174
Views
69
CrossRef citations to date
0
Altmetric
Review

Histone modification enzymes: novel targets for cancer drugs

, , &
Pages 135-154 | Published online: 02 Mar 2005

Bibliography

  • BALL P: Portrait of a molecule. Nature (2003) 421(6920:421–422.
  • FELSENFIELD G, GROUDINE M: Controlling the double helix. Nature (2003) 421(6921)448–453.
  • •Provides interesting discussion of epigenetic mechanisms.
  • KORNBERG RD: Chromatin structure: a repeating unit of histones and DNA. Science (1974) 184:868–871.
  • ••Landmark paper describing chromatinstructure.
  • WEINTRAUB H, GROUDINE M: Chromosomal subunits in active genes have an altered conformation. Science (1976) 193:848–856.
  • LUGER K, MADER AW, RICHMOND RK, SARGENT DF, RICHMOND TJ: Crystal structure of the nucleosome core particle at 2.8A resolution. Nature (1997) 389:251–260.
  • ••First report of the crystal structure of thenucleosome.
  • CHEUNG P, ALLIS CD, SASS ONE-CORSIP: Signalling to chromatin through histone modifications. Cell (2000) 103:263–271.
  • MARMORSTEIN R: Protein modules that manipulate histone tails for chromatin regulation. Nat. Rev Mol. Cell Biol(2001) 2:422–432.
  • •Useful review of histone-protein Interactions regulating chromatin.
  • STRAHL BD, ALLIS CD: The language of covalent histone modifications. Nature (2000) 403:41–45.
  • ••The first paper to suggest an interactivelanguage between various histone modifications termed the 'histone code'.
  • TURNER BM: Cellular memory and the histone code. Cell (2002) 111:285–291.
  • ••Further definition of the histone code.
  • BROWN R, STRATHDEE G: Epigenomics and epigenetic therapy of cancer. Trends Mol Med. (2002) 11:15–21.
  • CRESS WD, SETO E: Histone deacetylases, transcriptional control and cancer.j Cell Phys. (2000) 184:1–16.
  • •A thorough, early review of the association between histone deacetylation and cancer.
  • MAHLKNECHT U, HOELZER D: Histone acetylation modifiers in the pathogenesis of malignant disease. Mol Med. (2000) 6:623–644.
  • TIMMERMANN S, LEHRMANN H, POLESSKAYA A, HAREL-BELLAN A: Histone acetylation and disease. Cell Mol Life Sci. (2001) 58:728–736.
  • MARKS PA, RICHON VM, BRESLOW R, RIFKIND RA: Histone deacetylase inhibitors as new anti-cancer drugs. Curl: Opin. Oncol (2001) 13:477–483.
  • DAVIS PK, BRACKMANN RK: Chromatin remodelling and cancer. Cancer Bid. Ther. (2003) 2:22–29.
  • •Review focusing on the relationship between cancer and regulation of chromatin structure.
  • RICE JC, ALLIS CD: Histone methylation versus histone acetylation: new insights into epigenetic regulation. Curl: Opin. Cell Biol. (2001) 13:263–273.
  • ••In-depth discussion of the interactionbetween histone acetylation and methylation.
  • BANNISTER AJ, SCHNEIDER R, KOUZARIDES T: Histone methylation: dynamic or static? Cell (2002) 109:801–806.
  • ••Discussion of the role of histonemethylation in chromatin regulation and gene expression.
  • BEISEL C, IMHOF A, GREEN J, KREMMER E, SAUER F: Histone methylation by the Drosophila epigenetic transcriptional regulator Ash 1. Nature (2002) 419(6909):857–862.
  • HANS F, DIMITROV S: Histone H3 phosphorylation and cell division. Oncogene (2001) 20:3021–3027.
  • GOTO H, YASUI Y, NIGG EA, INAGAKI M: Aurora-B phosphorylates Histone H3 at serine 28 with regard to the mitotic chromosome condensation. Genes Cells (2002) 7:11–17.
  • RAMASWAMY V, WILLIAMS JS, ROBINSON KM, SOPKO RL, SCHULTZ MC: Global control of histone modification by the anaphase-promoting complex. Mol Cell Bid. (2003) 24:9136–9149.
  • MURNION ME, ADAMS RR, CALLISTER DM, ALLIS CD, EARNSHAW WC, SWEDLOW JR: Chromatin-associated protein phosphatase 1 regulates Aurora B and histone H3 phosphorylation. J. Biol. Chem. (2001) 276:26656–26665.
  • ALLFREY VG, FAULKNER R, MIRSKY AE: Acetylation and methylation of histones and their possible role in the regulation of RNA synthesis. Proc. Natl. Acad. Sci. USA (1964) 51:786–794.
  • GRUNSTEIN M: Histone acetylation in chromatin structure and transcription. Nature (1997) 389:349–352.
  • ••Excellent early review outlining theImportant effects of histone acetylation on gene expression.
  • STRUHL K: Histone acetylation and transcriptional regulatory mechanisms. Genes Dev. (1998) 12:599–606.
