373
Views
123
CrossRef citations to date
0
Altmetric
Review

Development of histone deacetylase inhibitors for cancer treatment

&
Pages 583-598 | Published online: 10 Jan 2014

References

  • Santos-Rosa H, Caldas C. Chromatin modifier enzymes, the histone code and cancer. Eur. J. Cancer41(16), 2381–2402 (2005).
  • Vogelstein B, Kinzler KW. Cancer genes and the pathways they control. Nat. Med.10(8), 789–799 (2004).
  • 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. USA51, 786–794 (1964).
  • Grunstein M. Histone acetylation in chromatin structure and transcription. Nature389(6649), 349–352 (1997).
  • Bird A. DNA methylation patterns and epigenetic memory. Genes Dev.16(1), 6–21 (2002).
  • Herman JG, Baylin SB. Gene silencing in cancer in association with promoter hypermethylation. N. Engl. J. Med.349(21), 2042–2054 (2003).
  • Jones PA, Baylin SB. The fundamental role of epigenetic events in cancer. Nat. Rev. Genet.3(6), 415–428 (2002).
  • Jones DO, Cowell IG, Singh PB. Mammalian chromodomain proteins: their role in genome organisation and expression. Bioessays22(2), 124–137 (2000).
  • Johnstone RW. Histone-deacetylase inhibitors: novel drugs for the treatment of cancer. Nat. Rev. Drug Discov.1(4), 287–299 (2002).
  • Taddei A, Roche D, Bickmore WA, Almouzni G. The effects of histone deacetylase inhibitors on heterochromatin: implications for anticancer therapy? EMBO Rep.6(6), 520–524 (2005).
  • Yang WM, Tsai SC, Wen YD, Fejer G, Seto E. Functional domains of histone deacetylase-3. J. Biol. Chem.277(11), 9447–9454 (2002).
  • Yang WM, Yao YL, Sun JM, Davie JR, Seto E. Isolation and characterization of cDNAs corresponding to an additional member of the human histone deacetylase gene family. J. Biol. Chem.272(44), 28001–28007 (1997).
  • Gao L, Cueto MA, Asselbergs F, Atadja P. Cloning and functional characterization of HDAC11, a novel member of the human histone deacetylase family. J. Biol. Chem.277(28), 25748–25755 (2002).
  • Longo VD, Kennedy BK. Sirtuins in aging and age-related disease. Cell126(2), 257–268 (2006).
  • Denu JM. The Sir 2 family of protein deacetylases. Curr. Opin. Chem. Biol.9(5), 431–440 (2005).
  • Buck SW, Gallo CM, Smith JS. Diversity in the Sir2 family of protein deacetylases. J. Leukoc. Biol.75(6), 939–950 (2004).
  • Vigushin DM, Coombes RC. Histone deacetylase inhibitors in cancer treatment. Anticancer Drugs13(1), 1–13 (2002).
  • Marks P, Rifkind RA, Richon VM et al. Histone deacetylases and cancer: causes and therapies. Nat. Rev. Cancer1(3), 194–202 (2001).
  • Jung M. Inhibitors of histone deacetylase as new anticancer agents. Curr. Med. Chem.8(12), 1505–1511 (2001).
  • Kelly WK, O’Connor OA, Marks PA. Histone deacetylase inhibitors: from target to clinical trials. Expert Opin. Investig. Drugs11(12), 1695–1713 (2002).
  • Arts J, de Schepper S, Van Emelen K. Histone deacetylase inhibitors: from chromatin remodeling to experimental cancer therapeutics. Curr. Med. Chem.10(22), 2343–2350 (2003).
  • Sengupta N, Seto E. Regulation of histone deacetylase activities. J. Cell. Biochem.93(1), 57–67 (2004).
  • Dokmanovic M, Marks PA. Prospects: histone deacetylase inhibitors. J. Cell Biochem.96(2), 293–304 (2005).
  • Zhang Z, Yamashita H, Toyama T et al. Quantitation of HDAC1 mRNA expression in invasive carcinoma of the breast. Breast Cancer Res. Treat.94(1), 11–16 (2005).
  • Krusche CA, Wulfing P, Kersting C et al. Histone deacetylase-1 and -3 protein expression in human breast cancer: a tissue microarray analysis. Breast Cancer Res. Treat.90(1), 15–23 (2005).
  • Choi JH, Kwon HJ, Yoon BI et al. Expression profile of histone deacetylase 1 in gastric cancer tissues. Jpn. J. Cancer Res.92(12), 1300–1304 (2001).
  • Toh Y, Yamamoto M, Endo K et al. Histone H4 acetylation and histone deacetylase 1 expression in esophageal squamous cell carcinoma. Oncol. Rep.10(2), 333–338 (2003).
  • Saji S, Kawakami M, Hayashi S et al. Significance of HDAC6 regulation via estrogen signaling for cell motility and prognosis in estrogen receptor-positive breast cancer. Oncogene24(28), 4531–4539 (2005).
  • Varshochi R, Halim F, Sunters A et al. ICI182,780 induces p21Waf1 gene transcription through releasing histone deacetylase 1 and estrogen receptor α from Sp1 sites to induce cell cycle arrest in MCF-7 breast cancer cell line. J. Biol. Chem.280(5), 3185–3196 (2005).
  • Osada H, Tatematsu Y, Saito H et al. Reduced expression of class II histone deacetylase genes is associated with poor prognosis in lung cancer patients. Int. J. Cancer112(1), 26–32 (2004).
  • Zhu P, Martin E, Mengwasser J et al. Induction of HDAC2 expression upon loss of APC in colorectal tumorigenesis. Cancer Cell5(5), 455–463 (2004).
  • Bradbury CA, Khanim FL, Hayden R et al. Histone deacetylases in acute myeloid leukaemia show a distinctive pattern of expression that changes selectively in response to deacetylase inhibitors. Leukemia19(10), 1751–1759 (2005).
  • Bolden JE, Peart MJ, Johnstone RW. Anticancer activities of histone deacetylase inhibitors. Nat. Rev. Drug Discov.5(9), 769–784 (2006).
