#PAGE_PARAMS# #ADS_HEAD_SCRIPTS# #MICRODATA#

Activation of HIV Transcription with Short-Course Vorinostat in HIV-Infected Patients on Suppressive Antiretroviral Therapy


The major barrier to curing HIV is the long term persistence of latently infected resting memory T-cells in HIV-infected patients on antiretroviral therapy (ART). One strategy being pursued to eliminate latently infected cells is to activate HIV production from latently infected cells with the aim of killing latently infected cells via virus induced cell death or stimulation of an HIV-specific immune response. Histone deacetylases (HDACs) are important in maintaining HIV latency. Vorinostat, an inhibitor of HDACs (HDACi) licensed for the treatment of some malignancies, has been shown in laboratory studies and a clinical study of selected individuals to disrupt HIV latency. We examined the ability of standard dose vorinostat given daily for 14 days to activate latent HIV infection in unselected HIV-infected individuals on ART. The study showed evidence of activation of latent HIV infection in 18/20 (90%) of individuals and was safe and generally well tolerated. There were significant early changes in host gene expression, which persisted during and after the period of vorinostat. No changes were seen in immune activation or number of latently infected cells. Vorinostat was able to activate latent HIV infection in most individuals. Additional interventions will be needed to eliminate latent HIV infection.


Vyšlo v časopise: Activation of HIV Transcription with Short-Course Vorinostat in HIV-Infected Patients on Suppressive Antiretroviral Therapy. PLoS Pathog 10(11): e32767. doi:10.1371/journal.ppat.1004473
Kategorie: Research Article
prolekare.web.journal.doi_sk: https://doi.org/10.1371/journal.ppat.1004473

Souhrn

The major barrier to curing HIV is the long term persistence of latently infected resting memory T-cells in HIV-infected patients on antiretroviral therapy (ART). One strategy being pursued to eliminate latently infected cells is to activate HIV production from latently infected cells with the aim of killing latently infected cells via virus induced cell death or stimulation of an HIV-specific immune response. Histone deacetylases (HDACs) are important in maintaining HIV latency. Vorinostat, an inhibitor of HDACs (HDACi) licensed for the treatment of some malignancies, has been shown in laboratory studies and a clinical study of selected individuals to disrupt HIV latency. We examined the ability of standard dose vorinostat given daily for 14 days to activate latent HIV infection in unselected HIV-infected individuals on ART. The study showed evidence of activation of latent HIV infection in 18/20 (90%) of individuals and was safe and generally well tolerated. There were significant early changes in host gene expression, which persisted during and after the period of vorinostat. No changes were seen in immune activation or number of latently infected cells. Vorinostat was able to activate latent HIV infection in most individuals. Additional interventions will be needed to eliminate latent HIV infection.


Zdroje

1. FinziD, HermankovaM, PiersonT, CarruthLM, BuckC, et al. (1997) Identification of a reservoir for HIV-1 in patients on highly active antiretroviral therapy. Science 278: 1295–1300.

2. ChunTW, CarruthL, FinziD, ShenX, DiGiuseppeJA, et al. (1997) Quantification of latent tissue reservoirs and total body viral load in HIV-1 infection. Nature 387: 183–188.

3. SmithMZ, WightmanF, LewinSR (2012) HIV reservoirs and strategies for eradication. Curr HIV/AIDS Rep 9: 5–15.

4. YlisastiguiL, ArchinNM, LehrmanG, BoschRJ, MargolisDM (2004) Coaxing HIV-1 from resting CD4 T cells: histone deacetylase inhibition allows latent viral expression. AIDS 18: 1101–1108.

5. Shehu-XhilagaM, RhodesD, WightmanF, LiuHB, SolomonA, et al. (2009) The novel histone deacetylase inhibitors metacept-1 and metacept-3 potently increase HIV-1 transcription in latently infected cells. AIDS 23: 2047–2050.

6. SavarinoA, MaiA, NorelliS, El DakerS, ValenteS, et al. (2009) “Shock and kill” effects of class I-selective histone deacetylase inhibitors in combination with the glutathione synthesis inhibitor buthionine sulfoximine in cell line models for HIV-1 quiescence. Retrovirology 6: 52.

7. KeedyKS, ArchinNM, GatesAT, EspesethA, HazudaDJ, et al. (2009) A limited group of class I histone deacetylases acts to repress human immunodeficiency virus type 1 expression. J Virol 83: 4749–4756.

8. ArchinNM, KeedyKS, EspesethA, DangH, HazudaDJ, et al. (2009) Expression of latent human immunodeficiency type 1 is induced by novel and selective histone deacetylase inhibitors. AIDS 23: 1799–1806.

