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.

  • Review Article
  • Published:

Drug Insight: histone deacetylase inhibitors—development of the new targeted anticancer agent suberoylanilide hydroxamic acid

Abstract

This review focuses on the discovery and development of the histone deacetylase (HDAC) inhibitor, suberoylanilide hydroxamic acid (SAHA). Post-translational modifications of the histones of chromatin are important factors in regulating gene expression—so-called epigenetic gene regulation. Acetylation and deacetylation of lysine residues in histone tails, controlled by the activities of HDACs and histone acetyltransferases, are among the most studied post-translational modification of histones. In addition to chromatin protein, transcription factors, cell-signaling regulatory proteins, and proteins regulating cell death are substrates of HDACs and may be altered in function by HDAC inhibitors. HDAC inhibitors have several remarkable aspects. For instance, despite HDACs being ubiquitously distributed through chromatin, SAHA selectively alters the transcription of relatively few genes, and normal cells are at least 10-fold more resistant than transformed cells to SAHA and related HDAC inhibitor-induced cell death. HDAC inhibitors represent a relatively new group of targeted anticancer compounds, which are showing significant promise as agents with activity against a broad spectrum of neoplasms, at doses that are well tolerated by cancer patients. SAHA is one of the HDAC inhibitors most advanced in development. It is in phase I and II clinical trials for patients with both hematologic and solid tumors.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Figure 1: Interaction showing SAHA inhibition of HDAC.

Similar content being viewed by others

References

  1. Miller TA et al. (2003) Histone deacetylase inhibitors. J Med Chem 46: 5097–5116

    Article  CAS  Google Scholar 

  2. Marks PA et al. (2004) Histone deacetylase inhibitors. Adv Cancer Res 91: 137–168

    Article  CAS  Google Scholar 

  3. Yoshida M et al. (1990) Potent and specific inhibition of mammalian histone deacetylase both in vivo and in vitro by trichostatin A. J Biol Chem 265: 17174–17179

    CAS  PubMed  Google Scholar 

  4. Kyrylenko S et al. (2003) Differential regulation of the Sir2 histone deacetylase gene family by inhibitors of class I and II histone deacetylases. Cell Mol Life Sci 60: 1990–1997

    Article  CAS  Google Scholar 

  5. Remiszewski SW (2003) The discovery of NVP-LAQ824: from concept to clinic. Curr Med Chem 10: 2393–2402

    Article  CAS  Google Scholar 

  6. Furumai R et al. (2002) FK228 (depsipeptide) as a natural prodrug that inhibits class I histone deacetylases. Cancer Res 62: 4916–4921

    CAS  PubMed  Google Scholar 

  7. Piekarz R and Bates S (2004) A review of depsipeptide and other histone deacetylase inhibitors in clinical trials. Curr Pharm Des 10: 2289–2298

    Article  CAS  Google Scholar 

  8. Thibault A et al. (1994) A phase I and pharmacokinetic study of intravenous phenylacetate in patients with cancer. Cancer Res 54: 1690–1694

    CAS  PubMed  Google Scholar 

  9. Chang SM et al. (1999) Phase II study of phenylacetate in patients with recurrent malignant glioma: a North American Brain Tumor Consortium report. J Clin Oncol 17: 984–990

    Article  CAS  Google Scholar 

  10. Reid T et al. (2004) Phase II trial of the histone deacetylase inhibitor pivaloyloxymethyl butyrate (Pivanex, AN-9) in advanced non-small cell lung cancer. Lung Cancer 45: 381–386

    Article  Google Scholar 

  11. Atmaca A et al. (2004) A dose escalating phase I study with valproic acid (VPA) in patients with advanced cancer. ASCO Annual Meeting Proceedings (Post-Meeting Edition) [abstract #3169] J Clin Oncol 22 (Suppl)

  12. Lucas DM et al. (2004) The histone deacetylase inhibitor MS-275 induces caspase-dependent apoptosis in B-cell chronic lymphocytic leukemia cells. Leukemia 18: 1207–1214

    Article  CAS  Google Scholar 

  13. Kimmel KA et al. (2001) A Phase I dose-finding study of CI-994 in combination with capecitabine in patients with advanced solid tumors. [abstract #345] Proc Am Soc Clin Oncol 87: a345