  • Minucci S, Pelicci PG. Histone deacetylase inhibitors and the promise of epigenetic (and more) treatments for cancer. Nat. Rev. Cancer6(1), 38–51 (2006).
  • Rosato RR, Grant S. Histone deacetylase inhibitors: insights into mechanisms of lethality. Expert Opin. Ther. Targets9(4), 809–824 (2005).
  • Caron C, Boyault C, Khochbin S. Regulatory cross-talk between lysine acetylation and ubiquitination: role in the control of protein stability. Bioessays27(4), 408–415 (2005).
  • Butler LM, Zhou X, Xu WS et al. The histone deacetylase inhibitor SAHA arrests cancer cell growth, up-regulates thioredoxin-binding protein-2, and down-regulates thioredoxin. Proc. Natl Acad. Sci. USA99(18), 11700–11705 (2002).
  • Marchion DC, Bicaku E, Daud AI et al. Sequence-specific potentiation of topoisomerase II inhibitors by the histone deacetylase inhibitor suberoylanilide hydroxamic acid. J. Cell Biochem.92(2), 223–237 (2004).
  • Marchion DC, Bicaku E, Daud AI, Sullivan DM, Munster PN. Valproic acid alters chromatin structure by regulation of chromatin modulation proteins. Cancer Res.65(9), 3815–3822 (2005).
  • Munster PN, Troso-Sandoval T, Rosen N et al. The histone deacetylase inhibitor suberoylanilide hydroxamic acid induces differentiation of human breast cancer cells. Cancer Res.61(23), 8492–8497 (2001).
  • Secrist JP, Zhou X, Richon VM. HDAC inhibitors for the treatment of cancer. Curr. Opin. Investig. Drugs4(12), 1422–1427 (2003).
  • Gray SG, Qian CN, Furge K, Guo X, Teh BT. Microarray profiling of the effects of histone deacetylase inhibitors on gene expression in cancer cell lines. Int. J. Oncol.24(4), 773–795 (2004).
  • Peart MJ, Smyth GK, van Laar RK et al. Identification and functional significance of genes regulated by structurally different histone deacetylase inhibitors. Proc. Natl Acad. Sci. USA102(10), 3697–3702 (2005).
  • Della Ragione F, Criniti V, Della Pietra V et al. Genes modulated by histone acetylation as new effectors of butyrate activity. FEBS Lett.499(3), 199–204 (2001).
  • Van Lint C, Emiliani S, Verdin E. The expression of a small fraction of cellular genes is changed in response to histone hyperacetylation. Gene Expr.5(4–5), 245–253 (1996).
  • Marks PA, Richon VM, Miller T, Kelly WK. Histone deacetylase inhibitors. Adv. Cancer Res.91, 137–168 (2004).
  • Richon VM, Sandhoff TW, Rifkind RA, Marks PA. Histone deacetylase inhibitor selectively induces p21WAF1 expression and gene-associated histone acetylation. Proc. Natl Acad. Sci. USA97(18), 10014–10019 (2000).
  • Bhalla KN. Epigenetic and chromatin modifiers as targeted therapy of hematologic malignancies. J. Clin. Oncol.23(17), 3971–3993 (2005).
  • Gui CY, Ngo L, Xu WS, Richon VM, Marks PA. Histone deacetylase (HDAC) inhibitor activation of p21WAF1 involves changes in promoter-associated proteins, including HDAC1. Proc. Natl Acad. Sci. USA101(5), 1241–1246 (2004).
  • Sambucetti LC, Fischer DD, Zabludoff S et al. Histone deacetylase inhibition selectively alters the activity and expression of cell cycle proteins leading to specific chromatin acetylation and antiproliferative effects. J. Biol. Chem.274(49), 34940–34947 (1999).
  • Zhang Y, Fatima N, Dufau ML. Coordinated changes in DNA methylation and histone modifications regulate silencing/derepression of luteinizing hormone receptor gene transcription. Mol. Cell. Biol.25(18), 7929–7939 (2005).
  • Reid G, Metivier R, Lin CY et al. Multiple mechanisms induce transcriptional silencing of a subset of genes, including oestrogen receptor α, in response to deacetylase inhibition by valproic acid and trichostatin A. Oncogene24(31), 4894–4907 (2005).
  • Archer SY, Meng S, Shei A, Hodin RA. p21(WAF1) is required for butyrate-mediated growth inhibition of human colon cancer cells. Proc. Natl Acad. Sci. USA95(12), 6791–6796 (1998).
  • Archer SY, Johnson J, Kim HJ et al. The histone deacetylase inhibitor butyrate downregulates cyclin B1 gene expression via a p21/WAF-1-dependent mechanism in human colon cancer cells. Am. J. Physiol. Gastrointest. Liver Physiol.289(4), G696–G703 (2005).
  • Hu J, Colburn NH. Histone deacetylase inhibition down-regulates cyclin D1 transcription by inhibiting nuclear factor-κB/p65 DNA binding. Mol. Cancer Res.3(2), 100–109 (2005).
  • Kurz EU, Wilson SE, Leader KB et al. The histone deacetylase inhibitor sodium butyrate induces DNA topoisomerase II α expression and confers hypersensitivity to etoposide in human leukemic cell lines. Mol. Cancer Ther.1(2), 121–131 (2002).
  • Li GC, Zhang X, Pan TJ, Chen Z, Ye ZQ. Histone deacetylase inhibitor trichostatin A inhibits the growth of bladder cancer cells through induction of p21WAF1 and G1 cell cycle arrest. Int. J. Urol.13(5), 581–586 (2006).
  • Marks PA, Michaeli J, Jackson J, Richon VM, Rifkind RA. Induced differentiation of murine erythroleukemia cells (MELC) by polar compounds: marked increased sensitivity of vincristine resistant MELC. Prog. Clin. Biol. Res.316B, 171–181 (1989).
  • Marks PA, Richon VM, Kiyokawa H, Rifkind RA. Inducing differentiation of transformed cells with hybrid polar compounds: a cell cycle-dependent process. Proc. Natl Acad. Sci. USA91(22), 10251–10254 (1994).
