Skip to main content
Top
Published in: Investigational New Drugs 1/2010

Open Access 01-12-2010 | SPECIAL ISSUE ARTICLE

Histone deacetylase inhibitors: potential targets responsible for their anti-cancer effect

Authors: Michael Dickinson, Ricky W. Johnstone, H. Miles Prince

Published in: Investigational New Drugs | Special Issue 1/2010

Login to get access

Summary

The histone deacetylase inhibitors (HDACi) have demonstrated anticancer efficacy across a range of malignancies, most impressively in the hematological cancers. It is uncertain whether this clinical efficacy is attributable predominantly to their ability to induce apoptosis and differentiation in the cancer cell, or to their ability to prime the cell to other pro-death stimuli such as those from the immune system. HDACi-induced apoptosis occurs through altered expression of genes encoding proteins in both intrinsic and extrinsic apoptotic pathways; through effects on the proteasome/aggresome systems; through the production of reactive oxygen species, possibly by directly inducing DNA damage; and through alterations in the tumor microenvironment. In addition HDACi increase the immunogenicity of tumor cells and modulate cytokine signaling and potentially T-cell polarization in ways that may contribute the anti-cancer effect in vivo. Here, we provide an overview of current thinking on the mechanisms of HDACi activity, with attention given to the hematological malignancies as well as scientific observations arising from the clinical trials. We also focus on the immune effects of these agents.
Literature
1.
go back to reference Richon VM, Sandhoff TW, Rifkind RA, Marks PA (2000) Histone deacetylase inhibitor selectively induces p21WAF1 expression and gene-associated histone acetylation. Proc Natl Acad Sci USA 97(18):10014–10019PubMedCrossRef Richon VM, Sandhoff TW, Rifkind RA, Marks PA (2000) Histone deacetylase inhibitor selectively induces p21WAF1 expression and gene-associated histone acetylation. Proc Natl Acad Sci USA 97(18):10014–10019PubMedCrossRef
2.
go back to reference Archer SY, Meng S, Shei A, Hodin RA (1998) p21(WAF1) is required for butyrate-mediated growth inhibition of human colon cancer cells. Proc Natl Acad Sci USA 95(12):6791–6796PubMedCrossRef Archer SY, Meng S, Shei A, Hodin RA (1998) p21(WAF1) is required for butyrate-mediated growth inhibition of human colon cancer cells. Proc Natl Acad Sci USA 95(12):6791–6796PubMedCrossRef
3.
go back to reference Sasakawa Y, Naoe Y, Inoue T, Sasakawa T, Matsuo M, Manda T, Mutoh S (2002) Effects of FK228, a novel histone deacetylase inhibitor, on human lymphoma U-937 cells in vitro and in vivo. Biochem Pharmacol 64(7):1079–1090PubMedCrossRef Sasakawa Y, Naoe Y, Inoue T, Sasakawa T, Matsuo M, Manda T, Mutoh S (2002) Effects of FK228, a novel histone deacetylase inhibitor, on human lymphoma U-937 cells in vitro and in vivo. Biochem Pharmacol 64(7):1079–1090PubMedCrossRef
5.
go back to reference Carew JS, Nawrocki ST, Kahue CN, Zhang H, Yang C, Chung L, Houghton JA, Huang P, Giles FJ, Cleveland JL (2007) Targeting autophagy augments the anticancer activity of the histone deacetylase inhibitor SAHA to overcome Bcr-Abl-mediated drug resistance. Blood 110(1):313–322PubMedCrossRef Carew JS, Nawrocki ST, Kahue CN, Zhang H, Yang C, Chung L, Houghton JA, Huang P, Giles FJ, Cleveland JL (2007) Targeting autophagy augments the anticancer activity of the histone deacetylase inhibitor SAHA to overcome Bcr-Abl-mediated drug resistance. Blood 110(1):313–322PubMedCrossRef
6.
go back to reference de Ruijter AJM, van Gennip AH, Caron HN, Kemp S, van Kuilenburg ABP (2003) Histone deacetylases (HDACs): characterization of the classical HDAC family. Biochem J 370(Pt 3):737–749. doi:10.1042/BJ20021321 PubMed de Ruijter AJM, van Gennip AH, Caron HN, Kemp S, van Kuilenburg ABP (2003) Histone deacetylases (HDACs): characterization of the classical HDAC family. Biochem J 370(Pt 3):737–749. doi:10.​1042/​BJ20021321 PubMed
7.
go back to reference Bolden JE, Peart MJ, Johnstone RW (2006) Anticancer activities of histone deacetylase inhibitors. Nat Rev 5(9):769–784CrossRef Bolden JE, Peart MJ, Johnstone RW (2006) Anticancer activities of histone deacetylase inhibitors. Nat Rev 5(9):769–784CrossRef
8.
go back to reference Dovey OM, Foster CT, Cowley SM (2010) Emphasizing the positive: a role for histone deacetylases in transcriptional activation. Cell Cycle 9(14):2700–2701PubMedCrossRef Dovey OM, Foster CT, Cowley SM (2010) Emphasizing the positive: a role for histone deacetylases in transcriptional activation. Cell Cycle 9(14):2700–2701PubMedCrossRef
12.
go back to reference Hubbert C, Guardiola A, Shao R, Kawaguchi Y, Ito A, Nixon A, Yoshida M, Wang X-F, Yao T-P (2002) HDAC6 is a microtubule-associated deacetylase. Nature 417(6887):455–458. doi:10.1038/417455a PubMedCrossRef Hubbert C, Guardiola A, Shao R, Kawaguchi Y, Ito A, Nixon A, Yoshida M, Wang X-F, Yao T-P (2002) HDAC6 is a microtubule-associated deacetylase. Nature 417(6887):455–458. doi:10.​1038/​417455a PubMedCrossRef
14.
go back to reference Insinga A, Monestiroli S, Ronzoni S, Gelmetti V, Marchesi F, Viale A, Altucci L, Nervi C, Minucci S, Pelicci PG (2005) Inhibitors of histone deacetylases induce tumor-selective apoptosis through activation of the death receptor pathway. Nat Med 11(1):71–76. doi:10.1038/nm1160 PubMedCrossRef Insinga A, Monestiroli S, Ronzoni S, Gelmetti V, Marchesi F, Viale A, Altucci L, Nervi C, Minucci S, Pelicci PG (2005) Inhibitors of histone deacetylases induce tumor-selective apoptosis through activation of the death receptor pathway. Nat Med 11(1):71–76. doi:10.​1038/​nm1160 PubMedCrossRef
15.
go back to reference Rosato RR, Almenara JA, Dai Y, Grant S (2003) 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):1273–1284PubMed Rosato RR, Almenara JA, Dai Y, Grant S (2003) 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):1273–1284PubMed
16.
go back to reference Nakata S, Yoshida T, Horinaka M, Shiraishi T, Wakada M, Sakai T (2004) Histone deacetylase inhibitors upregulate death receptor 5/TRAIL-R2 and sensitize apoptosis induced by TRAIL/APO2-L in human malignant tumor cells. Oncogene 23(37):6261–6271. doi:10.1038/sj.onc.1207830 PubMedCrossRef Nakata S, Yoshida T, Horinaka M, Shiraishi T, Wakada M, Sakai T (2004) Histone deacetylase inhibitors upregulate death receptor 5/TRAIL-R2 and sensitize apoptosis induced by TRAIL/APO2-L in human malignant tumor cells. Oncogene 23(37):6261–6271. doi:10.​1038/​sj.​onc.​1207830 PubMedCrossRef
17.
go back to reference Fandy TE, Shankar S, Ross DD, Sausville E, Srivastava RK (2005) Interactive effects of HDAC inhibitors and TRAIL on apoptosis are associated with changes in mitochondrial functions and expressions of cell cycle regulatory genes in multiple myeloma. Neoplasia 7(7):646–657PubMedCrossRef Fandy TE, Shankar S, Ross DD, Sausville E, Srivastava RK (2005) Interactive effects of HDAC inhibitors and TRAIL on apoptosis are associated with changes in mitochondrial functions and expressions of cell cycle regulatory genes in multiple myeloma. Neoplasia 7(7):646–657PubMedCrossRef
18.
go back to reference Nebbioso A, Clarke N, Voltz E, Germain E, Ambrosino C, Bontempo P, Alvarez R, Schiavone EM, Ferrara F, Bresciani F, Weisz A, de Lera AR, Gronemeyer H, Altucci L (2005) Tumor-selective action of HDAC inhibitors involves TRAIL induction in acute myeloid leukemia cells. Nat Med 11(1):77–84PubMedCrossRef Nebbioso A, Clarke N, Voltz E, Germain E, Ambrosino C, Bontempo P, Alvarez R, Schiavone EM, Ferrara F, Bresciani F, Weisz A, de Lera AR, Gronemeyer H, Altucci L (2005) Tumor-selective action of HDAC inhibitors involves TRAIL induction in acute myeloid leukemia cells. Nat Med 11(1):77–84PubMedCrossRef
19.
go back to reference Klener P, Molinsky J, Simonova T, Necas E, Andera L, Zivny J (2008) Acquired resistance to tumor necrosis factor-related apoptosis- inducing ligand (TRAIL): histone deacetylase inhibitors resensitize TRAIL-resistant Jurkat acute lymphocytic leukemia cells. ASH Annu Meet Abstr 112(11):5032 EP - Klener P, Molinsky J, Simonova T, Necas E, Andera L, Zivny J (2008) Acquired resistance to tumor necrosis factor-related apoptosis- inducing ligand (TRAIL): histone deacetylase inhibitors resensitize TRAIL-resistant Jurkat acute lymphocytic leukemia cells. ASH Annu Meet Abstr 112(11):5032 EP -
20.
go back to reference MacFarlane M, Harper N, Snowden RT, Dyer MJ, Barnett GA, Pringle JH, Cohen GM (2002) Mechanisms of resistance to TRAIL-induced apoptosis in primary B cell chronic lymphocytic leukaemia. Oncogene 21(44):6809–6818. doi:10.1038/sj.onc.1205853 PubMedCrossRef MacFarlane M, Harper N, Snowden RT, Dyer MJ, Barnett GA, Pringle JH, Cohen GM (2002) Mechanisms of resistance to TRAIL-induced apoptosis in primary B cell chronic lymphocytic leukaemia. Oncogene 21(44):6809–6818. doi:10.​1038/​sj.​onc.​1205853 PubMedCrossRef
21.
go back to reference Inoue S, Harper N, Walewska R, Dyer MJS, Cohen GM (2009) Enhanced Fas-associated death domain recruitment by histone deacetylase inhibitors is critical for the sensitization of chronic lymphocytic leukemia cells to TRAIL-induced apoptosis. Mol Cancer Ther 8(11):3088–3097. doi:10.1158/1535-7163.MCT-09-0451 PubMedCrossRef Inoue S, Harper N, Walewska R, Dyer MJS, Cohen GM (2009) Enhanced Fas-associated death domain recruitment by histone deacetylase inhibitors is critical for the sensitization of chronic lymphocytic leukemia cells to TRAIL-induced apoptosis. Mol Cancer Ther 8(11):3088–3097. doi:10.​1158/​1535-7163.​MCT-09-0451 PubMedCrossRef
22.
go back to reference Inoue S, MacFarlane M, Harper N, Wheat LMC, Dyer MJS, Cohen GM (2004) Histone deacetylase inhibitors potentiate TNF-related apoptosis-inducing ligand (TRAIL)-induced apoptosis in lymphoid malignancies. Cell Death Differ 11(Suppl 2):S193–S206. doi:10.1038/sj.cdd.4401535 PubMedCrossRef Inoue S, MacFarlane M, Harper N, Wheat LMC, Dyer MJS, Cohen GM (2004) Histone deacetylase inhibitors potentiate TNF-related apoptosis-inducing ligand (TRAIL)-induced apoptosis in lymphoid malignancies. Cell Death Differ 11(Suppl 2):S193–S206. doi:10.​1038/​sj.​cdd.​4401535 PubMedCrossRef
24.
go back to reference Guo F, Sigua C, Tao J, Bali P, George P, Li Y, Wittmann S, Moscinski L, Atadja P, Bhalla K (2004) Cotreatment with histone deacetylase inhibitor LAQ824 enhances Apo-2L/tumor necrosis factor-related apoptosis inducing ligand-induced death inducing signaling complex activity and apoptosis of human acute leukemia cells. Cancer Res 64(7):2580–2589PubMedCrossRef Guo F, Sigua C, Tao J, Bali P, George P, Li Y, Wittmann S, Moscinski L, Atadja P, Bhalla K (2004) Cotreatment with histone deacetylase inhibitor LAQ824 enhances Apo-2L/tumor necrosis factor-related apoptosis inducing ligand-induced death inducing signaling complex activity and apoptosis of human acute leukemia cells. Cancer Res 64(7):2580–2589PubMedCrossRef
25.
go back to reference Wood TE, Dalili S, Simpson CD, Sukhai MA, Hurren R, Anyiwe K, Mao X, Suarez Saiz F, Gronda M, Eberhard Y, MacLean N, Ketela T, Reed JC, Moffat J, Minden MD, Batey RA, Schimmer AD (2010) Selective inhibition of histone deacetylases sensitizes malignant cells to death receptor ligands. Mol Cancer Ther 9(1):246–256. doi:10.1158/1535-7163.MCT-09-0495 PubMedCrossRef Wood TE, Dalili S, Simpson CD, Sukhai MA, Hurren R, Anyiwe K, Mao X, Suarez Saiz F, Gronda M, Eberhard Y, MacLean N, Ketela T, Reed JC, Moffat J, Minden MD, Batey RA, Schimmer AD (2010) Selective inhibition of histone deacetylases sensitizes malignant cells to death receptor ligands. Mol Cancer Ther 9(1):246–256. doi:10.​1158/​1535-7163.​MCT-09-0495 PubMedCrossRef
26.
go back to reference Sanda T, Okamoto T, Uchida Y, Nakagawa H, Iida S, Kayukawa S, Suzuki T, Oshizawa T, Suzuki T, Miyata N, Ueda R (2007) Proteome analyses of the growth inhibitory effects of NCH-51, a novel histone deacetylase inhibitor, on lymphoid malignant cells. Leuk Official Journal Leuk Soc Am Leuk Res Fund UK 21(11):2344–2353. doi:10.1038/sj.leu.2404902 Sanda T, Okamoto T, Uchida Y, Nakagawa H, Iida S, Kayukawa S, Suzuki T, Oshizawa T, Suzuki T, Miyata N, Ueda R (2007) Proteome analyses of the growth inhibitory effects of NCH-51, a novel histone deacetylase inhibitor, on lymphoid malignant cells. Leuk Official Journal Leuk Soc Am Leuk Res Fund UK 21(11):2344–2353. doi:10.​1038/​sj.​leu.​2404902
27.
