA BDL is shown on background ART and on ART following a single 400 mg oral dose of VOR. For each subject, the differences are significant (p
Comment in
  • HIV: Shock and kill.
    Deeks SG. Deeks SG. Nature. 2012 Jul 25;487(7408):439-40. doi: 10.1038/487439a. Nature. 2012. PMID: 22836995 No abstract available.
  • Viral infection: Coaxing HIV out of hiding.
    Jermy A. Jermy A. Nat Rev Microbiol. 2012 Sep;10(9):596-7. doi: 10.1038/nrmicro2861. Epub 2012 Aug 13. Nat Rev Microbiol. 2012. PMID: 22886238 No abstract available.
Similar articles
Cited by
References
    1. Blankson JN, Persaud D, Siliciano RF. The challenge of viral reservoirs in HIV-1 infection. Annu Rev Med. 2002;53:557. - PubMed
    1. Margolis DM. Histone deacetylase inhibitors and HIV latency. Curr Opin HIV AIDS. 2011;6:25. - PMC - PubMed
    1. Archin NM, et al. Expression of Latent Human Immunodeficiency Type-1 is Induced by Novel and Selective Histone Deacetylase Inhibitors. AIDS. 2009;23:1799. - PMC - PubMed
    1. Archin NM, et al. Expression of Latent HIV Induced by the Potent HDAC Inhibitor Suberoylanilide Hydroxamic Acid. AIDS Res Hum Retroviruses. 2009;25:207. - PMC - PubMed
    1. Contreras X, et al. Suberoylanilide hydroxamic acid reactivates HIV from latently infected cells. J Biol Chem. 2009;284:6782. - PMC - PubMed
Show all 25 references
Publication types
MeSH terms
Associated data
[x]
Cite
Copy Download .nbib
Format: AMA APA MLA NLM
Figure 3. VOR upregulates HIV RNA expression
Figure 3. VOR upregulates HIV RNA expression
The relative HIV-1 RNA copy number (mean, s.d.) measured in the resting CD4+ T cells of eight HIV+ patients with plasma HIV RNA BDL is shown on background ART and on ART following a single 400 mg oral dose of VOR. For each subject, the differences are significant (p

References

    1. Blankson JN, Persaud D, Siliciano RF. The challenge of viral reservoirs in HIV-1 infection. Annu Rev Med. 2002;53:557.
    1. Margolis DM. Histone deacetylase inhibitors and HIV latency. Curr Opin HIV AIDS. 2011;6:25.
    1. Archin NM, et al. Expression of Latent Human Immunodeficiency Type-1 is Induced by Novel and Selective Histone Deacetylase Inhibitors. AIDS. 2009;23:1799.
    1. Archin NM, et al. Expression of Latent HIV Induced by the Potent HDAC Inhibitor Suberoylanilide Hydroxamic Acid. AIDS Res Hum Retroviruses. 2009;25:207.
    1. Contreras X, et al. Suberoylanilide hydroxamic acid reactivates HIV from latently infected cells. J Biol Chem. 2009;284:6782.
    1. Lehrman G, et al. Depletion of latent HIV infection in vivo: a proof of concept study. Lancet. 2005;366:549.
    1. Archin NM, et al. Valproic acid without intensified antiviral therapy has limited impact on persistent HIV infection of resting CD4+ T cells. AIDS. 2008;22:1131.
    1. Siliciano JD, et al. Stability of the latent reservoir for HIV-1 in patients receiving valproic acid. J Infect Dis. 2007;195:833.
    1. Sagot-Lerolle N, et al. Prolonged valproic acid treatment does not reduce the size of latent HIV reservoir. AIDS. 2008;22:1125.
    1. Archin NM, et al. Antiretroviral intensification and valproic acid have limited effect on residual HIV-1 viremia or resting CD4+ cell infection. PLoS One. 2010;5:e9390.
    1. Keedy KS, et al. A limited group of class I histone deacetylases act to repress human immunodeficiency virus type-1 expression. J Virol. 2009;83:4749.
    1. Edelstein LC, Micheva-Viteva S, Phelan BD, Dougherty JP. Activation of latent HIV type 1 gene expression by suberoylanilide hydroxamic acid (SAHA), an HDAC inhibitor approved for use to treat cutaneous T cell lymphoma. AIDS Res Hum Retroviruses. 2009;25:883.
    1. Gui CY, et al. Histone deacetylase (HDAC) inhibitor activation of p21WAF1 involves changes in promoter-associated proteins, including HDAC1. Proc Natl Acad Sci U S A. 2004;101:1241.
    1. Du L, Musson DG, Wang AQ. Stability studies of vorinostat and its two metabolites in human plasma, serum and urine. Rapid Commun Mass Spectrom. 2005;19:1779.
    1. Rubin EH, et al. A study to determine the effects of food and multiple dosing on the pharmacokinetics of vorinostat given orally to patients with advanced cancer. Clin Cancer Res. 2006;12:7039.
    1. Hermankova M, et al. Analysis of human immunodeficiency virus type 1 gene expression in latently infected resting CD4+ T lymphocytes in vivo. J Virol. 2003;77:7383.
    1. Palmer S, et al. New real-time reverse transcriptase-initiated PCR assay with single-copy sensitivity for human immunodeficiency virus type 1 RNA in plasma. J Clin Microbiol. 2003;41:4531.
    1. Lee JH, Choy ML, Ngo L, Foster SS, Marks PA. Histone deacetylase inhibitor induces DNA damage, which normal but not transformed cells can repair. Proc Natl Acad Sci U S A. 2010;107:14639.
    1. Lee JH, Choy ML, Ngo L, Venta-Perez G, Marks PA. Mechanisms of Resistance to Histone Deacetylase Inhibitors: Role of Chk1. Proc Natl Acad Sci U S A. 2011;108:19629.
    1. Marks PA. personal communication.
    1. Israel-Ballard K, et al. TaqMan RT-PCR and VERSANT HIV-1 RNA 3.0 (bDNA) assay Quantification of HIV-1 RNA viral load in breast milk. J Clin Virol. 2005;34:253.
    1. Robinson LH, Gale CV, Kleim JP. Inclusion of full-length human immunodeficiency virus type 1 (HIV-1) gag sequences in viral recombinants applied to drug susceptibility phenotyping. J Virol Methods. 2002;104:147.
    1. Radonić A, et al. Guideline to reference gene selection for quantitative real-time PCR. Biochem Biophys Res Commun. 2004;313:856.
    1. May RC, et al. Change-Point Models to Estimate the Limit of Detection. University of North Carolina at Chapel Hill Biostatistics Technical Report Series. Working Paper 26. 2012 Mar; .
    1. Tonelli R, et al. G1 cell-cycle arrest and apoptosis by histone deacetylase inhibition in MLL-AF9 acute myeloid leukemia cells is p21 dependent and MLL-AF9 independent. Leukemia. 2006;20:1307.

Source: PubMed

3
Tilaa