  • Mukhopadhyay NK, Weisberg E, Gilchrist D et al. Effectiveness of trichostatin A as a potential candidate for anticancer therapy in non-small-cell lung cancer. Ann. Thorac. Surg.81(3), 1034–1042 (2006).
  • Phillips A, Bullock T, Plant N. Sodium valproate induces apoptosis in the rat hepatoma cell line, FaO. Toxicology192(2–3), 219–227 (2003).
  • 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. Blood98(9), 2865–2868 (2001).
  • Planchon P, Magnien V, Beaupain R et al. Differential effects of butyrate derivatives on human breast cancer cells grown as organotypic nodules in vitro and as xenografts in vivo. In vivo6(6), 605–610 (1992).
  • Rosato RR, Maggio SC, Almenara JA et al. The histone deacetylase inhibitor LAQ824 induces human leukemia cell death through a process involving XIAP down-regulation, oxidative injury, and the acid sphingomyelinase-dependent generation of ceramide. Mol. Pharmacol.69(1), 216–225 (2006).
  • Ryu JK, Lee WJ, Lee KH et al. SK-7041, a new histone deacetylase inhibitor, induces G2-M cell cycle arrest and apoptosis in pancreatic cancer cell lines. Cancer Lett.237(1), 143–154 (2006).
  • Takai N, Ueda T, Nishida M, Nasu K, Narahara H. Anticancer activity of MS-275, a novel histone deacetylase inhibitor, against human endometrial cancer cells. Anticancer Res.26(2A), 939–945 (2006).
  • Tsai SC, Valkov N, Yang WM et al. Histone deacetylase interacts directly with DNA topoisomerase II. Nat. Genet.26(3), 349–353 (2000).
  • Weisberg E, Catley L, Kujawa J et al. Histone deacetylase inhibitor NVP-LAQ824 has significant activity against myeloid leukemia cells in vitro and in vivo. Leukemia18(12), 1951–1963 (2004).
  • Wetzel M, Premkumar DR, Arnold B, Pollack IF. Effect of trichostatin A, a histone deacetylase inhibitor, on glioma proliferation in vitro by inducing cell cycle arrest and apoptosis. J. Neurosurg.103(Suppl. 6), 549–556 (2005).
  • Vrana JA, Decker RH, Johnson CR et al. Induction of apoptosis in U937 human leukemia cells by suberoylanilide hydroxamic acid (SAHA) proceeds through pathways that are regulated by Bcl-2/Bcl-XL, c-Jun, and p21CIP1, but independent of p53. Oncogene18(50), 7016–7025 (1999).
  • Sandor V, Senderowicz A, Mertins S et al. p21-dependent g(1) arrest with downregulation of cyclin D1 and upregulation of cyclin E by the histone deacetylase inhibitor FR901228. Br. J. Cancer83(6), 817–825 (2000).
  • Abe M, Kufe DW. Sodium butyrate induction of milk-related antigens in human MCF-7 breast carcinoma cells. Cancer Res.44(10), 4574–4577 (1984).
  • Davis T, Kennedy C, Chiew YE, Clarke CL, deFazio A. Histone deacetylase inhibitors decrease proliferation and modulate cell cycle gene expression in normal mammary epithelial cells. Clin. Cancer Res.6(11), 4334–4342 (2000).
  • Zhou Q, Melkoumian ZK, Lucktong A et al. Rapid induction of histone hyperacetylation and cellular differentiation in human breast tumor cell lines following degradation of histone deacetylase-1. J. Biol. Chem.275(45), 35256–35263 (2000).
  • Wang S, Yan-Neale Y, Cai R, Alimov I, Cohen D. Activation of mitochondrial pathway is crucial for tumor selective induction of apoptosis by LAQ824. Cell Cycle5(15), 1662–1668 (2006).
  • Johnstone RW, Licht JD. Histone deacetylase inhibitors in cancer therapy: is transcription the primary target? Cancer Cell4(1), 13–18 (2003).
  • Peart MJ, Tainton KM, Ruefli AA et al. Novel mechanisms of apoptosis induced by histone deacetylase inhibitors. Cancer Res.63(15), 4460–4471 (2003).
  • Marks PA, Richon VM, Rifkind RA. Histone deacetylase inhibitors: inducers of differentiation or apoptosis of transformed cells. J. Natl Cancer Inst.92(15), 1210–1216 (2000).
  • Rosato RR, Almenara JA, Dai Y, Grant S. Simultaneous activation of the intrinsic and extrinsic pathways by histone deacetylase (HDAC) inhibitors and tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) synergistically induces mitochondrial damage and apoptosis in human leukemia cells. Mol. Cancer Ther.2(12), 1273–1284 (2003).
  • Zhao Y, Tan J, Zhuang L et al. Inhibitors of histone deacetylases target the Rb-E2F1 pathway for apoptosis induction through activation of proapoptotic protein Bim. Proc. Natl Acad. Sci. USA102(44), 16090–16095 (2005).
  • Rahmani M, Reese E, Dai Y et al. Cotreatment with suberanoylanilide hydroxamic acid and 17-allylamino 17-demethoxygeldanamycin synergistically induces apoptosis in Bcr-Abl+ cells sensitive and resistant to STI571 (imatinib mesylate) in association with down-regulation of Bcr-Abl, abrogation of signal transducer and activator of transcription 5 activity, and Bax conformational change. Mol. Pharmacol.67(4), 1166–1176 (2005).
  • Sawa H, Murakami H, Ohshima Y et al. Histone deacetylase inhibitors such as sodium butyrate and trichostatin A induce apoptosis through an increase of the bcl-2-related protein Bad. Brain Tumor Pathol.18(2), 109–114 (2001).
  • Strait KA, Warnick CT, Ford CD et al. Histone deacetylase inhibitors induce G2-checkpoint arrest and apoptosis in cisplatinum-resistant ovarian cancer cells associated with overexpression of the Bcl-2-related protein Bad. Mol. Cancer Ther.4(4), 603–611 (2005).
  • Bali P, Pranpat M, Swaby R et al. Activity of suberoylanilide hydroxamic acid against human breast cancer cells with amplification of her-2. Clin. Cancer Res.11(17), 6382–6389 (2005).