  • CAMERON EE, BACHMAN KE, MYOHANEN S, HERMAN JG, BAYLIN SB: Synergy of demethylation and histone deacetylase inhibition in the re-expression of genes silenced in cancer. Nat. Genet. (1999) 21:103–107.
  • •Description of the important synergistic interaction between methylation and acetylation.
  • KOUZARIDES T: Acetylation: a regulatory modification to rival phophorylation? EMBO J. (2000) 19:1176–1179.
  • GREGORY PD, WAGNER K, HORZ W: Histone acetylation and chromatin remodelling. Exp. Cell Res. (2001) 265:195–202.
  • MORALES V, RICHARD-FOY H: Role of histone N-terminal tails and their acetylation in nucleosome dynamics. Mol Cell Bid. (2000) 20:7230–7237.
  • WOLFFE AP, HAYES JJ: Chromatin disruption and modification. Nucleic Adds Res. (1999) 27:711–720.
  • STERNER DE, BERGER SL: Acetylation of histones and transcription-related factors. Microbial. Mol Biol. Rev (2000) 64:435–459.
  • DE RUIJTER AJ, VAN GENNIP AH, CARON HN, KEMP S, VAN KUILENBURG AB: Histone deacetylases: characterisation of the classical HDAC family. Biochem. 1 (2003) 370:737–749.
  • ••A concise and thorough review outliningthe known effects of individual HDAC enzymes.
  • GROZINGER CM, SCHREIBER SL: Deacetylase enzymes: biological functions and the use of small molecule inhibitors. Chem. Biol. (2002) 9:3–16.
  • ••Review covering both HDAC enzymebiology and HDACI effects.
  • ZHANG Y, LI N, CARON C et al: HDAC-6 interacts with and deacetylates tubulin and microtubules in vivo. EMBO (2003) 22:1168–79.
  • •One of the first reports describing substrate specificity for an HDAC enzyme.
  • LIU Y, COLOSIMO AL, YANG X-J, LIAO D: Adenovirus ElB 55-kilodalton oncoprotein inhibits p53 acetylation by PCAE Ma. Cell. Biol. (2000) 20:5540–5553.
  • GU W, ROEDER RG: Activation of p53 sequence-specific DNA binding by acetylation of the p53 C-terminal domain. Cell (1997) 90:595–606.
  • KUO MH, ALLIS CD: Roles of histone acetyltransferases and deacetylases in gene regulation. BioEssays (1998) 20:615–626.
  • MARTINEZ-BALBAS MA, BAUER UM, NEILSEN SJ, BREHM A, KOUZARIDES T: Regulation of E2F1 activity by acetylation. EMBO J. (2000) 19:662–671.
  • BOYES J, BYFIELD P, NAKATANI Y, OGRYZKO V: Regulation of activity of the transcription factor GATA-1 by acetylation. Nature (1998) 396:594–598.
  • DHALLUIN C, CARLSON JE, ZENG L, HE C, AGGARWAL AK, ZHOU MM: Structure and ligand of a histone acetyltransferase bromodomain. Nature (1999) 399:491–496.
  • •Demonstration of the HAT bromodomain structure.
  • WORKMAN P: Scoring a bull's-eye against cancer genome targets. Curr. Opin. Pharmacol (2001) 1:342–352.
  • OGRYZKO VV: Mammalian histone acetyltransferases and their complexes. Cell Mol Life Sci. (2001) 58:683–692.
  • NG HH, BIRD A: Histone deacetylases: silencers for hire. Trends Biochem. Sci. (2000) 25:121–126.
  • BERTOS NR, WANG AH, YANG XJ: Class II histone deacetylases: structure, function and regulation. Biochem. Cell Biol. (2001) 79:243–252.
  • FISCHLE W, EMILIANI S, HENDZEL MJ et al.: A new family of human histone deacetylases related to Saccharomyces cerevisiae HDAlp. J. Biol. Chem. (1999) 274:11713–11720.
  • FISCHLE W, KIERMER V, DEQUIEDT F, VERDIN E: The emerging role of class II histone deacetylases. Biochem. Cell Biol. (2001) 79:337–348.
  • IMAI S, ARMSTRONG CM, KAEBERLEIN M, GUARENTE L: Transcriptional silencing and longevity protein Sir2 is a NAD dependent histone deacetylase. Nature (2000) 403:795–800.
  • DUTNALL RN, PILLUS L: DecipheringNAD-dependent deacetylases. Cell (2001) 105:161–164.
  • PARSONS XH, GARCIA SM, PILLUS L, KADONAGA JT: Histone deacetylation by Sir2 generates a transcriptionally repressed nucleoprotein complex. Proc. Natl. Acad. Sci. USA (2003) 100:1609–1614.
  • FINNIN MS, DONIGIAN JR, COHEN A et al.: Structures of a histone deacetylase homologue bound to the TSA and SAHA inhibitors. Nature (1999) 401:188–193.
  • ••Landmark paper illustrating the molecularbinding between HDACI an HDAC enzyme homologues.
  • HUBBERT C, GUARDIOLA A, SHAO R et al: HDAC6 is a microtubule-associated deacetylase. Nature (2002) 417:455–458.