go back to reference Lucas DM, Davis ME, Parthun MR, Mone AP, Kitada S, Cunningham KD, Flax EL, Wickham J, Reed JC, Byrd JC, Grever MR (2004) The histone deacetylase inhibitor MS-275 induces caspase-dependent apoptosis in B-cell chronic lymphocytic leukemia cells. Leuk Official Journal Leuk Soc Am Leuk Res Fund UK 18(7):1207–1214. doi:10.1038/sj.leu.2403388 Lucas DM, Davis ME, Parthun MR, Mone AP, Kitada S, Cunningham KD, Flax EL, Wickham J, Reed JC, Byrd JC, Grever MR (2004) The histone deacetylase inhibitor MS-275 induces caspase-dependent apoptosis in B-cell chronic lymphocytic leukemia cells. Leuk Official Journal Leuk Soc Am Leuk Res Fund UK 18(7):1207–1214. doi:10.​1038/​sj.​leu.​2403388
28.
go back to reference Lucas DM, Alinari L, West DA, Davis ME, Edwards RB, Johnson AJ, Blum KA, Hofmeister CC, Freitas MA, Parthun MR, Wang D, Lehman A, Zhang X, Jarjoura D, Kulp SK, Croce CM, Grever MR, Chen C-S, Baiocchi RA, Byrd JC (2010) The novel deacetylase inhibitor AR-42 demonstrates pre-clinical activity in B-cell malignancies in vitro and in vivo. PLoS ONE 5(6):e10941. doi:10.1371/journal.pone.0010941 PubMedCrossRef Lucas DM, Alinari L, West DA, Davis ME, Edwards RB, Johnson AJ, Blum KA, Hofmeister CC, Freitas MA, Parthun MR, Wang D, Lehman A, Zhang X, Jarjoura D, Kulp SK, Croce CM, Grever MR, Chen C-S, Baiocchi RA, Byrd JC (2010) The novel deacetylase inhibitor AR-42 demonstrates pre-clinical activity in B-cell malignancies in vitro and in vivo. PLoS ONE 5(6):e10941. doi:10.​1371/​journal.​pone.​0010941 PubMedCrossRef
29.
go back to reference Tang R, Faussat A-M, Majdak P, Perrot J-Y, Chaoui D, Legrand O, Marie J-P (2004) Valproic acid inhibits proliferation and induces apoptosis in acute myeloid leukemia cells expressing P-gp and MRP1. Leuk Official Journal Leuk Soc Am Leuk Res Fund UK 18(7):1246–1251. doi:10.1038/sj.leu.2403390 Tang R, Faussat A-M, Majdak P, Perrot J-Y, Chaoui D, Legrand O, Marie J-P (2004) Valproic acid inhibits proliferation and induces apoptosis in acute myeloid leukemia cells expressing P-gp and MRP1. Leuk Official Journal Leuk Soc Am Leuk Res Fund UK 18(7):1246–1251. doi:10.​1038/​sj.​leu.​2403390
30.
go back to reference Morales JC, Ruiz-Magaña MJ, Carranza D, Ortiz-Ferrón G, Ruiz-Ruiz C (2010) HDAC inhibitors with different gene regulation activities depend on the mitochondrial pathway for the sensitization of leukemic T cells to TRAIL-induced apoptosis. Cancer Lett. doi:10.1016/j.canlet.2010.04.029 PubMed Morales JC, Ruiz-Magaña MJ, Carranza D, Ortiz-Ferrón G, Ruiz-Ruiz C (2010) HDAC inhibitors with different gene regulation activities depend on the mitochondrial pathway for the sensitization of leukemic T cells to TRAIL-induced apoptosis. Cancer Lett. doi:10.​1016/​j.​canlet.​2010.​04.​029 PubMed
31.
go back to reference Frew AJ, Lindemann RK, Martin BP, Clarke CJP, Sharkey J, Anthony DA, Banks K-M, Haynes NM, Gangatirkar P, Stanley K, Bolden JE, Takeda K, Yagita H, Secrist JP, Smyth MJ, Johnstone RW (2008) Combination therapy of established cancer using a histone deacetylase inhibitor and a TRAIL receptor agonist. Proc Natl Acad Sci USA 105(32):11317–11322. doi:10.1073/pnas.0801868105 PubMedCrossRef Frew AJ, Lindemann RK, Martin BP, Clarke CJP, Sharkey J, Anthony DA, Banks K-M, Haynes NM, Gangatirkar P, Stanley K, Bolden JE, Takeda K, Yagita H, Secrist JP, Smyth MJ, Johnstone RW (2008) Combination therapy of established cancer using a histone deacetylase inhibitor and a TRAIL receptor agonist. Proc Natl Acad Sci USA 105(32):11317–11322. doi:10.​1073/​pnas.​0801868105 PubMedCrossRef
32.
go back to reference Lindemann RK, Newbold A, Whitecross KF, Cluse LA, Frew AJ, Ellis L, Williams S, Wiegmans AP, Dear AE, Scott CL, Pellegrini M, Wei A, Richon VM, Marks PA, Lowe SW, Smyth MJ, Johnstone RW (2007) Analysis of the apoptotic and therapeutic activities of histone deacetylase inhibitors by using a mouse model of B cell lymphoma. Proc Natl Acad Sci USA 104(19):8071–8076. doi:10.1073/pnas.0702294104 PubMedCrossRef Lindemann RK, Newbold A, Whitecross KF, Cluse LA, Frew AJ, Ellis L, Williams S, Wiegmans AP, Dear AE, Scott CL, Pellegrini M, Wei A, Richon VM, Marks PA, Lowe SW, Smyth MJ, Johnstone RW (2007) Analysis of the apoptotic and therapeutic activities of histone deacetylase inhibitors by using a mouse model of B cell lymphoma. Proc Natl Acad Sci USA 104(19):8071–8076. doi:10.​1073/​pnas.​0702294104 PubMedCrossRef
33.
go back to reference Ellis L, Bots M, Lindemann RK, Bolden JE, Newbold A, Cluse LA, Scott CL, Strasser A, Atadja P, Lowe SW, Johnstone RW (2009) The histone deacetylase inhibitors LAQ824 and LBH589 do not require death receptor signaling or a functional apoptosome to mediate tumor cell death or therapeutic efficacy. Blood 114(2):380–393. doi:10.1182/blood-2008-10-182758 PubMedCrossRef Ellis L, Bots M, Lindemann RK, Bolden JE, Newbold A, Cluse LA, Scott CL, Strasser A, Atadja P, Lowe SW, Johnstone RW (2009) The histone deacetylase inhibitors LAQ824 and LBH589 do not require death receptor signaling or a functional apoptosome to mediate tumor cell death or therapeutic efficacy. Blood 114(2):380–393. doi:10.​1182/​blood-2008-10-182758 PubMedCrossRef
35.
go back to reference Zhang XD, Gillespie SK, Borrow JM, Hersey P (2004) 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):425–435PubMed Zhang XD, Gillespie SK, Borrow JM, Hersey P (2004) 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):425–435PubMed
36.
go back to reference Romanski A, Bacic B, Bug G, Pfeifer H, Gul H, Remiszewski S, Hoelzer D, Atadja P, Ruthardt M, Ottmann OG (2004) Use of a novel histone deacetylase inhibitor to induce apoptosis in cell lines of acute lymphoblastic leukemia. Haematologica 89(4):419–426PubMed Romanski A, Bacic B, Bug G, Pfeifer H, Gul H, Remiszewski S, Hoelzer D, Atadja P, Ruthardt M, Ottmann OG (2004) Use of a novel histone deacetylase inhibitor to induce apoptosis in cell lines of acute lymphoblastic leukemia. Haematologica 89(4):419–426PubMed
37.
go back to reference Mitsiades CS, Mitsiades NS, McMullan CJ, Poulaki V, Shringarpure R, Hideshima T, Akiyama M, Chauhan D, Munshi N, Gu X, Bailey C, Joseph M, Libermann TA, Richon VM, Marks PA, Anderson KC (2004) Transcriptional signature of histone deacetylase inhibition in multiple myeloma: biological and clinical implications. Proc Natl Acad Sci USA 101(2):540–545. doi:10.1073/pnas.2536759100 PubMedCrossRef Mitsiades CS, Mitsiades NS, McMullan CJ, Poulaki V, Shringarpure R, Hideshima T, Akiyama M, Chauhan D, Munshi N, Gu X, Bailey C, Joseph M, Libermann TA, Richon VM, Marks PA, Anderson KC (2004) Transcriptional signature of histone deacetylase inhibition in multiple myeloma: biological and clinical implications. Proc Natl Acad Sci USA 101(2):540–545. doi:10.​1073/​pnas.​2536759100 PubMedCrossRef
38.
go back to reference Wozniak MB, Villuendas R, Bischoff JR, Aparicio CB, Martínez Leal JF, de La Cueva P, Rodriguez ME, Herreros B, Martin-Perez D, Longo MI, Herrera M, Piris MA, Ortiz-Romero PL (2010) Vorinostat interferes with the signaling transduction pathway of T-cell receptor and synergizes with phosphoinositide-3 kinase inhibitors in cutaneous T-cell lymphoma. Haematologica 95(4):613–621. doi:10.3324/haematol.2009.013870 PubMedCrossRef Wozniak MB, Villuendas R, Bischoff JR, Aparicio CB, Martínez Leal JF, de La Cueva P, Rodriguez ME, Herreros B, Martin-Perez D, Longo MI, Herrera M, Piris MA, Ortiz-Romero PL (2010) Vorinostat interferes with the signaling transduction pathway of T-cell receptor and synergizes with phosphoinositide-3 kinase inhibitors in cutaneous T-cell lymphoma. Haematologica 95(4):613–621. doi:10.​3324/​haematol.​2009.​013870 PubMedCrossRef
40.
go back to reference Peart MJ, Smyth GK, van Laar RK, Bowtell DD, Richon VM, Marks PA, Holloway AJ, Johnstone RW (2005) Identification and functional significance of genes regulated by structurally different histone deacetylase inhibitors. Proc Natl Acad Sci USA 102(10):3697–3702. doi:10.1073/pnas.0500369102 PubMedCrossRef Peart MJ, Smyth GK, van Laar RK, Bowtell DD, Richon VM, Marks PA, Holloway AJ, Johnstone RW (2005) Identification and functional significance of genes regulated by structurally different histone deacetylase inhibitors. Proc Natl Acad Sci USA 102(10):3697–3702. doi:10.​1073/​pnas.​0500369102 PubMedCrossRef
41.
go back to reference Buglio D, Georgakis GV, Hanabuchi S, Arima K, Khaskhely NM, Liu Y-J, Younes A (2008) Vorinostat inhibits STAT6-mediated TH2 cytokine and TARC production and induces cell death in Hodgkin lymphoma cell lines. Blood 112(4):1424–1433. doi:10.1182/blood-2008-01-133769 PubMedCrossRef Buglio D, Georgakis GV, Hanabuchi S, Arima K, Khaskhely NM, Liu Y-J, Younes A (2008) Vorinostat inhibits STAT6-mediated TH2 cytokine and TARC production and induces cell death in Hodgkin lymphoma cell lines. Blood 112(4):1424–1433. doi:10.​1182/​blood-2008-01-133769 PubMedCrossRef
42.
go back to reference Fantin VR, Loboda A, Paweletz CP, Hendrickson RC, Pierce JW, Roth JA, Li L, Gooden F, Korenchuk S, Hou XS, Harrington EA, Randolph S, Reilly JF, Ware CM, Kadin ME, Frankel SR, Richon VM (2008) Constitutive activation of signal transducers and activators of transcription predicts vorinostat resistance in cutaneous T-cell lymphoma. Cancer Res 68(10):3785–3794. doi:10.1158/0008-5472.CAN-07-6091 PubMedCrossRef Fantin VR, Loboda A, Paweletz CP, Hendrickson RC, Pierce JW, Roth JA, Li L, Gooden F, Korenchuk S, Hou XS, Harrington EA, Randolph S, Reilly JF, Ware CM, Kadin ME, Frankel SR, Richon VM (2008) Constitutive activation of signal transducers and activators of transcription predicts vorinostat resistance in cutaneous T-cell lymphoma. Cancer Res 68(10):3785–3794. doi:10.​1158/​0008-5472.​CAN-07-6091 PubMedCrossRef
44.
go back to reference Wei Y, Kadia T, Tong W, Zhang M, Jia Y, Yang H, Hu Y, Tambaro FP, Viallet J, O’brien S, Garcia-Manero G (2010) The combination of a histone deacecetylase inhibitor with the BH3-Mimetic GX15-070 has synergistic antileukemia activity by activating both apoptosis and autophagy. Clin Cancer Res. doi:10.1158/1078-0432.CCR-10-0032 Wei Y, Kadia T, Tong W, Zhang M, Jia Y, Yang H, Hu Y, Tambaro FP, Viallet J, O’brien S, Garcia-Manero G (2010) The combination of a histone deacecetylase inhibitor with the BH3-Mimetic GX15-070 has synergistic antileukemia activity by activating both apoptosis and autophagy. Clin Cancer Res. doi:10.​1158/​1078-0432.​CCR-10-0032
45.
go back to reference Maiso P, Carvajal-Vergara X, Ocio EM, López-Pérez R, Mateo G, Gutiérrez N, Atadja P, Pandiella A, San Miguel JF (2006) The histone deacetylase inhibitor LBH589 is a potent antimyeloma agent that overcomes drug resistance. Cancer Res 66(11):5781–5789. doi:10.1158/0008-5472.CAN-05-4186 PubMedCrossRef Maiso P, Carvajal-Vergara X, Ocio EM, López-Pérez R, Mateo G, Gutiérrez N, Atadja P, Pandiella A, San Miguel JF (2006) The histone deacetylase inhibitor LBH589 is a potent antimyeloma agent that overcomes drug resistance. Cancer Res 66(11):5781–5789. doi:10.​1158/​0008-5472.​CAN-05-4186 PubMedCrossRef
46.