  • Sutheesophon K, Kobayashi Y, Takatoku MA et al. Histone deacetylase inhibitor depsipeptide (FK228) induces apoptosis in leukemic cells by facilitating mitochondrial translocation of Bax, which is enhanced by the proteasome inhibitor bortezomib. Acta Haematol.115(1–2), 78–90 (2006).
  • Cao XX, Mohuiddin I, Ece F, McConkey DJ, Smythe WR. Histone deacetylase inhibitor downregulation of bcl-xl gene expression leads to apoptotic cell death in mesothelioma. Am. J. Respir. Cell Mol. Biol.25(5), 562–568 (2001).
  • Shankar S, Singh TR, Fandy TE et al. Interactive effects of histone deacetylase inhibitors and TRAIL on apoptosis in human leukemia cells: involvement of both death receptor and mitochondrial pathways. Int. J. Mol. Med.16(6), 1125–1138 (2005).
  • Zhang XD, Gillespie SK, Borrow JM, Hersey P. The histone deacetylase inhibitor suberic bishydroxamate regulates the expression of multiple apoptotic mediators and induces mitochondria-dependent apoptosis of melanoma cells. Mol. Cancer Ther.3(4), 425–435 (2004).
  • Muhlethaler-Mottet A, Flahaut M, Bourloud KB et al. Histone deacetylase inhibitors strongly sensitise neuroblastoma cells to TRAIL-induced apoptosis by a caspases-dependent increase of the pro- to anti-apoptotic proteins ratio. BMC Cancer6, 214 (2006).
  • Kim HR, Kim EJ, Yang SH et al. Trichostatin A induces apoptosis in lung cancer cells via simultaneous activation of the death receptor-mediated and mitochondrial pathway? Exp. Mol. Med.38(6), 616–624 (2006).
  • Insinga A, Monestiroli S, Ronzoni S et al. Inhibitors of histone deacetylases induce tumor-selective apoptosis through activation of the death receptor pathway. Nat. Med.11(1), 71–76 (2005).
  • Hirano T. The ABCs of SMC proteins: two-armed ATPases for chromosome condensation, cohesion, and repair. Genes Dev.16(4), 399–414 (2002).
  • Wei Y, Yu L, Bowen J, Gorovsky MA, Allis CD. Phosphorylation of histone H3 is required for proper chromosome condensation and segregation. Cell97(1), 99–109 (1999).
  • Hayakawa T, Haraguchi T, Masumoto H, Hiraoka Y. Cell cycle behavior of human HP1 subtypes: distinct molecular domains of HP1 are required for their centromeric localization during interphase and metaphase. J. Cell Sci.116(Pt 16), 3327–3338 (2003).
  • Noma K, Sugiyama T, Cam H et al. RITS acts in cis to promote RNA interference-mediated transcriptional and post-transcriptional silencing. Nat. Genet.36(11), 1174–1180 (2004).
  • Carmeliet P, Dor Y, Herbert JM et al. Role of HIF-1α in hypoxia-mediated apoptosis, cell proliferation and tumour angiogenesis. Nature394(6692), 485–490 (1998).
  • Nor JE, Christensen J, Mooney DJ, Polverini PJ. Vascular endothelial growth factor (VEGF)-mediated angiogenesis is associated with enhanced endothelial cell survival and induction of Bcl-2 expression. Am. J. Pathol.154(2), 375–384 (1999).
  • Kim MS, Kwon HJ, Lee YM et al. Histone deacetylases induce angiogenesis by negative regulation of tumor suppressor genes. Nat. Med.7(4), 437–443 (2001).
  • Deroanne CF, Bonjean K, Servotte S et al. Histone deacetylases inhibitors as anti-angiogenic agents altering vascular endothelial growth factor signaling. Oncogene21(3), 427–436 (2002).
  • Rossig L, Li H, Fisslthaler B et al. Inhibitors of histone deacetylation downregulate the expression of endothelial nitric oxide synthase and compromise endothelial cell function in vasorelaxation and angiogenesis. Circ. Res.91(9), 837–844 (2002).
  • Kwon HJ, Kim MS, Kim MJ, Nakajima H, Kim KW. Histone deacetylase inhibitor FK228 inhibits tumor angiogenesis. Int. J. Cancer97(3), 290–296 (2002).
  • Pili R, Kruszewski MP, Hager BW, Lantz J, Carducci MA. Combination of phenylbutyrate and 13-cis retinoic acid inhibits prostate tumor growth and angiogenesis. Cancer Res.61(4), 1477–1485 (2001).
  • Qian DZ, Kato Y, Shabbeer S et al. Targeting tumor angiogenesis with histone deacetylase inhibitors: the hydroxamic acid derivative LBH589. Clin. Cancer Res.12(2), 634–642 (2006).
  • Qian DZ, Wang X, Kachhap SK et al. The histone deacetylase inhibitor NVP-LAQ824 inhibits angiogenesis and has a greater antitumor effect in combination with the vascular endothelial growth factor receptor tyrosine kinase inhibitor PTK787/ZK222584. Cancer Res.64(18), 6626–6634 (2004).
  • Huang N, Katz JP, Martin DR, Wu GD. Inhibition of IL-8 gene expression in Caco-2 cells by compounds which induce histone hyperacetylation. Cytokine9(1), 27–36 (1997).
  • Koyama Y, Adachi M, Sekiya M, Takekawa M, Imai K. Histone deacetylase inhibitors suppress IL-2-mediated gene expression prior to induction of apoptosis. Blood96(4), 1490–1495 (2000).
  • Takahashi I, Miyaji H, Yoshida T, Sato S, Mizukami T. Selective inhibition of IL-2 gene expression by trichostatin A, a potent inhibitor of mammalian histone deacetylase. J. Antibiot. (Tokyo)49(5), 453–457 (1996).
  • Blanchard F, Chipoy C. Histone deacetylase inhibitors: new drugs for the treatment of inflammatory diseases? Drug Discov. Today10(3), 197–204 (2005).
  • Dinarello CA. Inhibitors of histone deacetylases as anti-inflammatory drugs. Ernst Schering Res. Found. Workshop56, 45–60 (2006).