  • ••First evidence for HDAC6 substratespecificity.
  • ALLAND L, MUHLE R, HOU H Jr et al: Role for NCoR and HDAC in Sin3-mediated transcriptional repression. Nature (1997) 387(6628):49–55.
  • HEINZEL T, LAVINSKY RM, MULLEN TM et al: A complex containing NCoR, mSin3 and histone deacetylase mediates transcriptional repression. Nature (1997) 387:43–48.
  • YAO YL, YANG WM, SETO E: Regulation of transcription factor YY1 by acetylation and deacetylation. Ma. Cell. Biol. (2001) 21:5979–5991.
  • BLACK AR, BLACK JD, AZIZKHAN-CLIFFORD J: Spl and Kruppel-like factor family of transcription factors in cell growth regulation and cancer. J. Cell Physiol (2001) 188:143–160.
  • MAGNAGHI-JAULIN L, GROISMAN R, NAGUIBNEVA I et al: Retinoblastoma protein represses transcription by recruiting a histone deacetylase. Nature (1998) 391:601–605.
  • •Discovery of the link between Rb and HDAC enzymes.
  • CHEN D, MA H, HONG H et al.: Regulation of transcription by a protein methyltransferase. Science (1999) 284:2174–2177.
  • STRAHL BD, OHBA R, COOK RG, ALLIS CD: Methylation of histone H3 at lysine 4 is highly conserved and correlates with transcriptionally active nuclei in 7etrahymena. Proc. Natl. Acad. Sci. USA (1999) 96:14967–14972.
  • •Evidence for the role of histone methylation in transcriptional activation.
  • JENUWEIN T, ALLIS CD: Translating the histone code. Science (2001) 293:1074–1080.
  • SANTOS-ROSA H, SCHNEIDER R, BANNISTER AJ et al.: Active genes are tri-methylated at K4 of histone H3. Nature (2002) 419:407–411.
  • SCHNEIDER R, BANNISTER AJ, KOUZARIDES T: Unsafe SETs: histone lysine methyltransferases and cancer. Trends Biochem. Sci (2002) 27:396–402.
  • •Review of histone methyltransferase enzymes and discussion of their possible role in cancer.
  • FENG Q, WANG H, NG GH et al: Methylation of H3-lysine9 is mediated by a new family of HMTases without a SET domain. Curr. Biol. (2002) 12:1052–1058.
  • KOUZARIDES T: Histone methylation in transcriptional control. Curr. Opin. Genet. Dev. (2002) 9:40–48.
  • BREILING A, ORLANDO V: SET domain proteins reset gene expression. Nat. Struct. Biol. (2002) 9:894–896.
  • REA S, EISENHABER F, O'CARROLL D et al.: Regulation of chromatin structure by site-specific histone H3 methyltransferases. Nature (2000) 406:593–599.
  • •This review focuses on the mechanisms by which methyltransferase enzymes regulate chromatin.
  • XIAO B, JING C, WILSON JR et al: Structure and catalytic mechanism of the human histone methyltranferase SET7/9. Nature (2003) 421(6923):652–656.
  • •Discovery of the structure of a human histone methyltransferase.
  • ZHANG Y, REINBERG D: Transcription regulation by histone methylation: interplay between different covalent modifications of the core histone tails. Genes Dev. (2001) 15:2343–2360.
  • WANG H, HUANG ZQ, XIA L et al: Methylation of histone H4 at arginine 3 facilitates transcriptional activation by nuclear hormone receptor. Science (2001) 293(5530:853–857.
  • CHEN D, HUANG SM, STALLCUP MR: Synergistic p160 coactivator dependent enhancement of estrogen receptor function by CARM1 and p300. I Biol. Chem. (2000) 275:40810–40816.
  • WET Y, MIZZEN CA, COOK RG, GOROVSKY MA, ALLIS CD: Phosphorylation of histone H3 at serine 10 is correlated with chromosome condensation during mitosis and meiosis in 7etrahymena. Proc. Nati Acad. Sci. USA (1998) 95:7480–7484.
  • KATAYAMA H, BRINKLEY WR, SEN S: The Aurora kinases: role in cell transformation and tumorigenesis. Cancer Metastasis Rev (2003) 22:451–464.
  • ••Review and discussion of the biology ofAurora kinases.
  • BISCHOFF JR, ANDERSON L, ZHU Y et al.: A homologue of Drosophila Aurora kinase is oncogenic and amplified in human colorectal cancers. EMBO J. (1998) 17:3052–3065.
  • CARMENA M, EARNSHAW WC: The cellular geography of Aurora kinases. Nat. Rev Mol. Cell Biol. (2003) 4:842–854.
  • ••Review and discussion of the biology ofAurora kinases.
  • MOORE SC: The elusive structural role of ubiquitinated histones. Biochem. Cell Biol. (2002) 80:311–319.
  • SUN Z-W, ALLIS CD: Ubiquitination of histone H2B regulates H3 methylation and gene silencing in yeast. Nature (2002) 418:104–108.
  • ••Good description of the effects of histoneubiquitination.
