Valproic acid defines a novel class of HDAC inhibitors inducing differentiation of transformed cells

Histone deacetylases (HDACs) play important roles in transcriptional regulation and pathogenesis of cancer. Thus, HDAC inhibitors are candidate drugs for differentiation therapy of cancer. Here, we show that the well‐tolerated antiepileptic drug valproic acid is a powerful HDAC inhibitor. Valproic acid relieves HDAC‐dependent transcriptional repression and causes hyperacetylation of histones in cultured cells and in vivo. Valproic acid inhibits HDAC activity in vitro, most probably by binding to the catalytic center of HDACs. Most importantly, valproic acid induces differentiation of carcinoma cells, transformed hematopoietic progenitor cells and leukemic blasts from acute myeloid leukemia patients. More over, tumor growth and metastasis formation are significantly reduced in animal experiments. Therefore, valproic acid might serve as an effective drug for cancer therapy.

[1]  C. Van Lint,et al.  The expression of a small fraction of cellular genes is changed in response to histone hyperacetylation. , 1996, Gene expression.

[2]  T. Tomasi,et al.  Activation of MHC Class I, II, and CD40 Gene Expression by Histone Deacetylase Inhibitors1 , 2000, The Journal of Immunology.

[3]  Wolfgang Löscher,et al.  Valproate: a reappraisal of its pharmacodynamic properties and mechanisms of action , 1999, Progress in Neurobiology.

[4]  P. Marks,et al.  Histone deacetylase inhibitors: inducers of differentiation or apoptosis of transformed cells. , 2000, Journal of the National Cancer Institute.

[5]  C. Tribioli,et al.  Distinct interactions of PML-RARα and PLZF-RARα with co-repressors determine differential responses to RA in APL , 1998, Nature Genetics.

[6]  H. Stunnenberg,et al.  Identification of a retinoic acid responsive element in the retinoic acid receptor & beta;gene , 1990, Nature.

[7]  Sander Kersten,et al.  Roles of PPARs in health and disease , 2000, Nature.

[8]  T. Baillie,et al.  Cytochrome P-450--catalyzed formation of delta 4-VPA, a toxic metabolite of valproic acid. , 1987, Science.

[9]  K. Kishi,et al.  Hematopoietic cytokine-dependent differentiation to eosinophils and neutrophils in a newly established acute promyelocytic leukemia cell line with t(15;17). , 1998, Experimental hematology.

[10]  K. Kinzler,et al.  PPAR d Is an APC-Regulated Target of Nonsteroidal Anti-Inflammatory Drugs , 1999 .

[11]  T. Heinzel,et al.  Histone deacetylase as a therapeutic target , 2001, Trends in Endocrinology & Metabolism.

[12]  C. Young,et al.  Butyrate and phenylacetate as differentiating agents: Practical problems and opportunities , 1995, Journal of cellular biochemistry. Supplement.

[13]  C. Glass,et al.  Coactivator and corepressor complexes in nuclear receptor function. , 1999, Current opinion in genetics & development.

[14]  P. Pandolfi,et al.  Therapeutic targeting of transcription in acute promyelocytic leukemia by use of an inhibitor of histone deacetylase. , 1998, Journal of the National Cancer Institute.

[15]  Thorsten Heinzel,et al.  Ligand-independent repression by the thyroid hormone receptor mediated by a nuclear receptor co-repressor , 1995, Nature.

[16]  R. Evans,et al.  A transcriptional co-repressor that interacts with nuclear hormone receptors , 1995, Nature.

[17]  S. Inoue,et al.  Role of the histone deacetylase complex in acute promyelocytic leukaemia , 1998, Nature.

[18]  R. Evans,et al.  The LAZ3(BCL-6) oncoprotein recruits a SMRT/mSIN3A/histone deacetylase containing complex to mediate transcriptional repression. , 1998, Nucleic acids research.

[19]  H. Nau,et al.  Asymmetric synthesis and teratogenic activity of (R)- and (S)-2-ethylhexanoic acid, a metabolite of the plasticizer di-(2-ethylhexyl)phthalate. , 1990, Life sciences.

[20]  Pier Giuseppe Pelicci,et al.  Histone deacetylases: a common molecular target for differentiation treatment of acute myeloid leukemias? , 2001, Oncogene.

[21]  S. Horinouchi,et al.  Trichostatin A and trapoxin: Novel chemical probes for the role of histone acetylation in chromatin structure and function , 1995, BioEssays : news and reviews in molecular, cellular and developmental biology.

[22]  K. Kinzler,et al.  PPARδ Is an APC-Regulated Target of Nonsteroidal Anti-Inflammatory Drugs , 1999, Cell.

[23]  C. Glass,et al.  ETO, a Target of t(8;21) in Acute Leukemia, Interacts with the N-CoR and mSin3 Corepressors , 1998, Molecular and Cellular Biology.

[24]  M. Guenther,et al.  Histone Deacetylase Is a Direct Target of Valproic Acid, a Potent Anticonvulsant, Mood Stabilizer, and Teratogen* , 2001, The Journal of Biological Chemistry.