  • Camelo S, Iglesias AH, Hwang D et al. Transcriptional therapy with the histone deacetylase inhibitor trichostatin A ameliorates experimental autoimmune encephalomyelitis. J. Neuroimmunol.164(1–2), 10–21 (2005).
  • Chung YL, Lee MY, Wang AJ, Yao LF. A therapeutic strategy uses histone deacetylase inhibitors to modulate the expression of genes involved in the pathogenesis of rheumatoid arthritis. Mol. Ther.8(5), 707–717 (2003).
  • Reddy P, Maeda Y, Hotary K et al. Histone deacetylase inhibitor suberoylanilide hydroxamic acid reduces acute graft-versus-host disease and preserves graft-versus-leukemia effect. Proc. Natl Acad. Sci. USA101(11), 3921–3926 (2004).
  • Brogdon J, Xu Y, Szabo S et al. Histone deacetylase activities are required for innate immune cell control of Th1 but not Th2 effector cell function. Blood109(3), 1123–1130 (2007).
  • Leoni F, Zaliani A, Bertolini G et al. The antitumor histone deacetylase inhibitor suberoylanilide hydroxamic acid exhibits antiinflammatory properties via suppression of cytokines. Proc. Natl Acad. Sci. USA99(5), 2995–3000 (2002).
  • Mishra N, Reilly CM, Brown DR, Ruiz P, Gilkeson GS. Histone deacetylase inhibitors modulate renal disease in the MRL-lpr/lpr mouse. J. Clin. Invest.111(4), 539–552 (2003).
  • Suuronen T, Huuskonen J, Pihlaja R, Kyrylenko S, Salminen A. Regulation of microglial inflammatory response by histone deacetylase inhibitors. J. Neurochem.87(2), 407–416 (2003).
  • Ito K, Lim S, Caramori G et al. A molecular mechanism of action of theophylline: induction of histone deacetylase activity to decrease inflammatory gene expression. Proc. Natl Acad. Sci. USA99(13), 8921–8926 (2002).
  • Blanchard F, Kinzie E, Wang Y et al. FR901228, an inhibitor of histone deacetylases, increases the cellular responsiveness to IL-6 type cytokines by enhancing the expression of receptor proteins. Oncogene21(41), 6264–6277 (2002).
  • Kouzarides T. Acetylation: a regulatory modification to rival phosphorylation? EMBO J.19(6), 1176–1179 (2000).
  • Polevoda B, Sherman F. The diversity of acetylated proteins. Genome Biol.3(5), reviews0006 (2002).
  • Wang R, Cherukuri P, Luo J. Activation of Stat3 sequence-specific DNA binding and transcription by p300/CREB-binding protein-mediated acetylation. J. Biol. Chem.280(12), 11528–11534 (2005).
  • Yuan ZL, Guan YJ, Chatterjee D, Chin YE. Stat3 dimerization regulated by reversible acetylation of a single lysine residue. Science307(5707), 269–273 (2005).
  • Martinez-Balbas MA, Bauer UM, Nielsen SJ, Brehm A, Kouzarides T. Regulation of E2F1 activity by acetylation. EMBO J.19(4), 662–671 (2000).
  • Marzio G, Wagener C, Gutierrez MI et al. E2F family members are differentially regulated by reversible acetylation. J. Biol. Chem.275(15), 10887–10892 (2000).
  • Gu W, Roeder RG. Activation of p53 sequence-specific DNA binding by acetylation of the p53 C-terminal domain. Cell90(4), 595–606 (1997).
  • Sykes SM, Mellert HS, Holbert MA et al. Acetylation of the p53 DNA-binding domain regulates apoptosis induction. Mol. Cell24(6), 841–851 (2006).
  • Kramer OH, Baus D, Knauer SK et al. Acetylation of Stat1 modulates NF-κB activity. Genes Dev.20(4), 473–485 (2006).
  • Nusinzon I, Horvath CM. Interferon-stimulated transcription and innate antiviral immunity require deacetylase activity and histone deacetylase 1. Proc. Natl Acad. Sci. USA100(25), 14742–14747 (2003).
  • Vries RG, Prudenziati M, Zwartjes C et al. A specific lysine in c-Jun is required for transcriptional repression by E1A and is acetylated by p300. EMBO J.20(21), 6095–6103 (2001).
  • Glozak MA, Sengupta N, Zhang X, Seto E. Acetylation and deacetylation of non-histone proteins. Gene363, 15–23 (2005).
  • Piacentini P, Donadelli M, Costanzo C et al. Trichostatin A enhances the response of chemotherapeutic agents in inhibiting pancreatic cancer cell proliferation. Virchows Arch.448(6), 797–804 (2006).
  • Sato T, Suzuki M, Sato Y, Echigo S, Rikiishi H. Sequence-dependent interaction between cisplatin and histone deacetylase inhibitors in human oral squamous cell carcinoma cells. Int. J. Oncol.28(5), 1233–1241 (2006).
  • Zhang X, Yashiro M, Ren J, Hirakawa K. Histone deacetylase inhibitor, trichostatin A, increases the chemosensitivity of anticancer drugs in gastric cancer cell lines. Oncol. Rep.16(3), 563–568 (2006).
  • Chobanian NH, Greenberg VL, Gass JM et al. Histone deacetylase inhibitors enhance paclitaxel-induced cell death in ovarian cancer cell lines independent of p53 status. Anticancer Res.24(2B), 539–545 (2004).
  • Marchion DC, Bicaku E, Daud AI, Sullivan DM, Munster PN. In vivo synergy between topoisomerase II and histone deacetylase inhibitors: predictive correlates. Mol. Cancer Ther.4(12), 1993–2000 (2005).
  • Kim MS, Blake M, Baek JH et al. Inhibition of histone deacetylase increases cytotoxicity to anticancer drugs targeting DNA. Cancer Res.63(21), 7291–7300 (2003).
  • Johnson CA, Padget K, Austin CA, Turner BM. Deacetylase activity associates with topoisomerase II and is necessary for etoposide-induced apoptosis. J. Biol. Chem.276(7), 4539–4542 (2001).