  • BRIGGS SD, XIAO T, SUN ZW et al: Trans-histone regulatory pathway in chromatin. Nature (2002) 418:498.
  • LADURNER AG: Inactivating chromosomes: a macro domain that minimizes transcription. Mol. (2003) 12:1–3.
  • WADE PA: Transcriptional control at regulatory checkpoints by histone deacetylases: molecular connections between cancer and chromatin. Hum. Mol. Genet. (2001) 10:693–698.
  • •Detailed discussion of the aberrant molecular events linking HDAC activity with cancer.
  • JOHNSTONE R: Histone deacetylase inhibitors: novel drugs for the treatment of cancer. Nat. Rev Drug Discov. (2002) 1:287–299.
  • ••Excellent review of HDACIs.
  • CAIRNS BR: Emerging roles for chromatinremodelling in cancer biology. Trends Cell Biol. (2001) 11:S15–S21.
  • CHAKRABORTY S, SENYUK V, NUCIFORA G: Genetic lesions and perturbation of chromatin architecture: a road to cell transformation. J. Cell Biochem. (2001) 82:310–325.
  • WARNER SL, BEARSS DJ, HAN H, VON HOFF DD: Targeting Aurora-2 kinase in cancer. Mol. Cancer Ther (2003) 2:589–595.
  • GIORDANO A, AVANTAGGIATI ML: p300 and CBP: partners for life and death. J. Cell Physiol (1999) 181:218–230.
  • GAYTHER SA, BATLEY SJ, LINGER L, BANNISTER A, THORPE K, CHIN SF: Mutations truncating the EP300 acetylase in human cancers. Nat. Genet. (2000) 24:300–303.
  • ••Characterisation of mutations in theHAT, p300.
  • MURAOKA M, KONISHI M, KIKUCHI-YANOSHITA R et al: p300 gene alterations in colorectal and gastric carcinomas. Oncogene (1996) 12:1565–1569.
  • •Genetic alterations of p300 in cancer.
  • GILES RH, PETERS DJ, BREUNING MH: Conjunction dysfunction: CBP/p300 in human disease. Trends Genet. (1998) 14:178–183.
  • PETRIJ F, GILES RH, DAUWERSE HG et al.: Rubenstein-Taybi syndrome caused by mutations in the transcriptional co-activator CBP. Nature (1995) 376:348–351.
  • KUNG AL, REBEL VI, BRONSON RT et al.: Gene dose-dependent control of haematopoiesis and haematologic tumour suppression by CBP. Genes Dev. (2000) 14:272–277.
  • TANAKA T, NARUSE I, MACKAWA T, MASUYA H, SHIROISHI T, ISHII S: Abnormal skeletal patterning in embryos lacking a single CBPallele: a partial similarity with Rubenstein-Taybi syndrome. Proc. Natl. Acad. Sci. USA (1997) 94:10215–10220.
  • SAKAI K, NAGAHARA H, ABE K, OBATA J: Loss of heterozygosity on chromosome 16 in hepatocellular carcinoma. J. Gastroenterof Hepatol (1992) 7:88–92.
  • LUNDBLAD JR, KWOK RP, LAURANCE ME, HARTER ML, GOODMAN RH: Adenoviral E1A-associated protein p300 as a functional homologue of the transcriptional co-activator CBP. Nature (1995) 374:85–88.
  • ECKNER R, LUDLOW JW, LILL NL et al: Association of p300 and CBP with simian virus 40 large T antigen. Mol. Cell Biol. (1996) 16:3454–3464.
  • ANZICK SL, KONONEN J, WALKER RL et al.: AIB1, a steroid receptor coactivator amplified in breast and ovarian cancer. Science (1997) 277:965–968.
  • BERNS EM, VAN STAVEREN IL, KLIJN JG, FOEKENS JA: Predictive value of SRC-1 for tamoxifen response of recurrent breast cancer. Breast Cancer Res. Treat. (1998) 48:87–92.
  • ROWLEY JD, RESHMI S, SOBULO 0et al.: All patients with the t(11; 16) (q23;p13.3) that involves MLL and CBP have treatment related haematologic disorders. Blood (1997) 90: 535–541.
  • AYTON PM, CLEARY ML: Molecular mechanisms of leukaemogenesis mediated by MLL fusion proteins. Oncogene (2001) 20:5695–5707.
  • IDA K, KITABAYASHI I, TAKI T et al:Adenoviral E1A-associated protein p300 is involved in acute myeloid leukaemia with t(11;22)(q23;q13). Blood (1997) 90:4699–4704.
  • YAO YL, OH SP, FUCHS M et al: Gene dosage-dependent embryonic development and proliferation defects in mice lacking the transcriptional integrator p300. Cell (1998) 93:361-372. •In vivo study of p300knockdown in mice.
  • DEBES JD, SEBO J, LOHSE CM, MURPHY LM, HAUGEN DL, TINDALL DJ: p300 in prostate cancer proliferation and progression. Cancer Res. (2003) 63:7638–7640.
  • •p300 in prostate cancer.
  • MINUCCI S, NERVI C, LO COCO F, PELICCI PG: Histone deacetylases: a common molecular target for differentiation treatment of acute myeloid leukaemias? Oncogene (2001) 20:3110–3115.