    Google Scholar 

  14. Wolffe AP and Pruss D (1996) Deviant nucleosomes: the functional specialization of chromatin. Trends Genet 12: 58–62

    Article  CAS  Google Scholar 

  15. Luger K et al. (1997) Crystal structure of the nucleosome core particle at 2.8 Å resolution. Nature 389: 251–260

    Article  CAS  Google Scholar 

  16. Jenuwein T and Allis CD (2001) Translating the histone code. Science 293: 1074–1080

    Article  CAS  Google Scholar 

  17. Spotswood HT and Turner BM (2002) An increasingly complex code. J Clin Invest 110: 577–582

    Article  CAS  Google Scholar 

  18. Fischle W et al. (2003) Binary switches and modification cassettes in histone biology and beyond. Nature 425: 475–479

    Article  CAS  Google Scholar 

  19. Gui CY et al. (2004) Histone deacetylase (HDAC) inhibitor activation of p21WAF1 involves changes in promoter-associated proteins, including HDAC1. Proc Natl Acad Sci U S A 101: 1241–1246

    Article  CAS  Google Scholar 

  20. Arts J et al. (2003) Histone deacetylase inhibitors: from chromatin remodeling to experimental cancer therapeutics. Curr Med Chem 10: 2343–2350

    Article  CAS  Google Scholar 

  21. Glaser KB et al. (2003) Role of class I and class II histone deacetylases in carcinoma cells using siRNA. Biochem Biophys Res Commun 310: 529–536

    Article  CAS  Google Scholar 

  22. Cress WD and Seto E (2000) Histone deacetylases, transcriptional control, and cancer. J Cell Physiol 184: 1–16

    Article  CAS  Google Scholar 

  23. de Ruijter AJ et al. (2003) Histone deacetylases (HDACs): characterization of the classical HDAC family. Biochem J 370 (Part 3): 737–749

    Article  CAS  Google Scholar 

  24. Jones PA and Baylin SB (2002) The fundamental role of epigenetic events in cancer. Nat Rev Genet 3: 415–428

    Article  CAS  Google Scholar 

  25. Johnstone RW and Licht JD (2003) Histone deacetylase inhibitors in cancer therapy: is transcription the primary target? Cancer Cell 4: 13–18

    Article  CAS  Google Scholar 

  26. He LZ et al. (2001) Histone deacetylase inhibitors induce remission in transgenic models of therapy-resistant acute promyelocytic leukemia. J Clin Invest 108: 1321–1330

    Article  CAS  Google Scholar 

  27. Murata T et al. (2001) Defect of histone acetyltransferase activity of the nuclear transcriptional coactivator CBP in Rubinstein–Taybi syndrome. Hum Mol Genet 10: 1071–1076

    Article  CAS  Google Scholar 

  28. Finnin MS (1999) Structures of a histone deacetylase homologue bound to the TSA and SAHA inhibitors. Nature 401: 188–193

    Article  CAS  Google Scholar 

  29. Gray SG et al. (2004) Microarray profiling of the effects of histone deacetylase inhibitors on gene expression in cancer cell lines. Int J Oncol 24: 773–795

    CAS  PubMed  Google Scholar 

  30. Richon VM et al. (2000) Histone deacetylase inhibitor selectively induces p21WAF1 expression and gene-associated histone acetylation. Proc Natl Acad Sci U S A 97: 10014–10019

    Article  CAS  Google Scholar 

  31. Mitsiades CS et al. (2004) Transcriptional signature of histone deacetylase inhibition in multiple myeloma: biological and clinical implications. Proc Natl Acad Sci U S A 101: 540–545

    Article  CAS  Google Scholar 

  32. Butler LM et al. (2002) The histone deacetylase inhibitor SAHA arrests cancer cell growth, up-regulates thioredoxin-binding protein-2, and down-regulates thioredoxin. Proc Natl Acad Sci U S A 99: 11700–11705

    Article  CAS  Google Scholar 

  33. Warrener R et al. (2003) Tumor cell-selective cytotoxicity by targeting cell cycle checkpoints. FASEB J 17: 1550–1552

    Article  CAS  Google Scholar 

  34. Qiu L et al. (1999) Anti-tumour activity in vitro and in vivo of selective differentiating agents containing hydroxamate. Br J Cancer 80: 1252–1258

    Article  CAS  Google Scholar 

  35. Polevoda B and Sherman F (2002) The diversity of acetylated proteins. Genome Biol 3: reviews 0006

    Article  Google Scholar 

  36. Kouzarides T (2000) Acetylation: a regulatory modification to rival phosphorylation? EMBO J 19: 1176–1179

    Article  CAS  Google Scholar 

  37. Pei XY et al. (2004) Synergistic induction of oxidative injury and apoptosis in human multiple myeloma cells by the proteasome inhibitor bortezomib and histone deacetylase inhibitors. Clin Cancer Res 10: 3839–3852