9. CilloAR, SobolewskiMD, BoschRJ, FyneE, PiatakMJr, et al. (2014) Quantification of HIV-1 latency reversal in resting CD4+ T cells from patients on suppressive antiretroviral therapy. Proc Natl Acad Sci U S A 111: 7078–7083.

10. SpinaCA, AndersonJ, ArchinNM, BosqueA, ChanJ, et al. (2013) An in-depth comparison of latent HIV-1 reactivation in multiple cell model systems and resting CD4+ T cells from aviremic patients. PLoS Pathog 9: e1003834.

11. WeiDG, ChiangV, FyneE, BalakrishnanM, BarnesT, et al. (2014) Histone deacetylase inhibitor romidepsin induces HIV expression in CD4 T cells from patients on suppressive antiretroviral therapy at concentrations achieved by clinical dosing. PLoS Pathog 10: e1004071.

12. BullenCK, LairdGM, DurandCM, SilicianoJD, SilicianoRF (2014) New ex vivo approaches distinguish effective and ineffective single agents for reversing HIV-1 latency in vivo. Nat Med 20: 425–429.

13. PrinceHM, BishtonMJ, HarrisonSJ (2009) Clinical studies of histone deacetylase inhibitors. Clin Cancer Res 15: 3958–3969.

14. RasmussenTA, Schmeltz SogaardO, BrinkmannC, WightmanF, LewinSR, et al. (2013) Comparison of HDAC inhibitors in clinical development: Effect on HIV production in latently infected cells and T-cell activation. Hum Vaccin Immunother 9: 993–1001.

15. BouchatS, GatotJS, KabeyaK, CardonaC, ColinL, et al. (2012) Histone methyltransferase inhibitors induce HIV-1 recovery in resting CD4(+) T cells from HIV-1-infected HAART-treated patients. AIDS 26: 1473–1482.

16. ArchinNM, LibertyAL, KashubaAD, ChoudharySK, KurucJD, et al. (2012) Administration of vorinostat disrupts HIV-1 latency in patients on antiretroviral therapy. Nature 487: 482–485.

17. BlazkovaJ, MurrayD, JustementJS, FunkEK, NelsonAK, et al. (2012) Paucity of HIV DNA methylation in latently infected, resting CD4+ T cells from infected individuals receiving antiretroviral therapy. J Virol 86: 5390–5392.

18. SøgaardO, GraversenM, LethS, BrinkmannC, KjærA-S, et al. (2014) The HDAC inhibitor romidepsin is safe and effectively reverses HIV-1 latency in vivo as measured by standard clinical assays. Abstract TUAA0106LB. 20th International AIDS Conference. Melbourne

19. ArchinNM, BatesonR, TripathyMK, CrooksAM, YangKH, et al. (2014) HIV-1 Expression Within Resting CD4+ T Cells After Multiple Doses of Vorinostat. J Infect Dis 210: 728–35.

20. PalmerS, MaldarelliF, WiegandA, BernsteinB, HannaGJ, et al. (2008) Low-level viremia persists for at least 7 years in patients on suppressive antiretroviral therapy. Proc Natl Acad Sci U S A 105: 3879–3884.

21. BhaskaraS, JacquesV, RuscheJR, OlsonEN, CairnsBR, et al. (2013) Histone deacetylases 1 and 2 maintain S-phase chromatin and DNA replication fork progression. Epigenetics Chromatin 6: 27.

22. SmithS, FoxJ, MejiaM, RuangpraditW, SaberiA, et al. (2014) Histone deacetylase inhibitors selectively target homology dependent DNA repair defective cells and elevate non-homologous endjoining activity. PLoS One 9: e87203.

23. DarwishH, ChoJM, LoignonM, Alaoui-JamaliMA (2007) Overexpression of SERTAD3, a putative oncogene located within the 19q13 amplicon, induces E2F activity and promotes tumor growth. Oncogene 26: 4319–4328.

24. AliM, HomRA, BlakesleeW, IkenouyeL, KutateladzeTG (2014) Diverse functions of PHD fingers of the MLL/KMT2 subfamily. Biochim Biophys Acta 1843: 366–371.

25. MusselmanCA, LalondeME, CoteJ, KutateladzeTG (2012) Perceiving the epigenetic landscape through histone readers. Nat Struct Mol Biol 19: 1218–1227.

26. RafatiH, ParraM, HakreS, MoshkinY, VerdinE, et al. (2011) Repressive LTR nucleosome positioning by the BAF complex is required for HIV latency. PLoS Biol 9: e1001206.