  • LIN RJ, NAGY L, INOUE S, SHAO W, MILLER WH Jr, EVANS RM: Role of the histone deacetylase complex in acute promyelocytic leukaemia. Nature (1998) 391:811–814.
  • •Description of aberrant fusion protein activity in APL.
  • HE LZ, GUIDEZ F, TRIBIOLI C et al: Distinct interactions of PML-RAR alpha and PLZF-RAR alpha with co-repressors determine differential responses to RA in APL. Nat. Genet. (1998) 18:126–135.
  • GRIGNANI F, DE MATTEIS S, NERVI C et al.: Fusion proteins of the retinoic acid receptor-alpha recruit histone deacetylase in promyelocytic leukaemia. Nature (1998) 391:815–818.
  • DHORDAIN P, LIN RJ, QUIEF S et al: The LAZ3 (BCL6) oncoprotein recruits a SMRT/mSin3A/histone deacetylase containing complex to mediate transcriptional repression. Nur. Adds Res. (1998) 26:4645–4651.
  • WANG J, HOSHINO T, REDNER RL, KAJIGAYA S, LIU JM: ETO, fusion partner in t(8:21) acute myeloid leukaemia, represses transcription by interaction with the human NcoR/mSin3.HDAC1 complex. Proc. Natl. Acad. Sci. USA (1998) 95:10860–10865.
  • WANG J, SAUNTHARARAJAH Y, REDNER RL, LUI J: Inhibitors of histone deacetylase relieve ETO-mediated repression and induce differentiation of AML1-ETO leukaemia cells. Cancer Res. (1999) 59:2766–2769.
  • SEARS R, OHTANI K, NEVINS JR: Identification of positively and negatively acting elements regulating expression of the E2F2 gene in response to cell growth signals. Mol. Cell Biol. (1997) 17: 5227–5235.
  • MCARTHUR GA, LAHERTY CD, QUEVA C et al: The Mad protein family links transcriptional repression to cell differentiation. Cold Spring Harbour Symp. Quant Biol. (1998) 63:423–433.
  • DANG CV: c-Myc target genes involved in cell growth, apoptosis and metabolism. Mol. Cell. Biol. (1999) 19:1–11.
  • NOMURA T, KHAN MM, KAUL SC et al.: Ski is a component of the histone deacetylase complex required for transcriptional repression by Mad and thyroid hormone receptor. Genes Dev. (1999) 13:412–423.
  • BREHM A, MISKA EA, MCCANCE DJ, REID JL, BANNISTER AJ, KOUZARIDES T: Retinoblastoma protein recruits histone deacetylase to repress transcription. Nature (1998) 391:597–601.
  • SELLERS WR, KAELIN WG Jr: Role of the retinoblastoma protein in the pathogenesis of human cancer. J. Clin Onc. (1997) 15:3301–3312.
  • ZHANG Y, WOODFORD N, XIA X, HAMBURGER AW: Repression of E2F1-mediated transcription by the ErbB3 binding protein Ebp1 involves histone deacetylases. Nur. Acids Res. (2003) 31:2168–2177.
  • KIM MS, KWON HJ, LEE YM et al: Histone deacetylases induce angiogenesis by negative regulation of tumour suppressor genes. Nat. Med. (2001) 7:437–443.
  • ••HDAC effects on angiogenesis.
  • DEROANNE CF, BONJEAN K, SERVOTTE S et al: Histone deacetylase inhibitors as antiangiogenic agents altering vascular endothelial growth factor signalling. Oncogene (2002) 21:427–436.
  • ••HDAC effects on angiogenesis.
  • ROBERTSON KD: DNA methylation, methyltransferases and cancer. Oncogene (2001) 20:3139–3155.
  • ROUNTREE MR, BACHMAN KE, HERMAN JG, BAYLIN SB: DNA methylation, chromatin inheritance and cancer. Oncogene (2001) 20:3156–3165.
  • •Review of DNA methylation including synergism with histone acetylation in cancer.
  • JAENISCH R, BIRD A: Epigenetic regulation of gene expression: how the genome integrates intrinsic and environmental signals. Nat. Genet. Suppl. (2003) 33:245–254.
  • PETERS AHFM, O'CARROLL D, SCHERTHAN H et al: Loss of the 5uv39 histone methyltransferases impairs mammalian heterochromatin and genome stability. Cell (2001) 107:323–337.
  • NIELSEN PR, NIETLISPACH D, MOTT HR et al.: Structure of the HP1 chromodomain bound to histone H3 methylated at lysine 9. Nature (2002) 416(6876):103–107.
  • KONDO Y, SHEN L, ISSA JP: Critical role of histone methylation in tumour suppressor gene silencing in colorectal cancer. Mol. Cell. Bid. (2003) 23:206–215.
  • VARAMBALLY S, DHANASEKARAN SM, ZHOU M: The polycomb group protein EZH2 is involved in progression of prostate cancer. Nature (2002) 419:624–629.
  • •Evidence for the role of EZH2 in breast and prostate cancers.