[25]  S. Minucci,et al.  Oligomerization of RAR and AML1 transcription factors as a novel mechanism of oncogenic activation. , 2000, Molecular cell.

[26]  R A Rifkind,et al.  Suberoylanilide hydroxamic acid, an inhibitor of histone deacetylase, suppresses the growth of prostate cancer cells in vitro and in vivo. , 2000, Cancer research.

[27]  S. Minucci,et al.  Aberrant Recruitment of the Nuclear Receptor Corepressor-Histone Deacetylase Complex by the Acute Myeloid Leukemia Fusion Partner ETO , 1998, Molecular and Cellular Biology.

[28]  S. Schreiber,et al.  Nuclear Receptor Repression Mediated by a Complex Containing SMRT, mSin3A, and Histone Deacetylase , 1997, Cell.

[29]  S. Grégoire,et al.  Enantioselectivity in the induction of peroxisome proliferation by 2-ethylhexanoic acid. , 1992, Chirality.

[30]  C. Tribioli,et al.  Distinct interactions of PML-RARalpha and PLZF-RARalpha with co-repressors determine differential responses to RA in APL. , 1998, Nature genetics.

[31]  H Nau,et al.  New molecular bioassays for the estimation of the teratogenic potency of valproic acid derivatives in vitro: activation of the peroxisomal proliferator-activated receptor (PPARdelta). , 1999, Toxicology and applied pharmacology.

[32]  K. Umesono,et al.  Differential expression and activation of a family of murine peroxisome proliferator-activated receptors. , 1994, Proceedings of the National Academy of Sciences of the United States of America.

[33]  T. Suga,et al.  Enhancement of peroxisomal beta-oxidation in the liver of rats and mice treated with valproic acid. , 1985, Biochemical pharmacology.

[34]  W. Miller,et al.  Altered ligand binding and transcriptional regulation by mutations in the PML/RARalpha ligand-binding domain arising in retinoic acid-resistant patients with acute promyelocytic leukemia. , 2000, Blood.

[35]  T. Hoshino,et al.  ETO, fusion partner in t(8;21) acute myeloid leukemia, represses transcription by interaction with the human N-CoR/mSin3/HDAC1 complex. , 1998, Proceedings of the National Academy of Sciences of the United States of America.

[36]  J. Nadeau,et al.  A mouse model for valproate teratogenicity: parental effects, homeotic transformations, and altered HOX expression. , 2000, Human molecular genetics.

[37]  H. Nau,et al.  Valnoctamide, valpromide and valnoctic acid are much less teratogenic in mice than valproic acid , 1998, Epilepsy Research.

[38]  A. Zelent,et al.  Reduced retinoic acid-sensitivities of nuclear receptor corepressor binding to PML- and PLZF-RARalpha underlie molecular pathogenesis and treatment of acute promyelocytic leukemia. , 1998, Blood.

[39]  P. Marks,et al.  Structures of a histone deacetylase homologue bound to the TSA and SAHA inhibitors , 1999, Nature.

[40]  C. Allis,et al.  The language of covalent histone modifications , 2000, Nature.

[41]  H. Nau,et al.  Induction of differentiation in F9 cells and activation of peroxisome proliferator-activated receptor delta by valproic acid and its teratogenic derivatives. , 2001, Molecular pharmacology.

[42]  J. Gustafsson,et al.  Fatty acids activate a chimera of the clofibric acid-activated receptor and the glucocorticoid receptor. , 1992, Proceedings of the National Academy of Sciences of the United States of America.

[43]  C. Curry,et al.  The fetal valproate syndrome. , 1984, American journal of medical genetics.

[44]  P G Pelicci,et al.  Histone deacetylase-targeted treatment restores retinoic acid signaling and differentiation in acute myeloid leukemia. , 2001, Cancer research.

[45]  Wen‐Ming Yang,et al.  Histone Deacetylases Associated with the mSin3 Corepressor Mediate Mad Transcriptional Repression , 1997, Cell.

[46]  S. Minucci,et al.  Fusion proteins of the retinoic acid receptor-α recruit histone deacetylase in promyelocytic leukaemia , 1998, Nature.

[47]  L. Chin,et al.  Role for N-CoR and histone deacetylase in Sin3-mediated transcriptional repression , 1997, nature.

[48]  C. Glass,et al.  A complex containing N-CoR, mSln3 and histone deacetylase mediates transcriptional repression , 1997, nature.

[49]  H. Nau,et al.  Valproic acid-induced neural tube defects in mouse and human: aspects of chirality, alternative drug development, pharmacokinetics and possible mechanisms. , 1991, Pharmacology & toxicology.

[50]  C. Glass,et al.  The coregulator exchange in transcriptional functions of nuclear receptors. , 2000, Genes & development.

[51]  H. Nau,et al.  Pharmacological evaluation of various metabolites and analogues of valproic acid Anticonvulsant and toxic potencies in mice , 1985, Neuropharmacology.