go back to reference Chen J, Fiskus W, Eaton K, Fernandez P, Wang Y, Rao R, Lee P, Joshi R, Yang Y, Kolhe R, Balusu R, Chappa P, Natarajan K, Jillella A, Atadja P, Bhalla KN (2009) Cotreatment with BCL-2 antagonist sensitizes cutaneous T-cell lymphoma to lethal action of HDAC7-Nur77-based mechanism. Blood 113(17):4038–4048. doi:10.1182/blood-2008-08-176024 PubMedCrossRef Chen J, Fiskus W, Eaton K, Fernandez P, Wang Y, Rao R, Lee P, Joshi R, Yang Y, Kolhe R, Balusu R, Chappa P, Natarajan K, Jillella A, Atadja P, Bhalla KN (2009) Cotreatment with BCL-2 antagonist sensitizes cutaneous T-cell lymphoma to lethal action of HDAC7-Nur77-based mechanism. Blood 113(17):4038–4048. doi:10.​1182/​blood-2008-08-176024 PubMedCrossRef
47.
go back to reference High LM, Szymanska B, Wilczynska-Kalak U, Barber N, O’Brien R, Khaw SL, Vikstrom IB, Roberts AW, Lock RB (2009) The BH3-mimetic ABT-737 targets the apoptotic machinery in acute lymphoblastic leukemia resulting in synergistic in vitro and in vivo interactions with established drugs. Mol Pharmacol. doi:10.1124/mol.109.060780 PubMed High LM, Szymanska B, Wilczynska-Kalak U, Barber N, O’Brien R, Khaw SL, Vikstrom IB, Roberts AW, Lock RB (2009) The BH3-mimetic ABT-737 targets the apoptotic machinery in acute lymphoblastic leukemia resulting in synergistic in vitro and in vivo interactions with established drugs. Mol Pharmacol. doi:10.​1124/​mol.​109.​060780 PubMed
48.
go back to reference Peart MJ, Tainton KM, Ruefli AA, Dear AE, Sedelies KA, O’Reilly LA, Waterhouse NJ, Trapani JA, Johnstone RW (2003) Novel mechanisms of apoptosis induced by histone deacetylase inhibitors. Cancer Res 63(15):4460–4471PubMed Peart MJ, Tainton KM, Ruefli AA, Dear AE, Sedelies KA, O’Reilly LA, Waterhouse NJ, Trapani JA, Johnstone RW (2003) Novel mechanisms of apoptosis induced by histone deacetylase inhibitors. Cancer Res 63(15):4460–4471PubMed
49.
go back to reference Shao W, Growney JD, Feng Y, O’Connor G, Pu M, Zhu W, Yao Y-M, Kwon P, Fawell S, Atadja P (2010) Activity of deacetylase inhibitor panobinostat (LBH589) in cutaneous T-cell lymphoma models: defining molecular mechanisms of resistance. Int J Cancer J Int Cancer. doi:10.1002/ijc.25218 Shao W, Growney JD, Feng Y, O’Connor G, Pu M, Zhu W, Yao Y-M, Kwon P, Fawell S, Atadja P (2010) Activity of deacetylase inhibitor panobinostat (LBH589) in cutaneous T-cell lymphoma models: defining molecular mechanisms of resistance. Int J Cancer J Int Cancer. doi:10.​1002/​ijc.​25218
50.
go back to reference Ellis L, Pan Y, Smyth GK, George DJ, McCormack C, Williams-Truax R, Mita M, Beck J, Burris H, Ryan G, Atadja P, Butterfoss D, Dugan M, Culver K, Johnstone RW, Prince HM (2008) Histone deacetylase inhibitor panobinostat induces clinical responses with associated alterations in gene expression profiles in cutaneous T-cell lymphoma. Clin Cancer Res 14(14):4500–4510. doi:10.1158/1078-0432.ccr-07-4262 PubMedCrossRef Ellis L, Pan Y, Smyth GK, George DJ, McCormack C, Williams-Truax R, Mita M, Beck J, Burris H, Ryan G, Atadja P, Butterfoss D, Dugan M, Culver K, Johnstone RW, Prince HM (2008) Histone deacetylase inhibitor panobinostat induces clinical responses with associated alterations in gene expression profiles in cutaneous T-cell lymphoma. Clin Cancer Res 14(14):4500–4510. doi:10.​1158/​1078-0432.​ccr-07-4262 PubMedCrossRef
51.
go back to reference Ungerstedt JS, Sowa Y, Xu WS, Shao Y, Dokmanovic M, Perez G, Ngo L, Holmgren A, Jiang X, Marks PA (2005) Role of thioredoxin in the response of normal and transformed cells to histone deacetylase inhibitors. Proc Natl Acad Sci USA 102(3):673–678. doi:10.1073/pnas.0408732102 PubMedCrossRef Ungerstedt JS, Sowa Y, Xu WS, Shao Y, Dokmanovic M, Perez G, Ngo L, Holmgren A, Jiang X, Marks PA (2005) Role of thioredoxin in the response of normal and transformed cells to histone deacetylase inhibitors. Proc Natl Acad Sci USA 102(3):673–678. doi:10.​1073/​pnas.​0408732102 PubMedCrossRef
52.
go back to reference Rosato RR, Almenara JA, Grant S (2003) The histone deacetylase inhibitor MS-275 promotes differentiation or apoptosis in human leukemia cells through a process regulated by generation of reactive oxygen species and induction of p21CIP1/WAF1 1. Cancer Res 63(13):3637–3645PubMed Rosato RR, Almenara JA, Grant S (2003) The histone deacetylase inhibitor MS-275 promotes differentiation or apoptosis in human leukemia cells through a process regulated by generation of reactive oxygen species and induction of p21CIP1/WAF1 1. Cancer Res 63(13):3637–3645PubMed
53.
go back to reference Rosato RR, Maggio SC, Almenara JA, Payne SG, Atadja P, Spiegel S, Dent P, Grant S (2006) 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. doi:10.1124/mol.105.017145 PubMed Rosato RR, Maggio SC, Almenara JA, Payne SG, Atadja P, Spiegel S, Dent P, Grant S (2006) 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. doi:10.​1124/​mol.​105.​017145 PubMed
54.
go back to reference Rosato RR, Kolla SS, Hock SK, Almenara JA, Patel A, Amin S, Atadja P, Fisher PB, Dent P, Grant S (2010) Histone deacetylase inhibitors activate NF-kappaB in human leukemia cells through an ATM/NEMO-related pathway. J Biol Chem 285(13):10064–10077. doi:10.1074/jbc.M109.095208 PubMedCrossRef Rosato RR, Kolla SS, Hock SK, Almenara JA, Patel A, Amin S, Atadja P, Fisher PB, Dent P, Grant S (2010) Histone deacetylase inhibitors activate NF-kappaB in human leukemia cells through an ATM/NEMO-related pathway. J Biol Chem 285(13):10064–10077. doi:10.​1074/​jbc.​M109.​095208 PubMedCrossRef
55.
go back to reference Ruefli AA, Ausserlechner MJ, Bernhard D, Sutton VR, Tainton KM, Kofler R, Smyth MJ, Johnstone RW (2001) The histone deacetylase inhibitor and chemotherapeutic agent suberoylanilide hydroxamic acid (SAHA) induces a cell-death pathway characterized by cleavage of bid and production of reactive oxygen species. Proc Natl Acad Sci USA 98(19):10833–10838. doi:10.1073/pnas.191208598 PubMedCrossRef Ruefli AA, Ausserlechner MJ, Bernhard D, Sutton VR, Tainton KM, Kofler R, Smyth MJ, Johnstone RW (2001) The histone deacetylase inhibitor and chemotherapeutic agent suberoylanilide hydroxamic acid (SAHA) induces a cell-death pathway characterized by cleavage of bid and production of reactive oxygen species. Proc Natl Acad Sci USA 98(19):10833–10838. doi:10.​1073/​pnas.​191208598 PubMedCrossRef
56.
go back to reference Hu Y, Lu W, Chen G, Zhang H, Jia Y, Wei Y, Yang H, Zhang W, Fiskus W, Bhalla K, Keating M, Huang P, Garcia-Manero G (2010) Overcoming resistance to histone deacetylase inhibitors in human leukemia with the redox modulating compound {beta}-phenylethyl isothiocyanate. Blood. doi:10.1182/blood-2009-11-256354 Hu Y, Lu W, Chen G, Zhang H, Jia Y, Wei Y, Yang H, Zhang W, Fiskus W, Bhalla K, Keating M, Huang P, Garcia-Manero G (2010) Overcoming resistance to histone deacetylase inhibitors in human leukemia with the redox modulating compound {beta}-phenylethyl isothiocyanate. Blood. doi:10.​1182/​blood-2009-11-256354
57.
go back to reference Butler LM, Zhou X, Xu WS, Scher HI, Rifkind RA, Marks PA, Richon VM (2002) The histone deacetylase inhibitor SAHA arrests cancer cell growth, up-regulates thioredoxin-binding protein-2, and down-regulates thioredoxin. Proc Natl Acad Sci USA 99(18):11700–11705. doi:10.1073/pnas.182372299 PubMedCrossRef Butler LM, Zhou X, Xu WS, Scher HI, Rifkind RA, Marks PA, Richon VM (2002) The histone deacetylase inhibitor SAHA arrests cancer cell growth, up-regulates thioredoxin-binding protein-2, and down-regulates thioredoxin. Proc Natl Acad Sci USA 99(18):11700–11705. doi:10.​1073/​pnas.​182372299 PubMedCrossRef
59.
go back to reference Davenport EL, Morgan GJ, Davies FE (2008) Untangling the unfolded protein response. Cell Cycle 7(7):865–869PubMedCrossRef Davenport EL, Morgan GJ, Davies FE (2008) Untangling the unfolded protein response. Cell Cycle 7(7):865–869PubMedCrossRef
61.
go back to reference Davenport EL, Moore HE, Dunlop AS, Sharp SY, Workman P, Morgan GJ, Davies FE (2007) Heat shock protein inhibition is associated with activation of the unfolded protein response pathway in myeloma plasma cells. Blood 110(7):2641–2649PubMedCrossRef Davenport EL, Moore HE, Dunlop AS, Sharp SY, Workman P, Morgan GJ, Davies FE (2007) Heat shock protein inhibition is associated with activation of the unfolded protein response pathway in myeloma plasma cells. Blood 110(7):2641–2649PubMedCrossRef
62.
go back to reference Kopito RR (2003) The missing linker: an unexpected role for a histone deacetylase. Mol Cell 12(6):1349–1351PubMedCrossRef Kopito RR (2003) The missing linker: an unexpected role for a histone deacetylase. Mol Cell 12(6):1349–1351PubMedCrossRef
63.
go back to reference Kawaguchi Y, Kovacs JJ, McLaurin A, Vance JM, Ito A, Yao TP (2003) The deacetylase HDAC6 regulates aggresome formation and cell viability in response to misfolded protein stress. Cell 115(6):727–738PubMedCrossRef Kawaguchi Y, Kovacs JJ, McLaurin A, Vance JM, Ito A, Yao TP (2003) The deacetylase HDAC6 regulates aggresome formation and cell viability in response to misfolded protein stress. Cell 115(6):727–738PubMedCrossRef
64.
go back to reference Zhao Z, Xu H, Gong W (2009) Histone Deacetylase 6 (HDAC6) is an Independent Deacetylase for alpha-Tubulin. Protein Pept Lett Zhao Z, Xu H, Gong W (2009) Histone Deacetylase 6 (HDAC6) is an Independent Deacetylase for alpha-Tubulin. Protein Pept Lett
65.
go back to reference Hideshima T, Bradner JE, Wong J, Chauhan D, Richardson P, Schreiber SL, Anderson KC (2005) Small-molecule inhibition of proteasome and aggresome function induces synergistic antitumor activity in multiple myeloma. Proc Natl Acad Sci USA 102(24):8567–8572PubMedCrossRef Hideshima T, Bradner JE, Wong J, Chauhan D, Richardson P, Schreiber SL, Anderson KC (2005) Small-molecule inhibition of proteasome and aggresome function induces synergistic antitumor activity in multiple myeloma. Proc Natl Acad Sci USA 102(24):8567–8572PubMedCrossRef
67.
go back to reference Bali P, Pranpat M, Bradner J, Balasis M, Fiskus W, Guo F, Rocha K, Kumaraswamy S, Boyapalle S, Atadja P, Seto E, Bhalla K (2005) 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. doi:10.1074/jbc.C500186200 PubMedCrossRef Bali P, Pranpat M, Bradner J, Balasis M, Fiskus W, Guo F, Rocha K, Kumaraswamy S, Boyapalle S, Atadja P, Seto E, Bhalla K (2005) 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. doi:10.​1074/​jbc.​C500186200 PubMedCrossRef
68.
go back to reference Rao R, Nalluri S, Fiskus W, Savoie A, Buckley KM, Ha K, Balusu R, Joshi A, Coothankandaswamy V, Tao J, Sotomayor E, Atadja PW, Bhalla KN (2010) Role of C/EBP homologous protein (CHOP) in panobinostat-mediated potentiation of bortezomib-induced lethal ER stress in mantle cell lymphoma cells. Clin Cancer Res. doi:10.1158/1078-0432.CCR-10-0529 Rao R, Nalluri S, Fiskus W, Savoie A, Buckley KM, Ha K, Balusu R, Joshi A, Coothankandaswamy V, Tao J, Sotomayor E, Atadja PW, Bhalla KN (2010) Role of C/EBP homologous protein (CHOP) in panobinostat-mediated potentiation of bortezomib-induced lethal ER stress in mantle cell lymphoma cells. Clin Cancer Res. doi:10.​1158/​1078-0432.​CCR-10-0529
69.
go back to reference Prabhala RH, Pelluru D, Fulciniti M, Prabhala HK, Nanjappa P, Song W, Pai C, Amin S, Tai Y-T, Richardson PG, Ghobrial IM, Treon SP, Daley JF, Anderson KC, Kutok JL, Munshi NC (2010) Elevated IL-17 produced by TH17 cells promotes myeloma cell growth and inhibits immune function in multiple myeloma. Blood 115(26):5385–5392. doi:10.1182/blood-2009-10-246660 PubMedCrossRef Prabhala RH, Pelluru D, Fulciniti M, Prabhala HK, Nanjappa P, Song W, Pai C, Amin S, Tai Y-T, Richardson PG, Ghobrial IM, Treon SP, Daley JF, Anderson KC, Kutok JL, Munshi NC (2010) Elevated IL-17 produced by TH17 cells promotes myeloma cell growth and inhibits immune function in multiple myeloma. Blood 115(26):5385–5392. doi:10.​1182/​blood-2009-10-246660 PubMedCrossRef
70.