  • KLEER CG, CAO Q, VARAMBALLY S: EZH2 is a marker of aggressive breast cancer and promotes neoplastic transformation of breast epithelial cells. Proc. Natl. Acad. Li. USA (2003) 100:11606–11611.
  • •Evidence for the role of EZH2 in breast and prostate cancers.
  • BISCHOFF JR, PLOWMAN GD: The Aurora/IpIlp kinase family: regulators of chromosome segregation and cytokinesis. Trends Cell Bid. (1999) 9:454–459.
  • ZHOU H, KUANG J, ZHONG L et al: Tumour amplified kinase STK/BTAK induces centrosome amplification, aneuploidy and transformation. Nat. Genet. (1998) 20:189–193.
  • MIYOSHI Y, IWAO K, EGAWA C, NOGUCHI S: Association of centrosomal kinase STK15/BTAK mRNA expression with chromosomal instability in human breast cancers. Int. J. Cancer (2001) 92:370–373.
  • ROMANOV SR, KOZAKIEWICZ BK, HOLST CR, STAMPFER MR, HAUPT LM, TISTY TD: Normal human mammary epithelial cells spontaneously escape senescence and acquire genomic changes. Nature (2001) 409:633–637.
  • EWART-TOLAND A, BRIASSOULI P, DE KONIG JP et al: Identification of Stk6/STK15 as a candidate low-penetrance tumour-susceptibility gene in mouse and human. Nat. Genet. (2003) 34:403–412.
  • ANAND S, PENRHYN-LOWE S, VENKITARAMAN AR: Aurora-A amplification overrides the mitotic spindle assembly checkpoint, inducing resistance to Taxol. Cancer Cell (2003) 3:51–62.
  • LAU OD, KUNDU TK, SOCCIO RE et al.: HATs off: Selective synthetic inhibitors of the histone acetyltransferase p300 and PCAE MolCell (1999) 5:589–595.
  • ••First discovery of a HAT inhibitor.
  • CEBRAT M, KIM CM, THOMPSON PR, DAUGHERTY M, COLE PA: Synthesis and analysis of potential prodrugs of coenzyme A analogues for the inhibition of the histone acetyltransferase p300. Bioorg. Med. Chem. (2003) 11:3307–3313.
  • ••First development of a potential prodrugfor HAT inhibition.
  • POUX AN, MARMORSTEIN R: Molecular basis for Gcn5/PCAF histone acetyltransferase selectivity for histone and nonhistone substrates. Biochemistry (2003) 42:14366–14374.
  • TURLAIS F, HARDCASTLE A, ROWLANDS MG et al: High throughput screening (HTS) for identification of histone acetyltransferases using scintillating microplates (FlashPlate Anna]. Biochem. (2001) 298:62–68.
  • STIMSON L, ROWLANDS MG, NEWBATT Y et al: Isothiazolones as novel inhibitors of pcaf and p300 histone acetyltransferase activity. Clin. Cancer Res. (2003) 9(Suppl.):87.
  • BALASUBRAMANYAM K, SWAMINATHAN V, RANGANATHAN A, KUNDU TK: Small molecule modulators of histone acetyltransferase p300. J. Biol. Chem. (2003) 278:19134–19140.
  • CHEETHAM GM, KNEGTEL RM, COLL JT et al.: Crystal structure of Aurora-2, an oncogenic serineithreonine kinase. J. Biol. Chem. (2002) 277:42419–42422.
  • ••Discovery of the structure of Aurora 2.
  • MAHADEVAN D, BEARSS DJ, VANKAYALAPATI H: Structure-based design of novel anti-cancer agents targeting aurora kinases. Curl: Med. Chem. Anti-Canc. Agents (2003) 1:25–34.
  • YOSHIDA M, FURUMAI R, NISHIYAMA M, KOMATSU Y, NISHINO N, HORINOUCHI S: Histone deacetylase as a new target for cancer chemotherapy. Cancer Chemother. Pharmacol (2001) 48\(Suppl. 1):520–526.
  • REMISZEWSKI SW: Recent advances in the discovery of small molecule histone deacetylase inhibitors. Curr. Opin. Drug Discov. Devel. (2002) 5:487–499.
  • ELAUT G, TOROK G, VINKEN M et al: Major Phase I biotransformation pathways of Trichostatin A in rat hepatocytes and in rat and human liver microsomes. Drug Metab. Dispos. (2002) 30:1320–1328.
  • ATADJA P, GAOL, KWON P et al: Selective growth inhibition of tumour cells by a novel histone deacetylase inhibitor, NVP-LAQ824. Cancer Res. (2004) 64:689–695.
  • PLUMB JA, FINN PW, WILLIAMS RJ et al.: Pharmacodynamic response and inhibition of growth of human tumour xenografts by the novel histone deacetylase inhibitor PXD101. Mol. Cancer Ther. (2003) 2:721–728.
  • RICHON VM, SANDHOFF TW, RIFKIND RA, MARKS PA: Histone deacetylase inhibitor selectively induces p21WAF1 expression and gene-associated histone acetylation. Proc. Natl. Acad Sci USA (2000) 97:10014–10019.