27. Van DuyneR, GuendelI, NarayananA, GreggE, ShafagatiN, et al. (2011) Varying Modulation of HIV-1 LTR Activity by BAF Complexes. Journal of Molecular Biology 411: 581–596.

28. TolstorukovMY, SansamCG, LuP, KoellhofferEC, HelmingKC, et al. (2013) Swi/Snf chromatin remodeling/tumor suppressor complex establishes nucleosome occupancy at target promoters. Proc Natl Acad Sci U S A 110: 10165–10170.

29. HakreS, ChavezL, ShirakawaK, VerdinE (2012) HIV latency: experimental systems and molecular models. FEMS Microbiol Rev 36: 706–716.

30. MarounM, DelelisO, CoadouG, BaderT, SegeralE, et al. (2006) Inhibition of early steps of HIV-1 replication by SNF5/Ini1. J Biol Chem 281: 22736–22743.

31. BartholomeeusenK, FujinagaK, XiangY, PeterlinBM (2013) Histone deacetylase inhibitors (HDACis) that release the positive transcription elongation factor b (P-TEFb) from its inhibitory complex also activate HIV transcription. J Biol Chem 288: 14400–14407.

32. LuH, LiZ, XueY, Schulze-GahmenU, JohnsonJR, et al. (2014) AFF1 is a ubiquitous P-TEFb partner to enable Tat extraction of P-TEFb from 7SK snRNP and formation of SECs for HIV transactivation. Proc Natl Acad Sci U S A 111: E15–24.

33. LitvakV, RamseySA, RustAG, ZakDE, KennedyKA, et al. (2009) Function of C/EBPdelta in a regulatory circuit that discriminates between transient and persistent TLR4-induced signals. Nat Immunol 10: 437–443.

34. LassenKG, RamyarKX, BaileyJR, ZhouY, SilicianoRF (2006) Nuclear retention of multiply spliced HIV-1 RNA in resting CD4+ T cells. PLoS Pathog 2: e68.

35. HuangJ, WangF, ArgyrisE, ChenK, LiangZ, et al. (2007) Cellular microRNAs contribute to HIV-1 latency in resting primary CD4+ T lymphocytes. Nat Med 13: 1241–1247.

36. SalehS, WightmanF, RamanayakeS, AlexanderM, KumarN, et al. (2011) Expression and reactivation of HIV in a chemokine induced model of HIV latency in primary resting CD4+ T cells. Retrovirology 8: 80.

37. LassenKG, BaileyJR, SilicianoRF (2004) Analysis of human immunodeficiency virus type 1 transcriptional elongation in resting CD4+ T cells in vivo. J Virol 78: 9105–9114.

38. TaoR, de ZoetenEF, OzkaynakE, ChenC, WangL, et al. (2007) Deacetylase inhibition promotes the generation and function of regulatory T cells. Nat Med 13: 1299–1307.

39. LiN, ZhaoD, KirschbaumM, ZhangC, LinCL, et al. (2008) HDAC inhibitor reduces cytokine storm and facilitates induction of chimerism that reverses lupus in anti-CD3 conditioning regimen. Proc Natl Acad Sci U S A 105: 4796–4801.

40. GojoI, JiemjitA, TrepelJB, SparreboomA, FiggWD, et al. (2007) Phase 1 and pharmacologic study of MS-275, a histone deacetylase inhibitor, in adults with refractory and relapsed acute leukemias. Blood 109: 2781–2790.

41. BoseP, DaiY, GrantS (2014) Histone deacetylase inhibitor (HDACI) mechanisms of action: Emerging insights. Pharmacol Ther 143: 323–336.

42. ReuseS, CalaoM, KabeyaK, GuiguenA, GatotJS, et al. (2009) Synergistic activation of HIV-1 expression by deacetylase inhibitors and prostratin: implications for treatment of latent infection. PLoS One 4: e6093.

43. BoehmD, CalvaneseV, DarRD, XingS, SchroederS, et al. (2013) BET bromodomain-targeting compounds reactivate HIV from latency via a Tat-independent mechanism. Cell Cycle 12: 452–462.

44. RasmussenT (2014) the CLEAR study group (2014) Panobinostat Induces HIV Transcription and Plasma Viremia in HIV Patients on Suppressive cART. Conference on Retroviruses and Opportunistic Infections. Boston

45. WightmanF, LuHK, SolomonAE, SalehS, HarmanAN, et al. (2013) Entinostat is a histone deacetylase inhibitor selective for class 1 histone deacetylases and activates HIV production from latently infected primary T cells. AIDS 27: 2853–62.

