Elsevier

The Lancet HIV

Volume 1, Issue 1, October 2014, Pages e13-e21
The Lancet HIV

Articles
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

https://doi.org/10.1016/S2352-3018(14)70014-1Get rights and content

Summary

Background

Activating the expression of latent virus is an approach that might form part of an HIV cure. We assessed the ability of the histone deacetylase inhibitor panobinostat to disrupt HIV-1 latency and the safety of this strategy.

Methods

In this phase 1/2 clinical trial, we included aviraemic adults with HIV treated at Aarhus University Hospital, Denmark. Participants received oral panobinostat (20 mg) three times per week every other week for 8 weeks while maintaining combination antiretroviral therapy. The primary outcome was change from baseline of cell-associated unspliced HIV RNA. Secondary endpoints were safety, plasma HIV RNA, total and integrated HIV DNA, infectious units per million CD4 T cells, and time to viral rebound during an optional analytical treatment interruption of antiretroviral therapy. This trial is registered with ClinicalTrial.gov, number NCT01680094.

Findings

We enrolled 15 patients. The level of cell-associated unspliced HIV RNA increased significantly at all timepoints when patients were taking panobinostat (p<0·0001). The median maximum increase in cell-associated unspliced HIV RNA during panobinostat treatment was 3·5-fold (range 2·1–14·4). Panobinostat induced plasma viraemia with an odds ratio of 10·5 (95% CI 2·2–50·3; p=0·0002) compared with baseline. We recorded a transient decrease in total HIV DNA, but no cohort-wide reduction in total HIV DNA, integrated HIV DNA, or infectious units per million. Nine patients participated in the analytical treatment interruption, median time to viral rebound was 17 days (range 14–56). Panobinostat was well tolerated. 45 adverse events were reported, but only 16 (all grade 1) were presumed related to panobinostat.

Interpretation

Panobinostat effectively disrupts HIV latency in vivo and is a promising candidate for future combination clinical trials aimed at HIV eradication. However, panobinostat did not reduce the number of latently infected cells and this approach may need to be combined with others to significantly affect the latent HIV reservoir.

Funding

The Danish Council for Strategic Research and Aarhus University.

Introduction

Combination antiretroviral therapy effectively suppresses replication of HIV but persisting virus fuels rebound viraemia when treatment ceases.1 Thus, lifelong adherence to antiretroviral therapy is necessary for disease control, raising concerns of long-term adverse effects and the financial burden of treatment. The development of treatments that cure infection, therefore, is a fundamental goal in HIV research.

The most significant obstacle to eradication of HIV infection is the presence of replication-competent provirus in long-lived resting CD4 T cells.2, 3, 4 In the transcriptionally silent resting state, viral proteins are not expressed and, therefore, the infected status of resting cells remains invisible to the immune system and unresponsive to antiretroviral therapy. This reversibly non-productive state of infection is referred to as HIV latency.5 Drugs aimed at activating HIV from latency should ideally induce viral transcription, lead to viral protein production, and release of viral particles—essential prerequisites for immune-mediated elimination.6 However, the effect of this approach is subject to ongoing controversy with an ex-vivo study suggesting that none of the leading candidate drugs can alone disrupt the latent HIV reservoir.7 Clinical trials are the only experimental approaches to investigate whether drugs aimed at disrupting HIV latency in vivo are effective or not.

One of several mechanisms controlling HIV latency is the activity of histone deacetylases, which repress proviral transcription by promoting histone deacetylation.8, 9 Several studies have shown that histone deacetylase inhibitors disrupt HIV latency in vitro.10, 11, 12 Additionally, administration of a single dose of vorinostat—a histone deacetylase inhibitor approved by the US Food and Drug Administration—led to significant increases in cell-associated HIV RNA,13 as did multiple doses of vorinostat over 14 days.14 By contrast, three doses per week of vorinostat over 8 weeks only increased amounts of cell-associated HIV RNA above baseline for three of five participants.15 Increased plasma viraemia or reduction of the latent HIV reservoir have not been shown in studies of vorinostat. These findings have led researchers to question the ability of histone deacetylase inhibitors to disrupt latency to the extent that HIV proteins are expressed on the surface of infected cells, thus enabling immune-mediated elimination to occur.7, 16, 17

Panobinostat is a highly potent hydroxamic acid pan-histone deacetylase inhibitor in clinical development for the treatment of multiple myeloma.18 Therapeutic concentrations of panobinostat induce viral production in latently infected cells in vitro.19 On the basis of these data, we designed an interventional study to assess whether 8 weeks of cyclic panobinostat treatment added to antiretroviral therapy would increase HIV transcription, increase plasma viraemia, and affect the latent HIV reservoir.

Section snippets

Study design and participants

We did a single group, phase 1/2 trial at Aarhus University Hospital, Denmark, between Oct 1, 2012 and Jan 16, 2014. We enrolled HIV-infected adults taking antiretroviral therapy with virological suppression (<50 copies per mL, at least two measurements per year) for at least 2 years and CD4 counts above 500 cells per μL. Exclusion criteria included co-infection with hepatitis B or C viruses, clinically significant cardiac disease including QTc prolongation, and current use of a protease

Results

We enrolled 15 patients and all completed full panobinostat dosing (table 1). The amount of cell-associated unspliced HIV RNA increased during panobinostat treatment (p<0·0001; repeated measurement ANOVA incorporating all data from baseline and on panobinostat); with significant increases at all assayed timepoints compared with baseline (figure 1A, appendix p 3). Amounts of cell-associated unspliced HIV RNA increased rapidly, with a mean increase of 2·4-fold (95% CI 1·8–3·3; p<0·0001) measured

Discussion

8 weeks of cyclic treatment with panobinostat was safe, well tolerated, and effectively increased HIV transcription in patients taking antiretroviral therapy. Furthermore, panobinostat-induced HIV transcription was temporally associated with increased detection of plasma HIV RNA. Viral rebound followed treatment interruption, although the time to rebound varied between individuals. Together, these results provide evidence that panobinostat can effectively activate HIV from latency in vivo and

References (33)

  • C Van Lint et al.

    Transcriptional activation and chromatin remodeling of the HIV-1 promoter in response to histone acetylation

    Embo J

    (1996)
  • SA Williams et al.

    NF-kappaB p50 promotes HIV latency through HDAC recruitment and repression of transcriptional initiation

    Embo J

    (2006)
  • NM Archin et al.

    Expression of latent HIV induced by the potent HDAC inhibitor suberoylanilide hydroxamic acid

    AIDS Res Hum Retroviruses

    (2009)
  • F Wightman et al.

    Entinostat is a histone deacetylase inhibitor selective for class 1 histone deacetylases and activates HIV production from latently infected primary T cells

    AIDS

    (2013)
  • L Shan et al.

    Unique characteristics of histone deacetylase inhibitors in reactivation of latent HIV-1 in Bcl-2-transduced primary resting CD4+ T cells

    J Antimicrob Chemother

    (2014)
  • NM Archin et al.

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

    Nature

    (2012)
  • Cited by (496)

    • Strategies for HIV-1 elimination

      2024, HIV-Associated Neurocognitive Disorders
    View all citing articles on Scopus
    View full text