go back to reference Rao R, Fiskus W, Yang Y, Lee P, Joshi R, Fernandez P, Mandawat A, Atadja P, Bradner JE, Bhalla K (2008) HDAC6 inhibition enhances 17-AAG–mediated abrogation of hsp90 chaperone function in human leukemia cells. Blood 112(5):1886–1893. doi:10.1182/blood-2008-03-143644 PubMedCrossRef Rao R, Fiskus W, Yang Y, Lee P, Joshi R, Fernandez P, Mandawat A, Atadja P, Bradner JE, Bhalla K (2008) HDAC6 inhibition enhances 17-AAG–mediated abrogation of hsp90 chaperone function in human leukemia cells. Blood 112(5):1886–1893. doi:10.​1182/​blood-2008-03-143644 PubMedCrossRef
71.
go back to reference Catley L, Weisberg E, Kiziltepe T, Tai YT, Hideshima T, Neri P, Tassone P, Atadja P, Chauhan D, Munshi NC, Anderson KC (2006) Aggresome induction by proteasome inhibitor bortezomib and alpha-tubulin hyperacetylation by tubulin deacetylase (TDAC) inhibitor LBH589 are synergistic in myeloma cells. Blood 108(10):3441–3449PubMedCrossRef Catley L, Weisberg E, Kiziltepe T, Tai YT, Hideshima T, Neri P, Tassone P, Atadja P, Chauhan D, Munshi NC, Anderson KC (2006) Aggresome induction by proteasome inhibitor bortezomib and alpha-tubulin hyperacetylation by tubulin deacetylase (TDAC) inhibitor LBH589 are synergistic in myeloma cells. Blood 108(10):3441–3449PubMedCrossRef
72.
go back to reference Fotheringham S, Epping MT, Stimson L, Khan O, Wood V, Pezzella F, Bernards R, La Thangue NB (2009) Genome-wide loss-of-function screen reveals an important role for the proteasome in HDAC inhibitor-induced apoptosis. Cancer Cell 15(1):57–66. doi:10.1016/j.ccr.2008.12.001 PubMedCrossRef Fotheringham S, Epping MT, Stimson L, Khan O, Wood V, Pezzella F, Bernards R, La Thangue NB (2009) Genome-wide loss-of-function screen reveals an important role for the proteasome in HDAC inhibitor-induced apoptosis. Cancer Cell 15(1):57–66. doi:10.​1016/​j.​ccr.​2008.​12.​001 PubMedCrossRef
73.
go back to reference Khan O, Fotheringham S, Wood V, Stimson L, Zhang C, Pezzella F, Duvic M, Kerr DJ, La Thangue NB (2010) HR23B is a biomarker for tumor sensitivity to HDAC inhibitor-based therapy. Proc Natl Acad Sci USA 107(14):6532–6537. doi:10.1073/pnas.0913912107 PubMedCrossRef Khan O, Fotheringham S, Wood V, Stimson L, Zhang C, Pezzella F, Duvic M, Kerr DJ, La Thangue NB (2010) HR23B is a biomarker for tumor sensitivity to HDAC inhibitor-based therapy. Proc Natl Acad Sci USA 107(14):6532–6537. doi:10.​1073/​pnas.​0913912107 PubMedCrossRef
74.
go back to reference Boyault C, Zhang Y, Fritah S, Caron C, Gilquin B, Kwon SH, Garrido C, Yao T-P, Vourc’h C, Matthias P, Khochbin S (2007) HDAC6 controls major cell response pathways to cytotoxic accumulation of protein aggregates. Genes Dev 21(17):2172–2181. doi:10.1101/gad.436407 PubMedCrossRef Boyault C, Zhang Y, Fritah S, Caron C, Gilquin B, Kwon SH, Garrido C, Yao T-P, Vourc’h C, Matthias P, Khochbin S (2007) HDAC6 controls major cell response pathways to cytotoxic accumulation of protein aggregates. Genes Dev 21(17):2172–2181. doi:10.​1101/​gad.​436407 PubMedCrossRef
75.
go back to reference Ocio EM, Vilanova D, Atadja P, Maiso P, Crusoe E, Fernández-Lázaro D, Garayoa M, San-Segundo L, Hernández-Iglesias T, de Álava E, Shao W, Yao Y-M, Pandiella A, San-Miguel JF (2009) In vitro and in vivo rationale for the triple combination of panobinostat (LBH589) and dexamethasone with either bortezomib or lenalidomide in multiple myeloma. Haematologica. doi:10.3324/haematol.2009.015495 PubMed Ocio EM, Vilanova D, Atadja P, Maiso P, Crusoe E, Fernández-Lázaro D, Garayoa M, San-Segundo L, Hernández-Iglesias T, de Álava E, Shao W, Yao Y-M, Pandiella A, San-Miguel JF (2009) In vitro and in vivo rationale for the triple combination of panobinostat (LBH589) and dexamethasone with either bortezomib or lenalidomide in multiple myeloma. Haematologica. doi:10.​3324/​haematol.​2009.​015495 PubMed
76.
go back to reference Buglio D, Mamidipudi V, Khaskhely NM, Brady H, Heise C, Besterman J, Martell RE, MacBeth K, Younes A (2010) The class-I HDAC inhibitor MGCD0103 induces apoptosis in Hodgkin lymphoma cell lines and synergizes with proteasome inhibitors by an HDAC6-independent mechanism. Br J Haematol Epub Buglio D, Mamidipudi V, Khaskhely NM, Brady H, Heise C, Besterman J, Martell RE, MacBeth K, Younes A (2010) The class-I HDAC inhibitor MGCD0103 induces apoptosis in Hodgkin lymphoma cell lines and synergizes with proteasome inhibitors by an HDAC6-independent mechanism. Br J Haematol Epub
77.
go back to reference Harrison S, Quach H, Yuen K, Strayer A, Copeman M, Peinert S, Bishton M, Wolf M, Januszewicz H, Kenealy M, Herbert K, Westerman D, Carney D, Seymour J, Johnstone R, Ritchie D, Prince M (2008) High response rates with the combination of bortezomib, dexamethasone and the pan-histone deacetylase inhibitor romidepsin in patients with relapsed or refractory multiple myeloma in a phase I/II clinical trial. ASH Annu Meet Abstr 112(11):3698 EP - Harrison S, Quach H, Yuen K, Strayer A, Copeman M, Peinert S, Bishton M, Wolf M, Januszewicz H, Kenealy M, Herbert K, Westerman D, Carney D, Seymour J, Johnstone R, Ritchie D, Prince M (2008) High response rates with the combination of bortezomib, dexamethasone and the pan-histone deacetylase inhibitor romidepsin in patients with relapsed or refractory multiple myeloma in a phase I/II clinical trial. ASH Annu Meet Abstr 112(11):3698 EP -
78.
go back to reference Piekarz RL, Frye R, Turner M, Wright JJ, Allen SL, Kirschbaum MH, Zain J, Prince HM, Leonard JP, Geskin LJ, Reeder C, Joske D, Figg WD, Gardner ER, Steinberg SM, Jaffe ES, Stetler-Stevenson M, Lade S, Fojo AT, Bates SE (2009) Phase II multi-institutional trial of the histone deacetylase inhibitor romidepsin as monotherapy for patients with cutaneous T-cell lymphoma. J Clin Oncol 27(32):5410–5417. doi:10.1200/jco.2008.21.6150 PubMedCrossRef Piekarz RL, Frye R, Turner M, Wright JJ, Allen SL, Kirschbaum MH, Zain J, Prince HM, Leonard JP, Geskin LJ, Reeder C, Joske D, Figg WD, Gardner ER, Steinberg SM, Jaffe ES, Stetler-Stevenson M, Lade S, Fojo AT, Bates SE (2009) Phase II multi-institutional trial of the histone deacetylase inhibitor romidepsin as monotherapy for patients with cutaneous T-cell lymphoma. J Clin Oncol 27(32):5410–5417. doi:10.​1200/​jco.​2008.​21.​6150 PubMedCrossRef
80.
go back to reference Gu W, Roeder RG (1997) Activation of p53 sequence-specific DNA binding by acetylation of the p53 C-terminal domain. Cell 90(4):595–606PubMedCrossRef Gu W, Roeder RG (1997) Activation of p53 sequence-specific DNA binding by acetylation of the p53 C-terminal domain. Cell 90(4):595–606PubMedCrossRef
82.
go back to reference Terui T, Murakami K, Takimoto R, Takahashi M, Takada K, Murakami T, Minami S, Matsunaga T, Takayama T, Kato J, Niitsu Y (2003) Induction of PIG3 and NOXA through acetylation of p53 at 320 and 373 lysine residues as a mechanism for apoptotic cell death by histone deacetylase inhibitors. Cancer Res 63(24):8948–8954PubMed Terui T, Murakami K, Takimoto R, Takahashi M, Takada K, Murakami T, Minami S, Matsunaga T, Takayama T, Kato J, Niitsu Y (2003) Induction of PIG3 and NOXA through acetylation of p53 at 320 and 373 lysine residues as a mechanism for apoptotic cell death by histone deacetylase inhibitors. Cancer Res 63(24):8948–8954PubMed
83.
go back to reference Sakaguchi K, Herrera JE, Saito S, Miki T, Bustin M, Vassilev A, Anderson CW, Appella E (1998) DNA damage activates p53 through a phosphorylation-acetylation cascade. Genes Dev 12(18):2831–2841PubMedCrossRef Sakaguchi K, Herrera JE, Saito S, Miki T, Bustin M, Vassilev A, Anderson CW, Appella E (1998) DNA damage activates p53 through a phosphorylation-acetylation cascade. Genes Dev 12(18):2831–2841PubMedCrossRef
85.
go back to reference Juan LJ, Shia WJ, Chen MH, Yang WM, Seto E, Lin YS, Wu CW (2000) Histone deacetylases specifically down-regulate p53-dependent gene activation. J Biol Chem 275(27):20436–20443PubMedCrossRef Juan LJ, Shia WJ, Chen MH, Yang WM, Seto E, Lin YS, Wu CW (2000) Histone deacetylases specifically down-regulate p53-dependent gene activation. J Biol Chem 275(27):20436–20443PubMedCrossRef
87.
go back to reference Palani CD, Beck JF, Sonnemann J (2010) Histone deacetylase inhibitors enhance the anticancer activity of nutlin-3 and induce p53 hyperacetylation and downregulation of MDM2 and MDM4 gene expression. Investig New Drugs. doi:10.1007/s10637-010-9510-7 Palani CD, Beck JF, Sonnemann J (2010) Histone deacetylase inhibitors enhance the anticancer activity of nutlin-3 and induce p53 hyperacetylation and downregulation of MDM2 and MDM4 gene expression. Investig New Drugs. doi:10.​1007/​s10637-010-9510-7
89.
go back to reference Zhao Y, Lu S, Wu L, Chai G, Wang H, Chen Y, Sun J, Yu Y, Zhou W, Zheng Q, Wu M, Otterson GA, Zhu W-G (2006) Acetylation of p53 at lysine 373/382 by the histone deacetylase inhibitor depsipeptide induces expression of p21(Waf1/Cip1). Mol Cell Biol 26(7):2782–2790. doi:10.1128/MCB.26.7.2782-2790.2006 PubMedCrossRef Zhao Y, Lu S, Wu L, Chai G, Wang H, Chen Y, Sun J, Yu Y, Zhou W, Zheng Q, Wu M, Otterson GA, Zhu W-G (2006) Acetylation of p53 at lysine 373/382 by the histone deacetylase inhibitor depsipeptide induces expression of p21(Waf1/Cip1). Mol Cell Biol 26(7):2782–2790. doi:10.​1128/​MCB.​26.​7.​2782-2790.​2006 PubMedCrossRef
91.
go back to reference Sowa Y, Orita T, Minamikawa-Hiranabe S, Mizuno T, Nomura H, Sakai T (1999) Sp3, but not Sp1, mediates the transcriptional activation of the p21/WAF1/Cip1 gene promoter by histone deacetylase inhibitor. Cancer Res 59(17):4266–4270PubMed Sowa Y, Orita T, Minamikawa-Hiranabe S, Mizuno T, Nomura H, Sakai T (1999) Sp3, but not Sp1, mediates the transcriptional activation of the p21/WAF1/Cip1 gene promoter by histone deacetylase inhibitor. Cancer Res 59(17):4266–4270PubMed
92.
go back to reference Saito A, Yamashita T, Mariko Y, Nosaka Y, Tsuchiya K, Ando T, Suzuki T, Tsuruo T, Nakanishi O (1999) A synthetic inhibitor of histone deacetylase, MS-27-275, with marked in vivo antitumor activity against human tumors. Proc Natl Acad Sci USA 96(8):4592–4597PubMedCrossRef Saito A, Yamashita T, Mariko Y, Nosaka Y, Tsuchiya K, Ando T, Suzuki T, Tsuruo T, Nakanishi O (1999) A synthetic inhibitor of histone deacetylase, MS-27-275, with marked in vivo antitumor activity against human tumors. Proc Natl Acad Sci USA 96(8):4592–4597PubMedCrossRef
93.
go back to reference Gui C-Y, Ngo L, Xu WS, Richon VM, Marks PA (2004) Histone deacetylase (HDAC) inhibitor activation of p21WAF1 involves changes in promoter-associated proteins, including HDAC1. Proc Natl Acad Sci USA 101(5):1241–1246. doi:10.1073/pnas.0307708100 PubMedCrossRef Gui C-Y, Ngo L, Xu WS, Richon VM, Marks PA (2004) Histone deacetylase (HDAC) inhibitor activation of p21WAF1 involves changes in promoter-associated proteins, including HDAC1. Proc Natl Acad Sci USA 101(5):1241–1246. doi:10.​1073/​pnas.​0307708100 PubMedCrossRef
94.
go back to reference Johnstone RW, Licht JD (2003) Histone deacetylase inhibitors in cancer therapy: is transcription the primary target? Cancer Cell 4(1):13–18PubMedCrossRef Johnstone RW, Licht JD (2003) Histone deacetylase inhibitors in cancer therapy: is transcription the primary target? Cancer Cell 4(1):13–18PubMedCrossRef
95.
go back to reference Marks PA, Richon VM, Rifkind RA (2000) Histone deacetylase inhibitors: inducers of differentiation or apoptosis of transformed cells. J Natl Cancer Inst 92(15):1210–1216PubMedCrossRef Marks PA, Richon VM, Rifkind RA (2000) Histone deacetylase inhibitors: inducers of differentiation or apoptosis of transformed cells. J Natl Cancer Inst 92(15):1210–1216PubMedCrossRef
96.
go back to reference Sandor V, Senderowicz A, Mertins S, Sackett D, Sausville E, Blagosklonny MV, Bates SE (2000) P21-dependent g(1)arrest with downregulation of cyclin D1 and upregulation of cyclin E by the histone deacetylase inhibitor FR901228. Br J Cancer 83(6):817–825. doi:10.1054/bjoc.2000.1327 PubMedCrossRef Sandor V, Senderowicz A, Mertins S, Sackett D, Sausville E, Blagosklonny MV, Bates SE (2000) P21-dependent g(1)arrest with downregulation of cyclin D1 and upregulation of cyclin E by the histone deacetylase inhibitor FR901228. Br J Cancer 83(6):817–825. doi:10.​1054/​bjoc.​2000.​1327 PubMedCrossRef
98.