46. LewinSR, VesanenM, KostrikisL, HurleyA, DuranM, et al. (1999) Use of real-time PCR and molecular beacons to detect virus replication in human immunodeficiency virus type 1-infected individuals on prolonged effective antiretroviral therapy. Journal of Virology 73: 6099–6103.

47. StoltzfusCM, MadsenJM (2006) Role of viral splicing elements and cellular RNA binding proteins in regulation of HIV-1 alternative RNA splicing. Curr HIV Res 4: 43–55.

48. PasternakAO, JurriaansS, BakkerM, PrinsJM, BerkhoutB, et al. (2009) Cellular levels of HIV unspliced RNA from patients on combination antiretroviral therapy with undetectable plasma viremia predict the therapy outcome. PLoS ONE 4: e8490.

49. LewinSR, MurrayJM, SolomonA, WightmanF, CameronPU, et al. (2008) Virologic determinants of success after structured treatment interruptions of antiretrovirals in acute HIV-1 infection. J Acquir Immune Defic Syndr 47: 140–147.

50. ChomontN, El-FarM, AncutaP, TrautmannL, ProcopioFA, et al. (2009) HIV reservoir size and persistence are driven by T cell survival and homeostatic proliferation. Nat Med 15: 893–900.

51. YuklSA, GianellaS, SinclairE, EplingL, LiQ, et al. (2010) Differences in HIV burden and immune activation within the gut of HIV-positive patients receiving suppressive antiretroviral therapy. J Infect Dis 202: 1553–1561.

52. PasternakAO, AdemaKW, BakkerM, JurriaansS, BerkhoutB, et al. (2008) Highly sensitive methods based on seminested real-time reverse transcription-PCR for quantitation of human immunodeficiency virus type 1 unspliced and multiply spliced RNA and proviral DNA. J Clin Microbiol 46: 2206–2211.

53. HuY, SmythGK (2009) ELDA: extreme limiting dilution analysis for comparing depleted and enriched populations in stem cell and other assays. J Immunol Methods 347: 70–78.

54. HatanoH, JainV, HuntPW, LeeTH, SinclairE, et al. (2013) Cell-based measures of viral persistence are associated with immune activation and programmed cell death protein 1 (PD-1)-expressing CD4+ T cells. J Infect Dis 208: 50–56.

55. The R Development Core Team (2010) R: A Language and Environment for Statistical Computing: R Foundation for Statistical Computing.

56. Smyth GK (2005) Limma: linear models for microarray data. Bioinformatics and Computational Biology Solutions using R and Bioconductor. New York: Springer.

57. GentlemanRC, CareyVJ, BatesDM, BolstadB, DettlingM, et al. (2004) Bioconductor: open software development for computational biology and bioinformatics. Genome Biol 5: R80.

58. BenjaminiY, HochbergY (1995) Controlling the false discovery rate: a practical and powerful approach to multiple testing. Royal Statist Soc Series B (Methodological) 57.

59. SubramanianA, TamayoP, MoothaVK, MukherjeeS, EbertBL, et al. (2005) Gene set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profiles. Proc Natl Acad Sci U S A 102: 15545–15550.

60. NakayaHI, WrammertJ, LeeEK, RacioppiL, Marie-KunzeS, et al. (2011) Systems biology of vaccination for seasonal influenza in humans. Nat Immunol 12: 786–795.

61. SutradharB, RaoR (2001) On Marginal Quasi-Likelihood Inference in Generalized Linear Mixed Models. Journal of Multivariate Analysis 76: 1–34.

62. FeddagM, GramaI, MesbahM (2003) Generalised Estimating Equations (GEE) for Mixed Logistic Models. Communications in Statistics: Theory and Methods 32: 851–874.

Štítky
Hygiena a epidemiológia Infekčné lekárstvo Laboratórium

Článok vyšiel v časopise

PLOS Pathogens


2014 Číslo 11
Najčítanejšie tento týždeň
Najčítanejšie v tomto čísle
Kurzy

Zvýšte si kvalifikáciu online z pohodlia domova

Získaná hemofilie - Povědomí o nemoci a její diagnostika
nový kurz

Eozinofilní granulomatóza s polyangiitidou
Autori: doc. MUDr. Martina Doubková, Ph.D.

Všetky kurzy
Prihlásenie
Zabudnuté heslo

Zadajte e-mailovú adresu, s ktorou ste vytvárali účet. Budú Vám na ňu zasielané informácie k nastaveniu nového hesla.

Prihlásenie

Nemáte účet?  Registrujte sa

#ADS_BOTTOM_SCRIPTS#