  • MARKS PA, RICHON VM, RIFKIND RA: Histone deacetylase inhibitors: inducers of differentiation or apoptosis of transformed cells. J. Nati Cancer Inst. (2000) 92:1210–1216.
  • KELLY WK, RICHON VM, O'CONNOR 0 et al: Phase I clinical trial of histone deacetylase inhibitor: Suberoylanilide hydroxamic acid administered intravenously. Clin. Cancer Res. (2003) 9:3578–3588.
  • KELLY WK, O'CONNOR O, RICHON VM et al: Phase I clinical trial of an oral histone deacetylase inhibitor: Suberoylanilide hydroxamic acid (SAHA). Clin. Cancer Res. (2003) 9(Suppl.):226.
  • ARTS J, VAN EMELEM K, ANGIBAUD P et al.: Small molecule inhibitors of histone deacetylases (HDACs): identification of JNJ16241199, a potent oral antitumour agent. Clin. Cancer Res.. (2003) 9(Suppl.):87.
  • KRUH J: Effects of sodium butyrate, a new pharmacological agent, on cells in culture. Mol. Cell Biochem. (1982) 42:65–82.
  • KIJIMA M, YOSHIDA M, SUGITA K, HORINOUCHI S, BEPPU T: Trapoxin, an antitumour cyclic tetrapeptides an irreversible inhibitor of mammalian histone deacetylase. J. Biol. Chem. (1993) 268:22429–22435.
  • FURUMAI R, KOMATSU Y, NISHINO N, KHOCHBIN S, YOSHIDA M, HORINOUCHI S: Potent histone deacetylase inhibitors built from trichostatin A and cyclic tetrapeptide antibiotics including trapoxin. Proc. Natl. Acad. Sci. USA (2001) 98:87–92.
  • SAITO A, YAMASHITA T, MARIKO Y et al.: A synthetic inhibitor of histone deacetylase, MS-27-275, with marked in vivo antitumour activity against human tumours. Proc. Natl. Acad. Sci. USA (1999) 96:4592–4597.
  • SU GH, SOHN TA, RYU B, KERN SE: A novel histone deacetylase inhibitor identified by high-throughput transcriptional screening of a compound library. Cancer Res. (2000) 60:3137–3142.
  • GROZINGER CM, CHAO ED, BLACKWELL HE, MOAZED D, SCHREIBER SL: Identification of a class of small molecule inhibitors of the sirtuin family of NAD-dependent deacetylases by phenotypic screening. J. Biol. Chem. (2001) 276:38837–38843.
  • GLASER KB, LI J, STAVER MJ, WEI R-Q, ALBERT DH, DAVIDSEN SK: Role of Class I and Class II histone deacetylases in carcinoma cells using siRNA. Biochem. Biophys. Res. Comm. (2003) 310:529–536.
  • HUE, DUL E, SUNG CM et al.: Identification of novel isoform-selective inhibitors within Class I histone deacetylases. Pharmacol. Exp. Ther. (2003) 307:720–728.
  • GLASER KB, STAVER MJ, WARING JF, STENDER J, ULRICH RG, DAVIDSEN SK: Gene expression profiling of mutiple histone deacetylase (HDAC) inhibitors: defining a common gene set produced by HDAC inhibition in T24 and MDA carcinoma cell lines. Mol. Canc. Ther. (2003) 2:151–163.
  • BLAGOSKLONNY MV, ROBEY R, SACKETT DL et al.: Histone deacetylase inhibitors all induce p21 but differentially cause tubulin acetylation, mitotic arrest and cytotoxicity. Mol. Canc. Ther.(2002) 1: 937–941.
  • QIU L, BURGESS A, FAIRLIE DP, LEONARD H, PARSONS PG, GABRIELLI BG: Histone deacetylase inhibitors trigger a G2 checkpoint in normal cells that is defective in tumour cells. Mol. Biol. Cell (2000) 11:2069–2083.
  • SHI H, WEI S, LEU Y-W: Triple analysis of the cancer epigenome: an integrated microarray system for assessing gene expression, DNA methylation and histone acetylation. Cancer Res. (2003) 63:2164–2171.
  • VIGUSHIN DM, COOMBES RC: Histone deacetylase inhibitors in cancer treatment. Anticancer Drugs (2002) 13:1–13.
  • MAGNER WJ, KAZIM AL, STEWART C et al: Activation of MHC Class I, II and CD40 gene expression by histone deacetylase inhibitors. J. Immunol (2000) 165:7017–7024.
  • KWON HJ, KIM MS, KIM MJ, NAKAJIMA H, KIM KW: Histone deacetylase inhibitor FK228 inhibits tumour angiogenesis. Int. J. Cancer (2002) 97:290–296.
  • LIU LT, CHANG HC, CHIANG LC, HUNG WC: Histone deacetylase inhibitor up-regulates RECK to inhibit MMP-2 activation and cancer cell invasion. Cancer Res. (2003) 63:3069–3072.
  • HAN JW, AHN SH, PARK SH et al: Apicidin, a histone deacetylase inhibitor, inhibits proliferation of tumour cells via induction of p21WAF1/Cip 1 and gelsolin. Cancer Res. (2000) 60:6068–6074.