99.
go back to reference Burgess AJ, Pavey S, Warrener R, Hunter LJ, Piva TJ, Musgrove EA, Saunders N, Parsons PG, Gabrielli BG (2001) Up-regulation of p21(WAF1/CIP1) by histone deacetylase inhibitors reduces their cytotoxicity. Mol Pharmacol 60(4):828–837PubMed Burgess AJ, Pavey S, Warrener R, Hunter LJ, Piva TJ, Musgrove EA, Saunders N, Parsons PG, Gabrielli BG (2001) Up-regulation of p21(WAF1/CIP1) by histone deacetylase inhibitors reduces their cytotoxicity. Mol Pharmacol 60(4):828–837PubMed
101.
go back to reference Paulson M, Pisharody S, Pan L, Guadagno S, Mui AL, Levy DE (1999) Stat protein transactivation domains recruit p300/CBP through widely divergent sequences. J Biol Chem 274(36):25343–25349PubMedCrossRef Paulson M, Pisharody S, Pan L, Guadagno S, Mui AL, Levy DE (1999) Stat protein transactivation domains recruit p300/CBP through widely divergent sequences. J Biol Chem 274(36):25343–25349PubMedCrossRef
102.
104.
go back to reference Rascle A, Johnston JA, Amati B (2003) Deacetylase activity is required for recruitment of the basal transcription machinery and transactivation by STAT5. Mol Cell Biol 23(12):4162–4173PubMedCrossRef Rascle A, Johnston JA, Amati B (2003) Deacetylase activity is required for recruitment of the basal transcription machinery and transactivation by STAT5. Mol Cell Biol 23(12):4162–4173PubMedCrossRef
107.
go back to reference Chatterjee M, Stühmer T, Herrmann P, Bommert K, Dörken B, Bargou RC (2004) Combined disruption of both the MEK/ERK and the IL-6R/STAT3 pathways is required to induce apoptosis of multiple myeloma cells in the presence of bone marrow stromal cells. Blood 104(12):3712–3721. doi:10.1182/blood-2004-04-1670 PubMedCrossRef Chatterjee M, Stühmer T, Herrmann P, Bommert K, Dörken B, Bargou RC (2004) Combined disruption of both the MEK/ERK and the IL-6R/STAT3 pathways is required to induce apoptosis of multiple myeloma cells in the presence of bone marrow stromal cells. Blood 104(12):3712–3721. doi:10.​1182/​blood-2004-04-1670 PubMedCrossRef
108.
go back to reference Chatterjee M, Jain S, Stühmer T, Andrulis M, Ungethüm U, Kuban R-J, Lorentz H, Bommert K, Topp M, Krämer D, Müller-Hermelink HK, Einsele H, Greiner A, Bargou RC (2007) STAT3 and MAPK signaling maintain overexpression of heat shock proteins 90alpha and beta in multiple myeloma cells, which critically contribute to tumor-cell survival. Blood 109(2):720–728. doi:10.1182/blood-2006-05-024372 PubMedCrossRef Chatterjee M, Jain S, Stühmer T, Andrulis M, Ungethüm U, Kuban R-J, Lorentz H, Bommert K, Topp M, Krämer D, Müller-Hermelink HK, Einsele H, Greiner A, Bargou RC (2007) STAT3 and MAPK signaling maintain overexpression of heat shock proteins 90alpha and beta in multiple myeloma cells, which critically contribute to tumor-cell survival. Blood 109(2):720–728. doi:10.​1182/​blood-2006-05-024372 PubMedCrossRef
109.
go back to reference Löffler D, Brocke-Heidrich K, Pfeifer G, Stocsits C, Hackermüller J, Kretzschmar AK, Burger R, Gramatzki M, Blumert C, Bauer K, Cvijic H, Ullmann AK, Stadler PF, Horn F (2007) Interleukin-6 dependent survival of multiple myeloma cells involves the Stat3-mediated induction of microRNA-21 through a highly conserved enhancer. Blood 110(4):1330–1333. doi:10.1182/blood-2007-03-081133 PubMedCrossRef Löffler D, Brocke-Heidrich K, Pfeifer G, Stocsits C, Hackermüller J, Kretzschmar AK, Burger R, Gramatzki M, Blumert C, Bauer K, Cvijic H, Ullmann AK, Stadler PF, Horn F (2007) Interleukin-6 dependent survival of multiple myeloma cells involves the Stat3-mediated induction of microRNA-21 through a highly conserved enhancer. Blood 110(4):1330–1333. doi:10.​1182/​blood-2007-03-081133 PubMedCrossRef
110.
go back to reference Alas S, Bonavida B (2003) Inhibition of constitutive STAT3 activity sensitizes resistant non-Hodgkin’s lymphoma and multiple myeloma to chemotherapeutic drug-mediated apoptosis. Clin Cancer Res 9(1):316–326PubMed Alas S, Bonavida B (2003) Inhibition of constitutive STAT3 activity sensitizes resistant non-Hodgkin’s lymphoma and multiple myeloma to chemotherapeutic drug-mediated apoptosis. Clin Cancer Res 9(1):316–326PubMed
111.
go back to reference García JF, Camacho FI, Morente M, Fraga M, Montalbán C, Alvaro T, Bellas C, Castaño A, Díez A, Flores T, Martin C, Martinez MA, Mazorra F, Menárguez J, Mestre MJ, Mollejo M, Sáez AI, Sánchez L, Piris MA, Group SHLS (2003) Hodgkin and Reed-Sternberg cells harbor alterations in the major tumor suppressor pathways and cell-cycle checkpoints: analyses using tissue microarrays. Blood 101(2):681–689. doi:10.1182/blood-2002-04-1128 PubMedCrossRef García JF, Camacho FI, Morente M, Fraga M, Montalbán C, Alvaro T, Bellas C, Castaño A, Díez A, Flores T, Martin C, Martinez MA, Mazorra F, Menárguez J, Mestre MJ, Mollejo M, Sáez AI, Sánchez L, Piris MA, Group SHLS (2003) Hodgkin and Reed-Sternberg cells harbor alterations in the major tumor suppressor pathways and cell-cycle checkpoints: analyses using tissue microarrays. Blood 101(2):681–689. doi:10.​1182/​blood-2002-04-1128 PubMedCrossRef
112.
go back to reference Lamprecht B, Kreher S, Anagnostopoulos I, Jöhrens K, Monteleone G, Jundt F, Stein H, Janz M, Dörken B, Mathas S (2008) Aberrant expression of the Th2 cytokine IL-21 in Hodgkin lymphoma cells regulates STAT3 signaling and attracts Treg cells via regulation of MIP-3alpha. Blood 112(8):3339–3347. doi:10.1182/blood-2008-01-134783 PubMedCrossRef Lamprecht B, Kreher S, Anagnostopoulos I, Jöhrens K, Monteleone G, Jundt F, Stein H, Janz M, Dörken B, Mathas S (2008) Aberrant expression of the Th2 cytokine IL-21 in Hodgkin lymphoma cells regulates STAT3 signaling and attracts Treg cells via regulation of MIP-3alpha. Blood 112(8):3339–3347. doi:10.​1182/​blood-2008-01-134783 PubMedCrossRef
113.
go back to reference Baus D, Pfitzner E (2006) Specific function of STAT3, SOCS1, and SOCS3 in the regulation of proliferation and survival of classical Hodgkin lymphoma cells. Int J Cancer 118(6):1404–1413. doi:10.1002/ijc.21539 PubMedCrossRef Baus D, Pfitzner E (2006) Specific function of STAT3, SOCS1, and SOCS3 in the regulation of proliferation and survival of classical Hodgkin lymphoma cells. Int J Cancer 118(6):1404–1413. doi:10.​1002/​ijc.​21539 PubMedCrossRef
114.
115.
go back to reference Daigle D, Megyola C, El-Guindy A, Gradoville L, Tuck D, Miller G, Bhaduri-McIntosh S (2010) Upregulation of STAT3 marks Burkitt lymphoma cells refractory to Epstein-Barr virus lytic cycle induction by HDAC inhibitors. J Virol 84(2):993–1004. doi:10.1128/JVI.01745-09 PubMedCrossRef Daigle D, Megyola C, El-Guindy A, Gradoville L, Tuck D, Miller G, Bhaduri-McIntosh S (2010) Upregulation of STAT3 marks Burkitt lymphoma cells refractory to Epstein-Barr virus lytic cycle induction by HDAC inhibitors. J Virol 84(2):993–1004. doi:10.​1128/​JVI.​01745-09 PubMedCrossRef
116.
go back to reference Lam LT, Wright G, Davis RE, Lenz G, Farinha P, Dang L, Chan JW, Rosenwald A, Gascoyne RD, Staudt LM (2008) Cooperative signaling through the signal transducer and activator of transcription 3 and nuclear factor-{kappa}B pathways in subtypes of diffuse large B-cell lymphoma. Blood 111(7):3701–3713. doi:10.1182/blood-2007-09-111948 PubMedCrossRef Lam LT, Wright G, Davis RE, Lenz G, Farinha P, Dang L, Chan JW, Rosenwald A, Gascoyne RD, Staudt LM (2008) Cooperative signaling through the signal transducer and activator of transcription 3 and nuclear factor-{kappa}B pathways in subtypes of diffuse large B-cell lymphoma. Blood 111(7):3701–3713. doi:10.​1182/​blood-2007-09-111948 PubMedCrossRef
117.
go back to reference Eriksen KW, Kaltoft K, Mikkelsen G, Nielsen M, Zhang Q, Geisler C, Nissen MH, Röpke C, Wasik MA, Odum N (2001) Constitutive STAT3-activation in Sezary syndrome: tyrphostin AG490 inhibits STAT3-activation, interleukin-2 receptor expression and growth of leukemic Sezary cells. Leukemia 15(5):787–793PubMedCrossRef Eriksen KW, Kaltoft K, Mikkelsen G, Nielsen M, Zhang Q, Geisler C, Nissen MH, Röpke C, Wasik MA, Odum N (2001) Constitutive STAT3-activation in Sezary syndrome: tyrphostin AG490 inhibits STAT3-activation, interleukin-2 receptor expression and growth of leukemic Sezary cells. Leukemia 15(5):787–793PubMedCrossRef
118.
go back to reference Duvic M, Talpur R, Ni X, Zhang C, Hazarika P, Kelly C, Chiao JH, Reilly JF, Ricker JL, Richon VM, Frankel SR (2007) Phase 2 trial of oral vorinostat (suberoylanilide hydroxamic acid, SAHA) for refractory cutaneous T-cell lymphoma (CTCL). Blood 109(1):31–39PubMedCrossRef Duvic M, Talpur R, Ni X, Zhang C, Hazarika P, Kelly C, Chiao JH, Reilly JF, Ricker JL, Richon VM, Frankel SR (2007) Phase 2 trial of oral vorinostat (suberoylanilide hydroxamic acid, SAHA) for refractory cutaneous T-cell lymphoma (CTCL). Blood 109(1):31–39PubMedCrossRef
119.
go back to reference Scheeren FA, Diehl SA, Smit LA, Beaumont T, Naspetti M, Bende RJ, Blom B, Karube K, Ohshima K, Van Noesel CJM, Spits H (2008) IL-21 is expressed in Hodgkin lymphoma and activates STAT5: evidence that activated STAT5 is required for Hodgkin lymphomagenesis. Blood 111(9):4706–4715. doi:10.1182/blood-2007-08-105643 PubMedCrossRef Scheeren FA, Diehl SA, Smit LA, Beaumont T, Naspetti M, Bende RJ, Blom B, Karube K, Ohshima K, Van Noesel CJM, Spits H (2008) IL-21 is expressed in Hodgkin lymphoma and activates STAT5: evidence that activated STAT5 is required for Hodgkin lymphomagenesis. Blood 111(9):4706–4715. doi:10.​1182/​blood-2007-08-105643 PubMedCrossRef
120.
go back to reference Hinz M, Lemke P, Anagnostopoulos I, Hacker C, Krappmann D, Mathas S, Dörken B, Zenke M, Stein H, Scheidereit C (2002) Nuclear factor kappaB-dependent gene expression profiling of Hodgkin’s disease tumor cells, pathogenetic significance, and link to constitutive signal transducer and activator of transcription 5a activity. J Exp Med 196(5):605–617PubMedCrossRef Hinz M, Lemke P, Anagnostopoulos I, Hacker C, Krappmann D, Mathas S, Dörken B, Zenke M, Stein H, Scheidereit C (2002) Nuclear factor kappaB-dependent gene expression profiling of Hodgkin’s disease tumor cells, pathogenetic significance, and link to constitutive signal transducer and activator of transcription 5a activity. J Exp Med 196(5):605–617PubMedCrossRef
121.
go back to reference Buglio D, Georgiakis G, Hanabuchi S, Arima K, Khaskhely N, Liu Y, Younes A (2008) Vorinostat inhibits STAT6-mediated TH2 cytokine and TARC production and induces cell death in Hodgkin lymphoma cell lines. Blood. doi:10.1182/blood-2008-01-133769 PubMed Buglio D, Georgiakis G, Hanabuchi S, Arima K, Khaskhely N, Liu Y, Younes A (2008) Vorinostat inhibits STAT6-mediated TH2 cytokine and TARC production and induces cell death in Hodgkin lymphoma cell lines. Blood. doi:10.​1182/​blood-2008-01-133769 PubMed
124.
go back to reference Skinnider BF, Elia AJ, Gascoyne RD, Patterson B, Trumper L, Kapp U, Mak TW (2002) Signal transducer and activator of transcription 6 is frequently activated in Hodgkin and Reed-Sternberg cells of Hodgkin lymphoma. Blood 99(2):618–626PubMedCrossRef Skinnider BF, Elia AJ, Gascoyne RD, Patterson B, Trumper L, Kapp U, Mak TW (2002) Signal transducer and activator of transcription 6 is frequently activated in Hodgkin and Reed-Sternberg cells of Hodgkin lymphoma. Blood 99(2):618–626PubMedCrossRef
125.
go back to reference Yu H, Jove R (2004) The STATs of cancer–new molecular targets come of age. Nat Rev Cancer 4(2):97–105PubMedCrossRef Yu H, Jove R (2004) The STATs of cancer–new molecular targets come of age. Nat Rev Cancer 4(2):97–105PubMedCrossRef
126.
go back to reference Krämer OH, Knauer SK, Greiner G, Jandt E, Reichardt S, Gührs K-H, Stauber RH, Böhmer FD, Heinzel T (2009) A phosphorylation-acetylation switch regulates STAT1 signaling. Genes Dev 23(2):223–235. doi:10.1101/gad.479209 PubMedCrossRef Krämer OH, Knauer SK, Greiner G, Jandt E, Reichardt S, Gührs K-H, Stauber RH, Böhmer FD, Heinzel T (2009) A phosphorylation-acetylation switch regulates STAT1 signaling. Genes Dev 23(2):223–235. doi:10.​1101/​gad.​479209 PubMedCrossRef
127.
