Prospects: Histone deacetylase inhibitors

Histone deacetylase (HDAC), inhibitors represent a new class of targeted anti‐cancer agents. Several of these compounds are in clinical trials with significant activity against a spectrum of both hematologic and solid tumors at doses that are well tolerated by the patients. The HDAC inhibitors are a structurally diverse group of molecules that can induce growth arrest, differentiation, apoptosis, and autophagocytic cell death of cancer cells. While the base sequence of DNA provides the genetic code for proteins, the expression of genes is regulated, in large part, by the structure of the chromatin proteins around which the DNA is wrapped (epigenetic gene regulation). The acetylation and deacetylation of the lysines in the tails of the core histones, among the most extensively studied aspects of chromatin structure, is controlled by the action of two families of enzymes, histone deacetylases (HDACs) and histone acetyltransferases (HATs). Protein components of transcription factor complexes and many other non‐histone proteins are also substrates for HDACs and HATs. The structure and activity of these non‐histone proteins may be altered by acetylation/deacetylation with consequent effects on various cell functions including gene expression, cell cycle progression, and cell death pathways. This review focuses on several key questions with respect to the mechanism of action of HDACi, including, what are the different cell phenotypes induced by HDACi, why are normal cells compared to transformed cells relatively resistant to HDACi induced cell death, why are certain tumors more responsive to HDACi than others, and what is the basis of the selectivity of HDACi in altering gene expression. The answers to these questions will have therapeutic importance since we will identify targets for enhancing the efficacy and safety of HDACi. © 2005 Wiley‐Liss, Inc.

[1]  Ricardo Macarron,et al.  Identification of Novel Isoform-Selective Inhibitors within Class I Histone Deacetylases , 2003, Journal of Pharmacology and Experimental Therapeutics.

[2]  Minoru Yoshida,et al.  Novel histone deacetylase inhibitors: cyclic tetrapeptide with trifluoromethyl and pentafluoroethyl ketones. , 2004, Bioorganic & Medicinal Chemistry Letters.

[3]  Hing Y Leung,et al.  Upregulation and Nuclear Recruitment of HDAC1 in Hormone Refractory Prostate Cancer , 2004, The Prostate.

[4]  C. Benz,et al.  Clinical development of histone deacetylase inhibitors as anticancer agents. , 2005, Annual review of pharmacology and toxicology.

[5]  Stuart L Schreiber,et al.  Domain-selective small-molecule inhibitor of histone deacetylase 6 (HDAC6)-mediated tubulin deacetylation , 2003, Proceedings of the National Academy of Sciences of the United States of America.

[6]  K. Glaser,et al.  Role of class I and class II histone deacetylases in carcinoma cells using siRNA. , 2003, Biochemical and biophysical research communications.

[7]  T. Richmond,et al.  Crystal structure of the nucleosome core particle at 2.8 Å resolution , 1997, Nature.

[8]  K. Glaser,et al.  Histone deacetylase inhibitors: the Abbott experience. , 2003, Current medicinal chemistry.

[9]  P. Marks,et al.  A class of hybrid polar inducers of transformed cell differentiation inhibits histone deacetylases. , 1998, Proceedings of the National Academy of Sciences of the United States of America.

[10]  K. Furge,et al.  Microarray profiling of the effects of histone deacetylase inhibitors on gene expression in cancer cell lines. , 2004, International journal of oncology.

[11]  L. Guarente,et al.  The Sir2 family of protein deacetylases. , 2004, Annual review of biochemistry.

[12]  Sang Gyun Kim,et al.  Class I Histone Deacetylase-Selective Novel Synthetic Inhibitors Potently Inhibit Human Tumor Proliferation , 2004, Clinical Cancer Research.

[13]  A. V. van Kuilenburg,et al.  Histone deacetylases (HDACs): characterization of the classical HDAC family. , 2003, The Biochemical journal.

[14]  R A Rifkind,et al.  Second generation hybrid polar compounds are potent inducers of transformed cell differentiation. , 1996, Proceedings of the National Academy of Sciences of the United States of America.

[15]  L. Ngo,et al.  Histone deacetylase (HDAC) inhibitor activation of p21WAF1 involves changes in promoter-associated proteins, including HDAC1. , 2004, Proceedings of the National Academy of Sciences of the United States of America.

[16]  J. R. Somoza,et al.  Structural snapshots of human HDAC8 provide insights into the class I histone deacetylases. , 2004, Structure.

[17]  D. Reinberg,et al.  Transcription regulation by histone methylation: interplay between different covalent modifications of the core histone tails. , 2001, Genes & development.

[18]  H. Kwon,et al.  Expression Profile of Histone Deacetylase 1 in Gastric Cancer Tissues , 2001, Japanese journal of cancer research : Gann.

[19]  R. Johnstone,et al.  Histone-Deacetylase Inhibitors for the Treatment of Cancer , 2004, Cell cycle.