  • Bevins RL, Zimmer SG. It’s about time: scheduling alters effect of histone deacetylase inhibitors on camptothecin-treated cells. Cancer Res.65(15), 6957–6966 (2005).
  • Acharya MR, Figg WD. Histone deacetylase inhibitor enhances the anti-leukemic activity of an established nucleoside analogue. Cancer Biol. Ther.3(8), 719–720 (2004).
  • Maggio SC, Rosato RR, Kramer LB et al. The histone deacetylase inhibitor MS-275 interacts synergistically with fludarabine to induce apoptosis in human leukemia cells. Cancer Res.64(7), 2590–2600 (2004).
  • Adachi M, Zhang Y, Zhao X et al. Synergistic effect of histone deacetylase inhibitors FK228 and m-carboxycinnamic acid bis-hydroxamide with proteasome inhibitors PSI and PS-341 against gastrointestinal adenocarcinoma cells. Clin. Cancer Res.10(11), 3853–3862 (2004).
  • Bai J, Demirjian A, Sui J, Marasco W, Callery MP. Histone deacetylase inhibitor trichostatin A and proteasome inhibitor PS-341 synergistically induce apoptosis in pancreatic cancer cells. Biochem. Biophys. Res. Commun.348(4), 1245–1253 (2006).
  • Pei XY, Dai Y, Grant S. 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(11), 3839–3852 (2004).
  • Yu C, Rahmani M, Conrad D et al. The proteasome inhibitor bortezomib interacts synergistically with histone deacetylase inhibitors to induce apoptosis in Bcr/Abl+ cells sensitive and resistant to STI571. Blood102(10), 3765–3774 (2003).
  • Coffey DC, Kutko MC, Glick RD et al. Histone deacetylase inhibitors and retinoic acids inhibit growth of human neuroblastoma in vitro. Med. Pediatr. Oncol.35(6), 577–581 (2000).
  • Kuendgen A, Schmid M, Schlenk R et al. The histone deacetylase (HDAC) inhibitor valproic acid as monotherapy or in combination with all-trans retinoic acid in patients with acute myeloid leukemia. Cancer106(1), 112–119 (2006).
  • Kuendgen A, Strupp C, Aivado M et al. Treatment of myelodysplastic syndromes with valproic acid alone or in combination with all-trans retinoic acid. Blood104(5), 1266–1269 (2004).
  • Bug G, Ritter M, Wassmann B et al. Clinical trial of valproic acid and all-trans retinoic acid in patients with poor-risk acute myeloid leukemia. Cancer104(12), 2717–2725 (2005).
  • Massart C, Denais A, Gibassier J. Effect of all-trans retinoic acid and sodium butyrate in vitro and in vivo on thyroid carcinoma xenografts. Anticancer Drugs17(5), 559–563 (2006).
  • 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.21(1), 103–107 (1999).
  • Gore SD, Baylin S, Sugar E et al. Combined DNA methyltransferase and histone deacetylase inhibition in the treatment of myeloid neoplasms. Cancer Res.66(12), 6361–6369 (2006).
  • Hurtubise A, Momparler RL. Effect of histone deacetylase inhibitor LAQ824 on antineoplastic action of 5-aza-2´-deoxycytidine (decitabine) on human breast carcinoma cells. Cancer Chemother. Pharmacol.58(5), 618–625 (2006).
  • Zhu WG, Lakshmanan RR, Beal MD, Otterson GA. DNA methyltransferase inhibition enhances apoptosis induced by histone deacetylase inhibitors. Cancer Res.61(4), 1327–1333 (2001).
  • Camphausen K, Burgan W, Cerra M et al. Enhanced radiation-induced cell killing and prolongation of γH2AX foci expression by the histone deacetylase inhibitor MS-275. Cancer Res.64(1), 316–321 (2004).
  • Flatmark K, Nome RV, Folkvord S et al. Radiosensitization of colorectal carcinoma cell lines by histone deacetylase inhibition. Radiat. Oncol.1(1), 25 (2006).
  • Kim MS, Baek JH, Chakravarty D, Sidransky D, Carrier F. Sensitization to UV-induced apoptosis by the histone deacetylase inhibitor trichostatin A (TSA). Exp. Cell Res.306(1), 94–102 (2005).
  • Zhang Y, Adachi M, Kawamura R et al. Bmf contributes to histone deacetylase inhibitor-mediated enhancing effects on apoptosis after ionizing radiation. Apoptosis11(8), 1349–1357 (2006).
  • Chen Z, Brand NJ, Chen A et al. Fusion between a novel Kruppel-like zinc finger gene and the retinoic acid receptor-α locus due to a variant t(11;17) translocation associated with acute promyelocytic leukaemia. EMBO J.12(3), 1161–1167 (1993).
  • de The H, Lavau C, Marchio A et al. The PML-RAR α fusion mRNA generated by the t(15;17) translocation in acute promyelocytic leukemia encodes a functionally altered RAR. Cell66(4), 675–684 (1991).
  • Grignani F, De Matteis S, Nervi C et al. Fusion proteins of the retinoic acid receptor-α recruit histone deacetylase in promyelocytic leukaemia. Nature391(6669), 815–818 (1998).
  • Ruthardt M, Testa U, Nervi C et al. Opposite effects of the acute promyelocytic leukemia PML-retinoic acid receptor α (RAR α) and PLZF-RAR α fusion proteins on retinoic acid signalling. Mol. Cell. Biol.17(8), 4859–4869 (1997).
  • Senyuk V, Chakraborty S, Mikhail FM et al. The leukemia-associated transcription repressor AML1/MDS1/EVI1 requires CtBP to induce abnormal growth and differentiation of murine hematopoietic cells. Oncogene21(20), 3232–3240 (2002).
  • Minucci S, Horn V, Bhattacharyya N et al. A histone deacetylase inhibitor potentiates retinoid receptor action in embryonal carcinoma cells. Proc. Natl Acad. Sci. USA94(21), 11295–11300 (1997).
  • He LZ, Guidez F, Tribioli C et al. Distinct interactions of PML–RARα and PLZF–RARα with co-repressors determine differential responses to RA in APL. Nat. Genet.18(2), 126–135 (1998).