130.
go back to reference Zhang C, Richon V, Ni X, Talpur R, Duvic M (2005) Selective induction of apoptosis by histone deacetylase inhibitor SAHA in cutaneous T-cell lymphoma cells: relevance to mechanism of therapeutic action. J Investig Dermatol 125(5):1045–1052PubMedCrossRef Zhang C, Richon V, Ni X, Talpur R, Duvic M (2005) Selective induction of apoptosis by histone deacetylase inhibitor SAHA in cutaneous T-cell lymphoma cells: relevance to mechanism of therapeutic action. J Investig Dermatol 125(5):1045–1052PubMedCrossRef
133.
go back to reference Hideshima T, Ikeda H, Chauhan D, Okawa Y, Raje N, Podar K, Mitsiades C, Munshi NC, Richardson PG, Carrasco RD, Anderson KC (2009) Bortezomib induces canonical nuclear factor-kappaB activation in multiple myeloma cells. Blood 114(5):1046–1052. doi:10.1182/blood-2009-01-199604 PubMedCrossRef Hideshima T, Ikeda H, Chauhan D, Okawa Y, Raje N, Podar K, Mitsiades C, Munshi NC, Richardson PG, Carrasco RD, Anderson KC (2009) Bortezomib induces canonical nuclear factor-kappaB activation in multiple myeloma cells. Blood 114(5):1046–1052. doi:10.​1182/​blood-2009-01-199604 PubMedCrossRef
134.
go back to reference Li C, Chen S, Yue P, Deng X, Lonial S, Khuri FR, Sun S-Y (2010) The proteasome inhibitor PS-341 (Bortezomib) induces calpain-dependent I{kappa}B{alpha} degradation. J Biol Chem. doi:10.1074/jbc.M109.072694 Li C, Chen S, Yue P, Deng X, Lonial S, Khuri FR, Sun S-Y (2010) The proteasome inhibitor PS-341 (Bortezomib) induces calpain-dependent I{kappa}B{alpha} degradation. J Biol Chem. doi:10.​1074/​jbc.​M109.​072694
136.
go back to reference Maeda T, Towatari M, Kosugi H, Saito H (2000) Up-regulation of costimulatory/adhesion molecules by histone deacetylase inhibitors in acute myeloid leukemia cells. Blood 96(12):3847–3856PubMed Maeda T, Towatari M, Kosugi H, Saito H (2000) Up-regulation of costimulatory/adhesion molecules by histone deacetylase inhibitors in acute myeloid leukemia cells. Blood 96(12):3847–3856PubMed
137.
go back to reference Magner WJ, Kazim AL, Stewart C, Romano MA, Catalano G, Grande C, Keiser N, Santaniello F, Tomasi TB (2000) Activation of MHC class I, II, and CD40 gene expression by histone deacetylase inhibitors. J Immunol 165(12):7017–7024PubMed Magner WJ, Kazim AL, Stewart C, Romano MA, Catalano G, Grande C, Keiser N, Santaniello F, Tomasi TB (2000) Activation of MHC class I, II, and CD40 gene expression by histone deacetylase inhibitors. J Immunol 165(12):7017–7024PubMed
138.
140.
go back to reference Gialitakis M, Kretsovali A, Spilianakis C, Kravariti L, Mages J, Hoffmann R, Hatzopoulos AK, Papamatheakis J (2006) Coordinated changes of histone modifications and HDAC mobilization regulate the induction of MHC class II genes by Trichostatin A. Nucleic Acids Res 34(3):765–772. doi:10.1093/nar/gkj462 PubMedCrossRef Gialitakis M, Kretsovali A, Spilianakis C, Kravariti L, Mages J, Hoffmann R, Hatzopoulos AK, Papamatheakis J (2006) Coordinated changes of histone modifications and HDAC mobilization regulate the induction of MHC class II genes by Trichostatin A. Nucleic Acids Res 34(3):765–772. doi:10.​1093/​nar/​gkj462 PubMedCrossRef
143.
go back to reference Chambost H, Van Baren N, Brasseur F, Godelaine D, Xerri L, Landi SJ, Theate I, Plumas J, Spagnoli GC, Michel G, Coulie PG, Olive D (2000) Expression of gene MAGE-A4 in Reed-Sternberg cells. Blood 95(11):3530–3533PubMed Chambost H, Van Baren N, Brasseur F, Godelaine D, Xerri L, Landi SJ, Theate I, Plumas J, Spagnoli GC, Michel G, Coulie PG, Olive D (2000) Expression of gene MAGE-A4 in Reed-Sternberg cells. Blood 95(11):3530–3533PubMed
144.
go back to reference Cruz C, Leen A, Gerdemann U, Christin A, Tripic T, Shafer J, Younes A, Horton T, Shpall E, Heslop H, Rooney C, Bollard C (2009) Immune-based therapies targeting mage-A4 for relapsed/refractory Hodgkin’s lymphoma after stem cell transplant. ASH Annu Meet Abstr 114(22):4089 EP - Cruz C, Leen A, Gerdemann U, Christin A, Tripic T, Shafer J, Younes A, Horton T, Shpall E, Heslop H, Rooney C, Bollard C (2009) Immune-based therapies targeting mage-A4 for relapsed/refractory Hodgkin’s lymphoma after stem cell transplant. ASH Annu Meet Abstr 114(22):4089 EP -
146.
go back to reference Shichijo S, Yamada A, Sagawa K, Iwamoto O, Sakata M, Nagai K, Itoh K (1996) Induction of MAGE genes in lymphoid cells by the demethylating agent 5-aza-2′-deoxycytidine. Jpn J Cancer Res 87(7):751–756PubMed Shichijo S, Yamada A, Sagawa K, Iwamoto O, Sakata M, Nagai K, Itoh K (1996) Induction of MAGE genes in lymphoid cells by the demethylating agent 5-aza-2′-deoxycytidine. Jpn J Cancer Res 87(7):751–756PubMed
147.
go back to reference Goodyear O, Agathanggelou A, Novitzky-Basso I, Siddique S, McSkeane T, Ryan G, Vyas P, Cavenagh J, Stankovic T, Moss P, Craddock C (2010) Induction of a CD8+ T-cell response to the MAGE cancer testis antigen by combined treatment with azacitidine and sodium valproate in patients with acute myeloid leukemia and myelodysplasia. Blood. doi:10.1182/blood-2009-11-249474 PubMed Goodyear O, Agathanggelou A, Novitzky-Basso I, Siddique S, McSkeane T, Ryan G, Vyas P, Cavenagh J, Stankovic T, Moss P, Craddock C (2010) Induction of a CD8+ T-cell response to the MAGE cancer testis antigen by combined treatment with azacitidine and sodium valproate in patients with acute myeloid leukemia and myelodysplasia. Blood. doi:10.​1182/​blood-2009-11-249474 PubMed
148.
go back to reference Moreno A, Szmania S, Shi J, Barlogie B, Prentice G, van Rhee F (2008) Induction of the cancer-testis antigen MAGE-A3 in myeloma cell lines by 5′azacitidine and MGCD0103. ASCO Meet Abstr 26(15_suppl):14008 EP - Moreno A, Szmania S, Shi J, Barlogie B, Prentice G, van Rhee F (2008) Induction of the cancer-testis antigen MAGE-A3 in myeloma cell lines by 5′azacitidine and MGCD0103. ASCO Meet Abstr 26(15_suppl):14008 EP -
149.
go back to reference Khaskhely NM, Buglio D, Shafer J, Bollard CM, Younes A (2009) The histone deacetylase (HDAC) inhibitor entinostat (SNDX-275) targets Hodgkin lymphoma through a dual mechanism of immune modulation and apoptosis induction. Blood ASH Annu Meet Abstr 114(22):1562- Khaskhely NM, Buglio D, Shafer J, Bollard CM, Younes A (2009) The histone deacetylase (HDAC) inhibitor entinostat (SNDX-275) targets Hodgkin lymphoma through a dual mechanism of immune modulation and apoptosis induction. Blood ASH Annu Meet Abstr 114(22):1562-
150.
go back to reference Moreno-Bost A, Szmania S, Stone K, Shi J, Garg T, Shaughnessy J, Barlogie B, Prentice H, van Rhee F (2008) Recognition of myeloma by MAGE-A3 specific cytotoxic T lymphocytes induced by treatment with azacitidine and MGCD0103. ASH Annu Meet Abstr 112(11):3674 EP - Moreno-Bost A, Szmania S, Stone K, Shi J, Garg T, Shaughnessy J, Barlogie B, Prentice H, van Rhee F (2008) Recognition of myeloma by MAGE-A3 specific cytotoxic T lymphocytes induced by treatment with azacitidine and MGCD0103. ASH Annu Meet Abstr 112(11):3674 EP -
151.
go back to reference Stankovic T, McLarnon A, Agathanggelou A, Goodyear O, Craddock C, Moss P (2008) Epigenetic manipulation of cancer testis antigen (CTA) expression: a strategy for manipulating the graft-versus leukaemia response in patients allografted for haematological malignancies. ASH Annu Meet Abstr 112(11):600 EP - Stankovic T, McLarnon A, Agathanggelou A, Goodyear O, Craddock C, Moss P (2008) Epigenetic manipulation of cancer testis antigen (CTA) expression: a strategy for manipulating the graft-versus leukaemia response in patients allografted for haematological malignancies. ASH Annu Meet Abstr 112(11):600 EP -
154.
go back to reference Armeanu S, Bitzer M, Lauer UM, Venturelli S, Pathil A, Krusch M, Kaiser S, Jobst J, Smirnow I, Wagner A, Steinle A, Salih HR (2005) Natural killer cell-mediated lysis of hepatoma cells via specific induction of NKG2D ligands by the histone deacetylase inhibitor sodium valproate. Cancer Res 65(14):6321–6329. doi:10.1158/0008-5472.CAN-04-4252 PubMedCrossRef Armeanu S, Bitzer M, Lauer UM, Venturelli S, Pathil A, Krusch M, Kaiser S, Jobst J, Smirnow I, Wagner A, Steinle A, Salih HR (2005) Natural killer cell-mediated lysis of hepatoma cells via specific induction of NKG2D ligands by the histone deacetylase inhibitor sodium valproate. Cancer Res 65(14):6321–6329. doi:10.​1158/​0008-5472.​CAN-04-4252 PubMedCrossRef
155.
go back to reference Skov S, Pedersen MT, Andresen L, Straten PT, Woetmann A, Odum N (2005) Cancer cells become susceptible to natural killer cell killing after exposure to histone deacetylase inhibitors due to glycogen synthase kinase-3-dependent expression of MHC class I-related chain A and B. Cancer Res 65(23):11136–11145. doi:10.1158/0008-5472.CAN-05-0599 PubMedCrossRef Skov S, Pedersen MT, Andresen L, Straten PT, Woetmann A, Odum N (2005) Cancer cells become susceptible to natural killer cell killing after exposure to histone deacetylase inhibitors due to glycogen synthase kinase-3-dependent expression of MHC class I-related chain A and B. Cancer Res 65(23):11136–11145. doi:10.​1158/​0008-5472.​CAN-05-0599 PubMedCrossRef
156.
go back to reference Rohner A, Langenkamp U, Siegler U, Kalberer CP, Wodnar-Filipowicz A (2007) Differentiation-promoting drugs up-regulate NKG2D ligand expression and enhance the susceptibility of acute myeloid leukemia cells to natural killer cell-mediated lysis. Leuk Res 31(10):1393–1402. doi:10.1016/j.leukres.2007.02.020 PubMedCrossRef Rohner A, Langenkamp U, Siegler U, Kalberer CP, Wodnar-Filipowicz A (2007) Differentiation-promoting drugs up-regulate NKG2D ligand expression and enhance the susceptibility of acute myeloid leukemia cells to natural killer cell-mediated lysis. Leuk Res 31(10):1393–1402. doi:10.​1016/​j.​leukres.​2007.​02.​020 PubMedCrossRef
157.
go back to reference Diermayr S, Himmelreich H, Durovic B, Mathys-Schneeberger A, Siegler U, Langenkamp U, Hofsteenge J, Gratwohl A, Tichelli A, Paluszewska M, Wiktor-Jedrzejczak W, Kalberer CP, Wodnar-Filipowicz A (2008) NKG2D ligand expression in AML increases in response to HDAC inhibitor valproic acid and contributes to allorecognition by NK-cell lines with single KIR-HLA class I specificities. Blood 111(3):1428–1436. doi:10.1182/blood-2007-07-101311 PubMedCrossRef Diermayr S, Himmelreich H, Durovic B, Mathys-Schneeberger A, Siegler U, Langenkamp U, Hofsteenge J, Gratwohl A, Tichelli A, Paluszewska M, Wiktor-Jedrzejczak W, Kalberer CP, Wodnar-Filipowicz A (2008) NKG2D ligand expression in AML increases in response to HDAC inhibitor valproic acid and contributes to allorecognition by NK-cell lines with single KIR-HLA class I specificities. Blood 111(3):1428–1436. doi:10.​1182/​blood-2007-07-101311 PubMedCrossRef
158.