    Article  CAS  Google Scholar 

  38. Rahmani M et al. (2003) Coadministration of the heat shock protein 90 antagonist 17-allylamino- 17-demethoxygeldanamycin with suberoylanilide hydroxamic acid or sodium butyrate synergistically induces apoptosis in human leukemia cells. Cancer Res 63: 8420–8427

    CAS  PubMed  Google Scholar 

  39. Kelly WK et al. (2003) Phase I clinical trial of histone deacetylase inhibitor: suberoylanilide hydroxamic acid administered intravenously. Clin Cancer Res 9: 3578–3588

    CAS  Google Scholar 

  40. Duvic M et al. (2003) Phase II trial of oral suberoylanilide hydroxamic acid (SAHA) for cutaneous T-cell lymphoma (CTCL) and peripheral T-cell lymphoma (PTCL). [abstract #625] Proc Am Soc Hematol

  41. Marks PA and Kelly WK Histone deacetylase inhibitors: novel targeted anti-cancer agents. In DNA Methylation Epigenetics and Metastasis. (Ed. Esteller M) Boston, Dordrecht, London: Kluwer Academic Publisher, in press

  42. Ungerstedt JS et al. (2005) Role of thioredoxin in the response of normal and transformed cells to histone deacetylase inhibitor. Proc Natl Acad Sci U S A 102: 673–678

    Article  CAS  Google Scholar 

  43. Peart MJ et al. (2003) Novel mechanisms of apoptosis induced by histone deacetylase inhibitors. Cancer Res 63: 4460–4471

    CAS  PubMed  Google Scholar 

  44. Butler LM et al. (2000) Suberoylanilide hydroxamic acid, an inhibitor of histone deacetylase, suppresses the growth of prostate cancer cells in vitro and in vivo. Cancer Res 60: 5165–5170

    CAS  PubMed  Google Scholar 

  45. Kelly WK et al. Phase I study of the oral histone deacetylase inhibitor: suberoylanilide hydroxamic acid (SAHA), in patients with advanced cancer. J Clin Oncol, in press

  46. Piekarz RL et al. (2001) Inhibitor of histone deacetylation, depsipeptide (FR901228), in the treatment of peripheral and cutaneous T-cell lymphoma: a case report. Blood 98: 2865–2868

    Article  CAS  Google Scholar 

  47. Piekarz RL et al. (2004) T-cell lymphoma as a model for the use of histone deacetylase inhibitors in cancer therapy: impact of depsipeptide on molecular markers, therapeutic targets, and mechanisms of resistance. Blood 103: 4636–4643

    Article  CAS  Google Scholar 

Download references

Acknowledgements

The studies reviewed in this chapter representing research in the authors' laboratories were supported, in part, by grants from: the National Cancer Institute (P30-CA-08748-40); Robert J and Helen C Kleberg Foundation; DeWitt Wallace Fund for the Memorial Sloan-Kettering Cancer Center; Susan and Jack Rudin Foundation; and The David H Koch Prostate Cancer Research Award.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Paul A Marks.

Ethics declarations

Competing interests

Memorial Sloan-Kettering Cancer Center and Columbia University jointly hold patents on hydroxamic acid based polar compounds, including SAHA, which are exclusively licensed to Aton Pharma, Inc., a Biotechnology Company acquired by Merck, Inc. Paul A Marks was a founder of Aton and both institutions and the founder had an equity position in Aton Pharma, Inc. Paul A Marks is a scientific consultant to Merck.

Glossary

PML-RARα

A specific chromosomal translocation, t(15;17), results in the fusion of the promyelocytic leukemia gene (PML) and the retinoic acid receptor gene (RARα)

CBFA2T1

Core-binding factor 2 translocated to 1, also known as AML1/ETO

RUBINSTEIN–TAYBI SYNDROME

A rare genetic disorder that causes developmental abnormalities of many organs, caused by mutations in the gene encoding CREB binding protein (CBP)

CDKN1A

Gene encoding the cyclin-dependent kinase inhibitor-1A protein, which is an important inhibitor of cellular proliferation in response to DNA damage

TNFSF10

Tumor necrosis factor (ligand) superfamily member 10 (also known as TRAIL or APO-2L)

Rights and permissions

Reprints and permissions

About this article

Cite this article

Kelly, W., Marks, P. Drug Insight: histone deacetylase inhibitors—development of the new targeted anticancer agent suberoylanilide hydroxamic acid. Nat Rev Clin Oncol 2, 150–157 (2005). https://doi.org/10.1038/ncponc0106

Download citation

  • Received:

  • Accepted:

  • Issue Date:

  • DOI: https://doi.org/10.1038/ncponc0106

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