  • Lin RJ, Nagy L, Inoue S et al. Role of the histone deacetylase complex in acute promyelocytic leukaemia. Nature391(6669), 811–814 (1998).
  • Duvic M, Talpur R, Ni X et al. Phase II trial of oral vorinostat (suberoylanilide hydroxamic acid, SAHA) for refractory cutaneous T-cell lymphoma (CTCL). Blood109(1), 31–39 (2007).
  • Kelly WK, Richon VM, O’Connor O et al. Phase I clinical trial of histone deacetylase inhibitor: suberoylanilide hydroxamic acid administered intravenously. Clin. Cancer Res.9(10 Pt 1), 3578–3588 (2003).
  • Cohen LA, Amin S, Marks PA et al. Chemoprevention of carcinogen-induced mammary tumorigenesis by the hybrid polar cytodifferentiation agent, suberanilohydroxamic acid (SAHA). Anticancer Res.19(6B), 4999–5005 (1999).
  • Kelly WK, O’Connor OA, Krug LM et al. Phase I study of an oral histone deacetylase inhibitor, suberoylanilide hydroxamic acid, in patients with advanced cancer. J. Clin. Oncol.23(17), 3923–3931 (2005).
  • O’Connor OA, Heaney ML, Schwartz L et al. Clinical experience with intravenous and oral formulations of the novel histone deacetylase inhibitor suberoylanilide hydroxamic acid in patients with advanced hematologic malignancies. J. Clin. Oncol.24(1), 166–173 (2006).
  • Sandor V, Bakke S, Robey RW et al. Phase I trial of the histone deacetylase inhibitor, depsipeptide (FR901228, NSC 630176), in patients with refractory neoplasms. Clin. Cancer Res.8(3), 718–728 (2002).
  • Marshall JL, Rizvi N, Kauh J et al. A Phase I trial of depsipeptide (FR901228) in patients with advanced cancer. J. Exp. Ther. Oncol.2(6), 325–332 (2002).
  • Piekarz RL, Frye AR, Wright JJ et al. Cardiac studies in patients treated with depsipeptide, FK228, in a Phase II trial for T-cell lymphoma. Clin. Cancer Res.12(12), 3762–3773 (2006).
  • Stadler WM, Margolin K, Ferber S, McCulloch W, Thompson JA. A Phase II study of depsipeptide in refractory metastatic renal cell cancer. Clin. Genitourin. Cancer5(1), 57–60 (2006).
  • Shah MH, Binkley P, Chan K et al. Cardiotoxicity of histone deacetylase inhibitor depsipeptide in patients with metastatic neuroendocrine tumors. Clin. Cancer Res.12(13), 3997–4003 (2006).
  • Fouladi M, Furman WL, Chin T et al. Phase I study of depsipeptide in pediatric patients with refractory solid tumors: a Children’s Oncology Group report. J. Clin. Oncol.24(22), 3678–3685 (2006).
  • Gottlicher M. Valproic acid: an old drug newly discovered as inhibitor of histone deacetylases. Ann. Hematol.83(Suppl. 1), S91–S92 (2004).
  • Gottlicher M, Minucci S, Zhu P et al. Valproic acid defines a novel class of HDAC inhibitors inducing differentiation of transformed cells. EMBO J.20(24), 6969–6978 (2001).
  • Gurvich N, Tsygankova OM, Meinkoth JL, Klein PS. Histone deacetylase is a target of valproic acid-mediated cellular differentiation. Cancer Res.64(3), 1079–1086 (2004).
  • Williams RS, Cheng L, Mudge AW, Harwood AJ. A common mechanism of action for three mood-stabilizing drugs. Nature417(6886), 292–295 (2002).
  • Phiel CJ, Zhang F, Huang EY et al. Histone deacetylase is a direct target of valproic acid, a potent anticonvulsant, mood stabilizer, and teratogen. J. Biol. Chem.276(39), 36734–36741 (2001).
  • Yang H, Hoshino K, Sanchez-Gonzalez B, Kantarjian H, Garcia-Manero G. Antileukemia activity of the combination of 5-aza-2´-deoxycytidine with valproic acid. Leuk. Res.29(7), 739–748 (2005).
  • Garcia-Manero G, Kantarjian HM, Sanchez-Gonzalez B et al. Phase I/II study of the combination of 5-aza-2´-deoxycytidine with valproic acid in patients with leukemia. Blood108(10), 3271–3279 (2006).
  • Atadja P, Hsu M, Kwon P et al. Molecular and cellular basis for the anti-proliferative effects of the HDAC inhibitor LAQ824. Novartis Found. Symp.259, 249–266; discussion 266–268, 285–288 (2004).
  • Bali P, Pranpat M, Bradner J et al. Inhibition of histone deacetylase 6 acetylates and disrupts the chaperone function of heat shock protein 90: a novel basis for antileukemia activity of histone deacetylase inhibitors. J. Biol. Chem.280(29), 26729–26734 (2005).
  • Giles F, Fischer T, Cortes J et al. A Phase I study of intravenous LBH589, a novel cinnamic hydroxamic acid analogue histone deacetylase inhibitor, in patients with refractory hematologic malignancies. Clin. Cancer Res.12(15), 4628–4635 (2006).
  • Ryan QC, Headlee D, Acharya M et al. Phase I and pharmacokinetic study of MS-275, a histone deacetylase inhibitor, in patients with advanced and refractory solid tumors or lymphoma. J. Clin. Oncol.23(17), 3912–3922 (2005).
  • 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.7(8), 2292–2300 (2001).
  • Plumb JA, Finn PW, Williams RJ et al. Pharmacodynamic response and inhibition of growth of human tumor xenografts by the novel histone deacetylase inhibitor PXD101. Mol. Cancer Ther.2(8), 721–728 (2003).
  • Qian X, LaRochelle WJ, Ara G et al. Activity of PXD101, a histone deacetylase inhibitor, in preclinical ovarian cancer studies. Mol. Cancer Ther.5(8), 2086–2095 (2006).