go back to reference Lu X, Ohata K, Kondo Y, Luis Espinoza J, Qi Z, Nakao S (2009) Hydroxyurea upregulates NKG2D ligand expression in myeloid leukemia cells synergistically with valproic acid and potentially enhances susceptibility of leukemic cells to natural killer cell-mediated cytolysis. Cancer Sci. doi:10.1111/j.1349-7006.2009.01439.x Lu X, Ohata K, Kondo Y, Luis Espinoza J, Qi Z, Nakao S (2009) Hydroxyurea upregulates NKG2D ligand expression in myeloid leukemia cells synergistically with valproic acid and potentially enhances susceptibility of leukemic cells to natural killer cell-mediated cytolysis. Cancer Sci. doi:10.​1111/​j.​1349-7006.​2009.​01439.​x
162.
go back to reference Song W, Tai Y-T, Tian Z, Hideshima T, Chauhan D, Nanjappa P, Exley M, Anderson K, Munshi N (2009) HDAC inhibition by LBH589 affects phenotype and function of human dendritic cells. ASH Annu Meet Abstr 114(22):1646 EP - Song W, Tai Y-T, Tian Z, Hideshima T, Chauhan D, Nanjappa P, Exley M, Anderson K, Munshi N (2009) HDAC inhibition by LBH589 affects phenotype and function of human dendritic cells. ASH Annu Meet Abstr 114(22):1646 EP -
164.
go back to reference Reddy P, Sun Y, Toubai T, Duran-Struuck R, Clouthier SG, Weisiger E, Maeda Y, Tawara I, Krijanovski O, Gatza E, Liu C, Malter C, Mascagni P, Dinarello CA, Ferrara JLM (2008) Histone deacetylase inhibition modulates indoleamine 2, 3-dioxygenase-dependent DC functions and regulates experimental graft-versus-host disease in mice. J Clin Invest 118(7):2562–2573. doi:10.1172/JCI34712 PubMed Reddy P, Sun Y, Toubai T, Duran-Struuck R, Clouthier SG, Weisiger E, Maeda Y, Tawara I, Krijanovski O, Gatza E, Liu C, Malter C, Mascagni P, Dinarello CA, Ferrara JLM (2008) Histone deacetylase inhibition modulates indoleamine 2, 3-dioxygenase-dependent DC functions and regulates experimental graft-versus-host disease in mice. J Clin Invest 118(7):2562–2573. doi:10.​1172/​JCI34712 PubMed
166.
go back to reference Dickinson M, Ritchie D, Deangelo DJ, Spencer A, Ottmann OG, Fischer T, Bhalla KN, Liu A, Parker K, Scott JW, Bishton M, Miles Prince H (2009) Preliminary evidence of disease response to the pan deacetylase inhibitor panobinostat (LBH589) in refractory Hodgkin Lymphoma. Br J Haematol:1–5. doi:10.1111/j.1365-2141.2009.07837.x Dickinson M, Ritchie D, Deangelo DJ, Spencer A, Ottmann OG, Fischer T, Bhalla KN, Liu A, Parker K, Scott JW, Bishton M, Miles Prince H (2009) Preliminary evidence of disease response to the pan deacetylase inhibitor panobinostat (LBH589) in refractory Hodgkin Lymphoma. Br J Haematol:1–5. doi:10.​1111/​j.​1365-2141.​2009.​07837.​x
167.
170.
go back to reference Tao R, de Zoeten EF, Ozkaynak E, Chen C, Wang L, Porrett PM, Li B, Turka LA, Olson EN, Greene MI, Wells AD, Hancock WW (2007) Deacetylase inhibition promotes the generation and function of regulatory T cells. Nat Med 13(11):1299–1307. doi:10.1038/nm1652 PubMedCrossRef Tao R, de Zoeten EF, Ozkaynak E, Chen C, Wang L, Porrett PM, Li B, Turka LA, Olson EN, Greene MI, Wells AD, Hancock WW (2007) Deacetylase inhibition promotes the generation and function of regulatory T cells. Nat Med 13(11):1299–1307. doi:10.​1038/​nm1652 PubMedCrossRef
172.
go back to reference Prabhala RH, Neri P, Bae JE, Tassone P, Shammas MA, Allam CK, Daley JF, Chauhan D, Blanchard E, Thatte HS, Anderson KC, Munshi NC (2006) Dysfunctional T regulatory cells in multiple myeloma. Blood 107(1):301–304. doi:10.1182/blood-2005-08-3101 PubMedCrossRef Prabhala RH, Neri P, Bae JE, Tassone P, Shammas MA, Allam CK, Daley JF, Chauhan D, Blanchard E, Thatte HS, Anderson KC, Munshi NC (2006) Dysfunctional T regulatory cells in multiple myeloma. Blood 107(1):301–304. doi:10.​1182/​blood-2005-08-3101 PubMedCrossRef
174.
go back to reference van Loosdregt J, Vercoulen Y, Guichelaar T, Gent YYJ, Beekman JM, van Beekum O, Brenkman AB, Hijnen D-J, Mutis T, Kalkhoven E, Prakken BJ, Coffer PJ (2010) Regulation of Treg functionality by acetylation-mediated Foxp3 protein stabilization. Blood 115(5):965–974. doi:10.1182/blood-2009-02-207118 PubMedCrossRef van Loosdregt J, Vercoulen Y, Guichelaar T, Gent YYJ, Beekman JM, van Beekum O, Brenkman AB, Hijnen D-J, Mutis T, Kalkhoven E, Prakken BJ, Coffer PJ (2010) Regulation of Treg functionality by acetylation-mediated Foxp3 protein stabilization. Blood 115(5):965–974. doi:10.​1182/​blood-2009-02-207118 PubMedCrossRef
178.
go back to reference Urbich C, Rössig L, Kaluza D, Potente M, Boeckel J-N, Knau A, Diehl F, Geng J-G, Hofmann W-K, Zeiher AM, Dimmeler S (2009) HDAC5 is a repressor of angiogenesis and determines the angiogenic gene expression pattern of endothelial cells. Blood 113(22):5669–5679. doi:10.1182/blood-2009-01-196485 PubMedCrossRef Urbich C, Rössig L, Kaluza D, Potente M, Boeckel J-N, Knau A, Diehl F, Geng J-G, Hofmann W-K, Zeiher AM, Dimmeler S (2009) HDAC5 is a repressor of angiogenesis and determines the angiogenic gene expression pattern of endothelial cells. Blood 113(22):5669–5679. doi:10.​1182/​blood-2009-01-196485 PubMedCrossRef
179.
go back to reference Kim MS, Kwon HJ, Lee YM, Baek JH, Jang JE, Lee SW, Moon EJ, Kim HS, Lee SK, Chung HY, Kim CW, Kim KW (2001) Histone deacetylases induce angiogenesis by negative regulation of tumor suppressor genes. Nat Med 7(4):437–443. doi:10.1038/86507 PubMedCrossRef Kim MS, Kwon HJ, Lee YM, Baek JH, Jang JE, Lee SW, Moon EJ, Kim HS, Lee SK, Chung HY, Kim CW, Kim KW (2001) Histone deacetylases induce angiogenesis by negative regulation of tumor suppressor genes. Nat Med 7(4):437–443. doi:10.​1038/​86507 PubMedCrossRef
180.
go back to reference Deroanne CF, Bonjean K, Servotte S, Devy L, Colige A, Clausse N, Blacher S, Verdin E, Foidart J-M, Nusgens BV, Castronovo V (2002) Histone deacetylases inhibitors as anti-angiogenic agents altering vascular endothelial growth factor signaling. Oncogene 21(3):427–436. doi:10.1038/sj.onc.1205108 PubMedCrossRef Deroanne CF, Bonjean K, Servotte S, Devy L, Colige A, Clausse N, Blacher S, Verdin E, Foidart J-M, Nusgens BV, Castronovo V (2002) Histone deacetylases inhibitors as anti-angiogenic agents altering vascular endothelial growth factor signaling. Oncogene 21(3):427–436. doi:10.​1038/​sj.​onc.​1205108 PubMedCrossRef
181.
go back to reference Qian DZ, Wang X, Kachhap SK, Kato Y, Wei Y, Zhang L, Atadja P, Pili R (2004) 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. doi:10.1158/0008-5472.CAN-04-0540 PubMedCrossRef Qian DZ, Wang X, Kachhap SK, Kato Y, Wei Y, Zhang L, Atadja P, Pili R (2004) 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. doi:10.​1158/​0008-5472.​CAN-04-0540 PubMedCrossRef
183.
go back to reference Qian DZ, Kato Y, Shabbeer S, Wei Y, Verheul HMW, Salumbides B, Sanni T, Atadja P, Pili R (2006) Targeting tumor angiogenesis with histone deacetylase inhibitors: the hydroxamic acid derivative LBH589. Clin Cancer Res 12(2):634–642. doi:10.1158/1078-0432.CCR-05-1132 PubMedCrossRef Qian DZ, Kato Y, Shabbeer S, Wei Y, Verheul HMW, Salumbides B, Sanni T, Atadja P, Pili R (2006) Targeting tumor angiogenesis with histone deacetylase inhibitors: the hydroxamic acid derivative LBH589. Clin Cancer Res 12(2):634–642. doi:10.​1158/​1078-0432.​CCR-05-1132 PubMedCrossRef
184.
go back to reference Kwon HJ, Kim MS, Kim MJ, Nakajima H, Kim K-W (2002) Histone deacetylase inhibitor FK228 inhibits tumor angiogenesis. Int J Cancer 97(3):290–296PubMedCrossRef Kwon HJ, Kim MS, Kim MJ, Nakajima H, Kim K-W (2002) Histone deacetylase inhibitor FK228 inhibits tumor angiogenesis. Int J Cancer 97(3):290–296PubMedCrossRef
186.
go back to reference Aurora AB, Biyashev D, Mirochnik Y, Zaichuk TA, Sánchez-Martinez C, Renault M-A, Losordo D, Volpert OV (2010) NF-kappaB balances vascular regression and angiogenesis via chromatin remodeling and NFAT displacement. Blood 116(3):475–484. doi:10.1182/blood-2009-07-232132 PubMedCrossRef Aurora AB, Biyashev D, Mirochnik Y, Zaichuk TA, Sánchez-Martinez C, Renault M-A, Losordo D, Volpert OV (2010) NF-kappaB balances vascular regression and angiogenesis via chromatin remodeling and NFAT displacement. Blood 116(3):475–484. doi:10.​1182/​blood-2009-07-232132 PubMedCrossRef
187.
go back to reference Wolf J, Siegel D, Matous J, Lonial S, Goldschmidt H, Schmitt S, Vij R, De Malgalhaes-Silverman M, Abonour R, Jalaluddin M, Li M, Hazell K, Bourquelot P, Mateos M-V, Anderson K, Spencer A, Harousseau J-L, Blade J (2008) A phase II study of oral panobinostat (LBH589) in adult patients with advanced refractory multiple myeloma. ASH Annu Meet Abstr 112(11):2774 EP - Wolf J, Siegel D, Matous J, Lonial S, Goldschmidt H, Schmitt S, Vij R, De Malgalhaes-Silverman M, Abonour R, Jalaluddin M, Li M, Hazell K, Bourquelot P, Mateos M-V, Anderson K, Spencer A, Harousseau J-L, Blade J (2008) A phase II study of oral panobinostat (LBH589) in adult patients with advanced refractory multiple myeloma. ASH Annu Meet Abstr 112(11):2774 EP -
188.
go back to reference Verheul HMW, Salumbides B, Van Erp K, Hammers H, Qian DZ, Sanni T, Atadja P, Pili R (2008) Combination strategy targeting the hypoxia inducible factor-1 alpha with mammalian target of rapamycin and histone deacetylase inhibitors. Clin Cancer Res 14(11):3589–3597. doi:10.1158/1078-0432.CCR-07-4306 PubMedCrossRef Verheul HMW, Salumbides B, Van Erp K, Hammers H, Qian DZ, Sanni T, Atadja P, Pili R (2008) Combination strategy targeting the hypoxia inducible factor-1 alpha with mammalian target of rapamycin and histone deacetylase inhibitors. Clin Cancer Res 14(11):3589–3597. doi:10.​1158/​1078-0432.​CCR-07-4306 PubMedCrossRef
190.
go back to reference Trumpp A, Wiestler OD (2008) Mechanisms of disease: cancer stem cells–targeting the evil twin. Nat Clin Prac Oncol 5(6):337–347. doi:10.1038/ncponc1110 Trumpp A, Wiestler OD (2008) Mechanisms of disease: cancer stem cells–targeting the evil twin. Nat Clin Prac Oncol 5(6):337–347. doi:10.​1038/​ncponc1110
194.
go back to reference Matsui W, Wang Q, Barber JP, Brennan S, Smith BD, Borrello I, McNiece I, Lin L, Ambinder RF, Peacock C, Watkins DN, Huff CA, Jones RJ (2008) Clonogenic multiple myeloma progenitors, stem cell properties, and drug resistance. Cancer Res 68(1):190–197. doi:10.1158/0008-5472.CAN-07-3096 PubMedCrossRef Matsui W, Wang Q, Barber JP, Brennan S, Smith BD, Borrello I, McNiece I, Lin L, Ambinder RF, Peacock C, Watkins DN, Huff CA, Jones RJ (2008) Clonogenic multiple myeloma progenitors, stem cell properties, and drug resistance. Cancer Res 68(1):190–197. doi:10.​1158/​0008-5472.​CAN-07-3096 PubMedCrossRef
195.
go back to reference Jones RJ, Matsui WH, Smith BD (2004) Cancer stem cells: are we missing the target? J Natl Cancer Inst 96(8):583–585PubMedCrossRef Jones RJ, Matsui WH, Smith BD (2004) Cancer stem cells: are we missing the target? J Natl Cancer Inst 96(8):583–585PubMedCrossRef
196.