  • FINZER P, KUNTZEN C, SOTO U, ZUR HAUSEN H, ROSL F: Inhibitors of histone deacetylase arrest cell cycle and induce apoptosis in cervical carcinoma cells circumventing human papilloma virus oncogene expression. Oncogene (2001) 20:4768–4776.
  • SANDOR V, SENDEROWICZ A, MERTINS S et al: p21-dependent G1 arrest with downregulation of cyclin D1 with upregulation of cyclin E by the histone deacetylase inhibitor FR901228. BE J. Cancer (2000) 83:817–825.
  • ZHU WG, LAKSHMANAN RR, BEAL MD, OTTERS ON GA: DNA methyltransferase inhibition enhances apoptosis induced by histone deacetylase inhibitors. Cancer Res. (2001) 61:1327–1333.
  • YU X, GUO ZS, MARCU MG et al: Modulation of p53, erbB1, erbB2 and raf-1 expression in lung cancer cells by depsipeptide FR901228. J. Natl. Cancer Inst. (2002) 94:504–513.
  • MALONEY A, WORKMAN P: Hsp90 as a new therapeutic target for cancer therapy: the story unfolds. Expert Opia Biol. Ther. (2002) 2:3–24.
  • CATLEY L, WEISBERG E, TAI Y-T et al: NVP-LAQ824 is a potent novel histone deacetylase inhibitor with significant activity against multiple myeloma. Blood (2003) 102:2615–2622.
  • QIU L, KELSO MJ, HANSEN C, WEST ML, FAIRLIE DP, PARSONS PG: Anti-tumour activity in vitro and in vivo of selective differentiating agents containing hydroxamate. Br. J. Cancer (1999) 80:1252–1258.
  • KIM YB, LEE KH, SUGITA K, YOSHIDA M, HORINOUCHI S: Oxamflatin is a novel antitumour compound that inhibits mammalian histone deacetylase. Oncogene (1999) 18:2461–2470.
  • UEDA H, MANDA T, MATSUMOTO S et al: FR901228, a novel antitumour bicyclic depsipeptide produced by Chromobactedum violaceum No. 968 III. Antitumour activities on experimental tumours in mice. J. Antibiot. (Tokyo) (1994) 47:315–323.
  • GILBERT J, BAKER SD, BOWLING MK et al: A Phase I dose escalation and bioavailability study of oral sodium phenylbutyrate in patients with refractory solid tumor malignancies. Clin. Cancer Res. (2001) 8:2292–2300.
  • CARDUCCI MA, GILBERT J, BOWLING MK et al: A Phase I clinical and pharmacological evaluation of sodium phenylbutyrate on an 120-h infusion schedule. Clin. Cancer Res. (2001) 10:3047–3055.
  • WARRELL RP Jr, HE LZ, RICHON V, CALLEJA E, PANDOLFI PP: Therapeutic targeting of transcription in acute promyelocytic leukaemia by use of an inhibitor of histone deacetylase. J. Natl. Cancer Inst. (1998) 90:1621–1625.
  • PIEKARZ RL, ROBEY R, SANDOR V et al: Inhibitor of histone deacetylation, depsipeptide (FR901228), in the treatment of peripheral and cutaneous T-cell lymphoma: a case report. Blood (2001) 98:2865–2868.
  • SANDOR V, BAKKE S, ROBEY RW et al.: Phase I trial of the histone deacetylase inhibitor, depsipeptide (FR901228, NSC 630176), in patients with refractory neoplasams. Clin. Cancer Res. (2002) 8:718–728.
  • WISINSKI KB: A Phase I study of an oral histone deacetylase inhibitor, MS-275, in patients (pts) with refractory solid tumours and lymphomas. OM. Cancer Res. (2003) 9(Suppl.):92.
  • NIMMANAPALLI R, FUINO L, BALI P et al: Histone deacetylase inhibitor LAQ824 both lowers expression and promotes proteasomal degradation of Bcr-Abl and induces apoptosis of imatinib mesylate-sensitive or refractory chronic myelogenous leukaemia-blast crisis cells. Cancer Res. (2003) 63:5126–5135.
  • AHERNE GW, ROWLANDS MG, STIMSON L, WORKMAN P: Assays for the identification and evaluation of histone acetyltransferase inhibitors. Methods (2002) 26:245–253.
  • SCHMEICHEL KL: Centrosome cycle studies reveal promising candidates for anti-cancer drug design. Breast Cancer Res. (2003) 5:59.
  • BRINKMANN H, DAHLER AL, POPA C et al.: Histone hyperacetylation induced by histone deacetylase inhibitors is not sufficient to cause growth inhibition in human dermal fibroblasts. I Biol. Chem. (2001) 276:22491–22499.
  • HANAHAN D, WEINBERG RA: The hallmarks of cancer. Cell (2000) 100:57–70.
  • WORKMAN P: The opportunities and challenges of personalised genome-based molecular therapies for cancer: targets, technologies and molecular chaperones. Cancer Chemother. Pharmacol. (2003) 52\(Suppl. 1):545–556.

Websites

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.