  • Munster PN, Marchion DC, Bicaku E et al. Phase I trial of histone deacetylase inhibition by valproic acid (VPA) followed by the topoisomerase II inhibitor epirubicin in advanced solid tumors: a clinical and translational study. J. Clin. Oncol. (2007) (In Press).
  • Laherty CD, Yang WM, Sun JM et al. Histone deacetylases associated with the mSin3 corepressor mediate mad transcriptional repression. Cell89(3), 349–356 (1997).
  • Hassig CA, Fleischer TC, Billin AN, Schreiber SL, Ayer DE. Histone deacetylase activity is required for full transcriptional repression by mSin3A. Cell89(3), 341–347 (1997).
  • Nagy L, Kao HY, Chakravarti D et al. Nuclear receptor repression mediated by a complex containing SMRT, mSin3A, and histone deacetylase. Cell89(3), 373–380 (1997).
  • Humphrey GW, Wang Y, Russanova VR et al. Stable histone deacetylase complexes distinguished by the presence of SANT domain proteins CoREST/kiaa0071 and Mta-L1. J. Biol. Chem.276(9), 6817–6824 (2001).
  • You A, Tong JK, Grozinger CM, Schreiber SL. CoREST is an integral component of the CoREST–human histone deacetylase complex. Proc. Natl Acad. Sci. USA98(4), 1454–1458 (2001).
  • Glaser KB, Li J, Staver MJ et al. Role of class I and class II histone deacetylases in carcinoma cells using siRNA. Biochem. Biophys. Res. Commun.310(2), 529–536 (2003).
  • Lehrmann H, Pritchard LL, Harel-Bellan A. Histone acetyltransferases and deacetylases in the control of cell proliferation and differentiation. Adv. Cancer Res.86, 41–65 (2002).
  • Calonghi N, Cappadone C, Pagnotta E et al. Histone deacetylase 1: a target of 9-hydroxystearic acid in the inhibition of cell growth in human colon cancer. J. Lipid Res.46(8), 1596–1603 (2005).
  • Lagger G, O’Carroll D, Rembold M et al. Essential function of histone deacetylase 1 in proliferation control and CDK inhibitor repression. EMBO J.21(11), 2672–2681 (2002).
  • Stadler JA, Shkumatava A, Norton WH et al. Histone deacetylase 1 is required for cell cycle exit and differentiation in the zebrafish retina. Dev. Dyn.233(3), 883–889 (2005).
  • Huang BH, Laban M, Leung CH et al. Inhibition of histone deacetylase 2 increases apoptosis and p21Cip1/WAF1 expression, independent of histone deacetylase 1. Cell Death Differ.12(4), 395–404 (2005).
  • Hrzenjak A, Moinfar F, Kremser ML et al. Valproate inhibition of histone deacetylase 2 affects differentiation and decreases proliferation of endometrial stromal sarcoma cells. Mol. Cancer Ther.5(9), 2203–2210 (2006).
  • Wilson AJ, Byun DS, Popova N et al. Histone deacetylase 3 (HDAC3) and other class I HDACs regulate colon cell maturation and p21 expression and are deregulated in human colon cancer. J. Biol. Chem.281(19), 13548–13558 (2006).
  • Fischle W, Dequiedt F, Fillion M et al. Human HDAC7 histone deacetylase activity is associated with HDAC3 in vivo. J. Biol. Chem.276(38), 35826–35835 (2001).
  • Fischle W, Dequiedt F, Hendzel MJ et al. Enzymatic activity associated with class II HDACs is dependent on a multiprotein complex containing HDAC3 and SMRT/N-CoR. Mol. Cell9(1), 45–57 (2002).
  • Hug BA. HDAC4: a corepressor controlling bone development. Cell119(4), 448–449 (2004).
  • Dressel U, Bailey PJ, Wang SC et al. A dynamic role for HDAC7 in MEF2-mediated muscle differentiation. J. Biol. Chem.276(20), 17007–17013 (2001).
  • McKinsey TA, Zhang CL, Lu J, Olson EN. Signal-dependent nuclear export of a histone deacetylase regulates muscle differentiation. Nature408(6808), 106–111 (2000).
  • Miska EA, Langley E, Wolf D et al. Differential localization of HDAC4 orchestrates muscle differentiation. Nucleic Acids Res.29(16), 3439–3447 (2001).
  • Hubbert C, Guardiola A, Shao R et al. HDAC6 is a microtubule-associated deacetylase. Nature417(6887), 455–458 (2002).
  • Kovacs JJ, Hubbert C, Yao TP. The HDAC complex and cytoskeleton. Novartis Found. Symp.259, 170–177; discussion 178–181, 223–225 (2004).
  • Matsuyama A, Shimazu T, Sumida Y et al. In vivo destabilization of dynamic microtubules by HDAC6-mediated deacetylation. EMBO J.21(24), 6820–6831 (2002).
  • Chang S, Young BD, Li S et al. Histone deacetylase 7 maintains vascular integrity by repressing matrix metalloproteinase 10. Cell126(2), 321–334 (2006).
  • Waltregny D, De Leval L, Glenisson W et al. Expression of histone deacetylase 8, a class I histone deacetylase, is restricted to cells showing smooth muscle differentiation in normal human tissues. Am. J. Pathol.165(2), 553–564 (2004).
  • Waltregny D, Glenisson W, Tran SL et al. Histone deacetylase HDAC8 associates with smooth muscle α-actin and is essential for smooth muscle cell contractility. FASEB J.19(8), 966–968 (2005).
  • Kramer OH, Zhu P, Ostendorff HP et al. The histone deacetylase inhibitor valproic acid selectively induces proteasomal degradation of HDAC2. EMBO J.22(13), 3411–3420 (2003).
  • Hu E, Dul E, Sung CM et al. Identification of novel isoform-selective inhibitors within class I histone deacetylases. J. Pharmacol. Exp. Ther.307(2), 720–728 (2003).
  • Ropero S, Fraga MF, Ballestar E et al. A truncating mutation of HDAC2 in human cancers confers resistance to histone deacetylase inhibition. Nat. Genet.38(5), 566–569 (2006).

Website

  • US National Institutes of Health Clinical Trials www.clinicaltrials.gov

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.