go back to reference Peacock CD, Wang Q, Gesell GS, Corcoran-Schwartz IM, Jones E, Kim J, Devereux WL, Rhodes JT, Huff CA, Beachy PA, Watkins DN, Matsui W (2007) Hedgehog signaling maintains a tumor stem cell compartment in multiple myeloma. Proc Natl Acad Sci USA 104(10):4048–4053. doi:10.1073/pnas.0611682104 PubMedCrossRef Peacock CD, Wang Q, Gesell GS, Corcoran-Schwartz IM, Jones E, Kim J, Devereux WL, Rhodes JT, Huff CA, Beachy PA, Watkins DN, Matsui W (2007) Hedgehog signaling maintains a tumor stem cell compartment in multiple myeloma. Proc Natl Acad Sci USA 104(10):4048–4053. doi:10.​1073/​pnas.​0611682104 PubMedCrossRef
197.
go back to reference Sharma SV, Lee DY, Li B, Quinlan MP, Takahashi F, Maheswaran S, Mcdermott U, Azizian N, Zou L, Fischbach MA, Wong K-K, Brandstetter K, Wittner B, Ramaswamy S, Classon M, Settleman J (2010) A chromatin-mediated reversible drug-tolerant state in cancer cell subpopulations. Cell 141(1):69–80. doi:10.1016/j.cell.2010.02.027 PubMedCrossRef Sharma SV, Lee DY, Li B, Quinlan MP, Takahashi F, Maheswaran S, Mcdermott U, Azizian N, Zou L, Fischbach MA, Wong K-K, Brandstetter K, Wittner B, Ramaswamy S, Classon M, Settleman J (2010) A chromatin-mediated reversible drug-tolerant state in cancer cell subpopulations. Cell 141(1):69–80. doi:10.​1016/​j.​cell.​2010.​02.​027 PubMedCrossRef
199.
go back to reference Grignani F, De Matteis S, Nervi C, Tomassoni L, Gelmetti V, Cioce M, Fanelli M, Ruthardt M, Ferrara FF, Zamir I, Seiser C, Grignani F, Lazar MA, Minucci S, Pelicci PG (1998) Fusion proteins of the retinoic acid receptor-alpha recruit histone deacetylase in promyelocytic leukaemia. Nature 391(6669):815–818. doi:10.1038/35901 PubMedCrossRef Grignani F, De Matteis S, Nervi C, Tomassoni L, Gelmetti V, Cioce M, Fanelli M, Ruthardt M, Ferrara FF, Zamir I, Seiser C, Grignani F, Lazar MA, Minucci S, Pelicci PG (1998) Fusion proteins of the retinoic acid receptor-alpha recruit histone deacetylase in promyelocytic leukaemia. Nature 391(6669):815–818. doi:10.​1038/​35901 PubMedCrossRef
200.
go back to reference Lin RJ, Nagy L, Inoue S, Shao W, Miller WH, Evans RM (1998) Role of the histone deacetylase complex in acute promyelocytic leukaemia. Nature 391(6669):811–814. doi:10.1038/35895 PubMedCrossRef Lin RJ, Nagy L, Inoue S, Shao W, Miller WH, Evans RM (1998) Role of the histone deacetylase complex in acute promyelocytic leukaemia. Nature 391(6669):811–814. doi:10.​1038/​35895 PubMedCrossRef
201.
go back to reference He LZ, Tolentino T, Grayson P, Zhong S, Warrell RP, Rifkind RA, Marks PA, Richon VM, Pandolfi PP (2001) Histone deacetylase inhibitors induce remission in transgenic models of therapy-resistant acute promyelocytic leukemia. J Clin Invest 108(9):1321–1330. doi:10.1172/JCI11537 PubMed He LZ, Tolentino T, Grayson P, Zhong S, Warrell RP, Rifkind RA, Marks PA, Richon VM, Pandolfi PP (2001) Histone deacetylase inhibitors induce remission in transgenic models of therapy-resistant acute promyelocytic leukemia. J Clin Invest 108(9):1321–1330. doi:10.​1172/​JCI11537 PubMed
202.
go back to reference Warrell RP, He LZ, Richon V, Calleja E, Pandolfi PP (1998) Therapeutic targeting of transcription in acute promyelocytic leukemia by use of an inhibitor of histone deacetylase. J Natl Cancer Inst 90(21):1621–1625PubMedCrossRef Warrell RP, He LZ, Richon V, Calleja E, Pandolfi PP (1998) Therapeutic targeting of transcription in acute promyelocytic leukemia by use of an inhibitor of histone deacetylase. J Natl Cancer Inst 90(21):1621–1625PubMedCrossRef
203.
go back to reference Zhou DC, Kim SH, Ding W, Schultz C, Warrell RP Jr, Gallagher RE (2002) Frequent mutations in the ligand-binding domain of PML-RARalpha after multiple relapses of acute promyelocytic leukemia: analysis for functional relationship to response to all-trans retinoic acid and histone deacetylase inhibitors in vitro and in vivo. Blood 99(4):1356–1363PubMedCrossRef Zhou DC, Kim SH, Ding W, Schultz C, Warrell RP Jr, Gallagher RE (2002) Frequent mutations in the ligand-binding domain of PML-RARalpha after multiple relapses of acute promyelocytic leukemia: analysis for functional relationship to response to all-trans retinoic acid and histone deacetylase inhibitors in vitro and in vivo. Blood 99(4):1356–1363PubMedCrossRef
205.
go back to reference Wang J, Hoshino T, Redner RL, Kajigaya S, Liu JM (1998) ETO, fusion partner in t(8;21) acute myeloid leukemia, represses transcription by interaction with the human N-CoR/mSin3/HDAC1 complex. Proc Natl Acad Sci USA 95(18):10860–10865PubMedCrossRef Wang J, Hoshino T, Redner RL, Kajigaya S, Liu JM (1998) ETO, fusion partner in t(8;21) acute myeloid leukemia, represses transcription by interaction with the human N-CoR/mSin3/HDAC1 complex. Proc Natl Acad Sci USA 95(18):10860–10865PubMedCrossRef
206.
go back to reference Fazi F, Zardo G, Gelmetti V, Travaglini L, Ciolfi A, Di Croce L, Rosa A, Bozzoni I, Grignani F, Lo-Coco F, Pelicci PG, Nervi C (2007) Heterochromatic gene repression of the retinoic acid pathway in acute myeloid leukemia. Blood 109(10):4432–4440. doi:10.1182/blood-2006-09-045781 PubMedCrossRef Fazi F, Zardo G, Gelmetti V, Travaglini L, Ciolfi A, Di Croce L, Rosa A, Bozzoni I, Grignani F, Lo-Coco F, Pelicci PG, Nervi C (2007) Heterochromatic gene repression of the retinoic acid pathway in acute myeloid leukemia. Blood 109(10):4432–4440. doi:10.​1182/​blood-2006-09-045781 PubMedCrossRef
207.
go back to reference Tabe Y, Jin L, Contractor R, Gold D, Ruvolo P, Radke S, Xu Y, Tsutusmi-Ishii Y, Miyake K, Miyake N, Kondo S, Ohsaka A, Nagaoka I, Andreeff M, Konopleva M (2007) Novel role of HDAC inhibitors in AML1/ETO AML cells: activation of apoptosis and phagocytosis through induction of annexin A1. Cell Death Differ 14(8):1443–1456. doi:10.1038/sj.cdd.4402139 PubMedCrossRef Tabe Y, Jin L, Contractor R, Gold D, Ruvolo P, Radke S, Xu Y, Tsutusmi-Ishii Y, Miyake K, Miyake N, Kondo S, Ohsaka A, Nagaoka I, Andreeff M, Konopleva M (2007) Novel role of HDAC inhibitors in AML1/ETO AML cells: activation of apoptosis and phagocytosis through induction of annexin A1. Cell Death Differ 14(8):1443–1456. doi:10.​1038/​sj.​cdd.​4402139 PubMedCrossRef
208.
go back to reference Wang J, Saunthararajah Y, Redner RL, Liu JM (1999) Inhibitors of histone deacetylase relieve ETO-mediated repression and induce differentiation of AML1-ETO leukemia cells. Cancer Res 59(12):2766–2769PubMed Wang J, Saunthararajah Y, Redner RL, Liu JM (1999) Inhibitors of histone deacetylase relieve ETO-mediated repression and induce differentiation of AML1-ETO leukemia cells. Cancer Res 59(12):2766–2769PubMed
209.
go back to reference Odenike OM, Alkan S, Sher D, Godwin JE, Huo D, Brandt SJ, Green M, Xie J, Zhang Y, Vesole DH, Stiff P, Wright J, Larson RA, Stock W (2008) Histone deacetylase inhibitor romidepsin has differential activity in core binding factor acute myeloid leukemia. Clin Cancer Res 14(21):7095–7101. doi:10.1158/1078-0432.CCR-08-1007 PubMedCrossRef Odenike OM, Alkan S, Sher D, Godwin JE, Huo D, Brandt SJ, Green M, Xie J, Zhang Y, Vesole DH, Stiff P, Wright J, Larson RA, Stock W (2008) Histone deacetylase inhibitor romidepsin has differential activity in core binding factor acute myeloid leukemia. Clin Cancer Res 14(21):7095–7101. doi:10.​1158/​1078-0432.​CCR-08-1007 PubMedCrossRef
210.
go back to reference Xia ZB, Anderson M, Diaz MO, Zeleznik-Le NJ (2003) MLL repression domain interacts with histone deacetylases, the polycomb group proteins HPC2 and BMI-1, and the corepressor C-terminal-binding protein. Proc Natl Acad Sci USA 100(14):8342–8347. doi:10.1073/pnas.1436338100 PubMedCrossRef Xia ZB, Anderson M, Diaz MO, Zeleznik-Le NJ (2003) MLL repression domain interacts with histone deacetylases, the polycomb group proteins HPC2 and BMI-1, and the corepressor C-terminal-binding protein. Proc Natl Acad Sci USA 100(14):8342–8347. doi:10.​1073/​pnas.​1436338100 PubMedCrossRef
212.
go back to reference Sobulo OM, Borrow J, Tomek R, Reshmi S, Harden A, Schlegelberger B, Housman D, Doggett NA, Rowley JD, Zeleznik-Le NJ (1997) MLL is fused to CBP, a histone acetyltransferase, in therapy-related acute myeloid leukemia with a t(11;16)(q23;p13.3). Proc Natl Acad Sci USA 94(16):8732–8737PubMedCrossRef Sobulo OM, Borrow J, Tomek R, Reshmi S, Harden A, Schlegelberger B, Housman D, Doggett NA, Rowley JD, Zeleznik-Le NJ (1997) MLL is fused to CBP, a histone acetyltransferase, in therapy-related acute myeloid leukemia with a t(11;16)(q23;p13.3). Proc Natl Acad Sci USA 94(16):8732–8737PubMedCrossRef
213.
go back to reference Thirman MJ, Gill HJ, Burnett RC, Mbangkollo D, McCabe NR, Kobayashi H, Ziemin-van der Poel S, Kaneko Y, Morgan R, Sandberg AA et al (1993) Rearrangement of the MLL gene in acute lymphoblastic and acute myeloid leukemias with 11q23 chromosomal translocations. N Engl J Med 329(13):909–914PubMedCrossRef Thirman MJ, Gill HJ, Burnett RC, Mbangkollo D, McCabe NR, Kobayashi H, Ziemin-van der Poel S, Kaneko Y, Morgan R, Sandberg AA et al (1993) Rearrangement of the MLL gene in acute lymphoblastic and acute myeloid leukemias with 11q23 chromosomal translocations. N Engl J Med 329(13):909–914PubMedCrossRef
216.
go back to reference Tonelli R, Sartini R, Fronza R, Freccero F, Franzoni M, Dongiovanni D, Ballarini M, Ferrari S, D’Apolito M, Di Cola G, Capranico G, Khobta A, Campanini R, Paolucci P, Minucci S, Pession A (2006) G1 cell-cycle arrest and apoptosis by histone deacetylase inhibition in MLL-AF9 acute myeloid leukemia cells is p21 dependent and MLL-AF9 independent. Leukemia 20(7):1307–1310. doi:10.1038/sj.leu.2404221 PubMedCrossRef Tonelli R, Sartini R, Fronza R, Freccero F, Franzoni M, Dongiovanni D, Ballarini M, Ferrari S, D’Apolito M, Di Cola G, Capranico G, Khobta A, Campanini R, Paolucci P, Minucci S, Pession A (2006) G1 cell-cycle arrest and apoptosis by histone deacetylase inhibition in MLL-AF9 acute myeloid leukemia cells is p21 dependent and MLL-AF9 independent. Leukemia 20(7):1307–1310. doi:10.​1038/​sj.​leu.​2404221 PubMedCrossRef
217.
go back to reference Burbury KL, Bishton MJ, Johnstone RW, Dickinson M, Szer J, Prince HM (2010) Sustained complete cytogenetic remission in MLL-aberrant leukemia following treatment with a Histone Deacetylase (HDAC) inhibitor (HDACi). Ann Hematol In Press. doi:10.1007/s00277-010-1099-6 Burbury KL, Bishton MJ, Johnstone RW, Dickinson M, Szer J, Prince HM (2010) Sustained complete cytogenetic remission in MLL-aberrant leukemia following treatment with a Histone Deacetylase (HDAC) inhibitor (HDACi). Ann Hematol In Press. doi:10.​1007/​s00277-010-1099-6
Metadata
Title
Histone deacetylase inhibitors: potential targets responsible for their anti-cancer effect
Authors
Michael Dickinson
Ricky W. Johnstone
H. Miles Prince
Publication date
01-12-2010
Publisher
Springer US
Published in
Investigational New Drugs / Issue Special Issue 1/2010
Print ISSN: 0167-6997
Electronic ISSN: 1573-0646
DOI
https://doi.org/10.1007/s10637-010-9596-y

Other articles of this Special Issue 1/2010

Investigational New Drugs 1/2010 Go to the issue
Webinar | 19-02-2024 | 17:30 (CET)

Keynote webinar | Spotlight on antibody–drug conjugates in cancer

Antibody–drug conjugates (ADCs) are novel agents that have shown promise across multiple tumor types. Explore the current landscape of ADCs in breast and lung cancer with our experts, and gain insights into the mechanism of action, key clinical trials data, existing challenges, and future directions.

Dr. Véronique Diéras
Prof. Fabrice Barlesi
Developed by: Springer Medicine