[20]  C. Monneret,et al.  Histone deacetylase inhibitors. , 2005, European journal of medicinal chemistry.

[21]  J. Johnston,et al.  Machinery and Transactivation by Stat5 Recruitment of the Basal Transcription Deacetylase Activity Is Required For , 2022 .

[22]  直江 知樹,et al.  Histone Deacetylase Inhibitor , 1998, Definitions.

[23]  Gordon K Smyth,et al.  Identification and functional significance of genes regulated by structurally different histone deacetylase inhibitors. , 2005, Proceedings of the National Academy of Sciences of the United States of America.

[24]  H. Scher,et al.  The histone deacetylase inhibitor SAHA arrests cancer cell growth, up-regulates thioredoxin-binding protein-2, and down-regulates thioredoxin , 2002, Proceedings of the National Academy of Sciences of the United States of America.

[25]  Wolfgang Fischle,et al.  Binary switches and modification cassettes in histone biology and beyond , 2003, Nature.

[26]  S. Bates,et al.  A review of depsipeptide and other histone deacetylase inhibitors in clinical trials. , 2004, Current pharmaceutical design.

[27]  J. Melo,et al.  Biology of Chronic Myeloid Leukemia and Possible Therapeutic Approaches to Imatinib-Resistant Disease , 2004, International journal of hematology.

[28]  Minoru Yoshida,et al.  From discovery to the coming generation of histone deacetylase inhibitors. , 2003, Current medicinal chemistry.

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

[30]  Ivan V. Gregoretti,et al.  Molecular evolution of the histone deacetylase family: functional implications of phylogenetic analysis. , 2004, Journal of molecular biology.

[31]  John M. Shelton,et al.  Histone Deacetylase 4 Controls Chondrocyte Hypertrophy during Skeletogenesis , 2004, Cell.

[32]  J. Denu The Sir 2 family of protein deacetylases. , 2005, Current opinion in chemical biology.

[33]  S. Grant,et al.  Histone deacetylase inhibitors in clinical development , 2004, Expert opinion on investigational drugs.

[34]  Elias S. J. Arnér,et al.  Physiological functions of thioredoxin and thioredoxin reductase. , 2000, European journal of biochemistry.

[35]  G. Perez,et al.  Role of thioredoxin in the response of normal and transformed cells to histone deacetylase inhibitors. , 2005, Proceedings of the National Academy of Sciences of the United States of America.

[36]  L. Schwartz,et al.  Phase I study of an oral histone deacetylase inhibitor, suberoylanilide hydroxamic acid, in patients with advanced cancer. , 2005, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[37]  P. Marks,et al.  Drug Insight: histone deacetylase inhibitors—development of the new targeted anticancer agent suberoylanilide hydroxamic acid , 2005, Nature Clinical Practice Oncology.

[38]  F. Dequiedt,et al.  Class II histone deacetylases: versatile regulators. , 2003, Trends in genetics : TIG.

[39]  B. Turner,et al.  An increasingly complex code. , 2002, The Journal of clinical investigation.

[40]  Y. Yatabe,et al.  Reduced expression of class II histone deacetylase genes is associated with poor prognosis in lung cancer patients , 2004, International journal of cancer.

[41]  M. Caraglia,et al.  Acetylation of proteins as novel target for antitumor therapy: Review article , 2004, Amino Acids.

[42]  Ricky W Johnstone,et al.  Histone deacetylase inhibitors in cancer therapy: is transcription the primary target? , 2003, Cancer cell.

[43]  A. Harel-Bellan,et al.  Histone acetyltransferases and deacetylases in the control of cell proliferation and differentiation. , 2002, Advances in cancer research.

[44]  E. Seto,et al.  Regulation of histone deacetylase activities , 2004, Journal of cellular biochemistry.

[45]  S. Grant,et al.  Synergistic Induction of Oxidative Injury and Apoptosis in Human Multiple Myeloma Cells by the Proteasome Inhibitor Bortezomib and Histone Deacetylase Inhibitors , 2004, Clinical Cancer Research.

[46]  S. Bates,et al.  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. , 2004, Blood.

[47]  Y. Bang,et al.  Histone Deacetylase Inhibitors for Cancer Therapy , 2006, Epigenetics.

[48]  Marie Joseph,et al.  Transcriptional signature of histone deacetylase inhibition in multiple myeloma: biological and clinical implications. , 2004, Proceedings of the National Academy of Sciences of the United States of America.

[49]  P. Marks,et al.  Histone deacetylases and cancer: causes and therapies , 2001, Nature Reviews Cancer.

[50]  A. Mirsky,et al.  ACETYLATION AND METHYLATION OF HISTONES AND THEIR POSSIBLE ROLE IN THE REGULATION OF RNA SYNTHESIS. , 1964, Proceedings of the National Academy of Sciences of the United States of America.