113
Views
30
CrossRef citations to date
0
Altmetric
Review

Epigenetic targets in hematological malignancies: combination therapies with HDACis and demethylating agents

, , , &
Pages 1439-1449 | Published online: 10 Jan 2014

References

  • Jones PA, Baylin SB. The fundamental role of epigenetic events in cancer. Nat. Rev. Genet.3(6), 415–428 (2002).
  • Rice JC, Allis CD. Code of silence. Nature414(6861), 258–261 (2001).
  • Redner RL, Wang J, Liu JM. Chromatin remodeling and leukemia: new therapeutic paradigms. Blood94(2), 417–428 (1999).
  • Jones LK, Saha V. Chromatin modification, leukaemia and implications for therapy. Br. J. Haematol.118(3), 714–727 (2002).
  • Herman JG, Baylin SB. Gene silencing in cancer in association with promoter hypermethylation. N. Engl. J. Med.349(21), 2042–2054 (2003).
  • Jones PA, Baylin SB. The epigenomics of cancer. Cell128(4), 683–692 (2007).
  • Sparmann A, van Lohuizen M. Polycomb silencers control cell fate, development and cancer. Nat. Rev. Cancer6(11), 846–856 (2006).
  • Marks PA, Richon VM, Rifkind RA. Histone deacetylase inhibitors: inducers of differentiation or apoptosis of transformed cells. J. Natl Cancer Inst.92(15), 1210–1216 (2000).
  • Kelly WK, Marks PA. Drug insight: histone deacetylase inhibitors – development of the new targeted anticancer agent suberoylanilide hydroxamic acid. Nat. Clin. Pract. Oncol.2(3), 150–157 (2005).
  • Piekarz RL, Robey R, Sandor V et al. Inhibitor of histone deacetylation, depsipeptide (FR901228), in the treatment of peripheral and cutaneous T-cell lymphoma: a case report. Blood98(9), 2865–2868 (2001).
  • Zhang C, Richon V, Ni X, Talpur R, Duvic M. Selective induction of apoptosis by histone deacetylase inhibitor SAHA in cutaneous T-cell lymphoma cells: relevance to mechanism of therapeutic action. J. Invest. Dermatol.125(5), 1045–1052 (2005).
  • Peart MJ, Tainton KM, Ruefli AA et al. Novel mechanisms of apoptosis induced by histone deacetylase inhibitors. Cancer Res.63(15), 4460–4471 (2003).
  • Burgess AJ, Pavey S, Warrener R et al. Up-regulation of p21(WAF1/CIP1) by histone deacetylase inhibitors reduces their cytotoxicity. Mol. Pharmacol.60(4), 828–837 (2001).
  • Peart MJ, Smyth GK, van Laar RK et al. Identification and functional significance of genes regulated by structurally different histone deacetylase inhibitors. Proc. Natl Acad. Sci. USA102(10), 3697–3702 (2005).
  • Bhalla KN. Epigenetic and chromatin modifiers as targeted therapy of hematologic malignancies. J. Clin. Oncol.23(17), 3971–3993 (2005).
  • Dokmanovic M, Marks PA. Prospects: histone deacetylase inhibitors. J. Cell Biochem.96(2), 293–304 (2005).
  • Santini V, Kantarjian HM, Issa JP. Changes in DNA methylation in neoplasia: pathophysiology and therapeutic implications. Ann. Intern. Med.134, 573–586 (2001).
  • Uchida T, Knoshita T, Nagai H et al. Hypermethylation of p15ink4b gene in myelodysplastic syndromes. Blood90, 1403–1409 (1997).
  • Quesnel B, Guillerm G, Verecque R et al. Methylation of the p15INK4b gene in myelodysplastic syndromes is frequent and acquired during disease progression. Blood91, 2985–2990 (1998).
  • Esteller M, Corn PG, Baylin SB, Herman JG. A gene hypermethylation profile of human cancer. Cancer Res.61(8), 3225–3229 (2001).
  • Jenuwein T, Allis CD. Translating the histone code. Science293(5532), 1074–1080 (2001).
  • Mann BS, Johnson JR, He K et al. Vorinostat for treatment of cutaneous manifestations of advanced primary cutaneous T-cell lymphoma. Clin. Cancer Res.13(8), 2318–2322 (2007).
  • Piekarz RL, Frye R, Turner M et al. Update of the NCI multiinstitutional Phase II trial of romidepsin, FK228, for patients with cutaneous or peripheral T-cell lymphoma. American Society of Clinical Oncology Annual Meeting Abstracts25, 8027 (2007).
  • Lerner A, Demierre MF, Whittaker S et al. Romidepsin (depsipeptide, FK228) induces clinically significant responses in treatment-refractory CTCL: interim report of a Phase II multicenter study. American Society of Hematology Annual Meeting Abstracts (2006) (Abstract 2468).
  • Prince HM, George D, Patnaik A et al. Phase I study of oral LBH589, a novel deacetylase (DAC) inhibitor in advanced solid tumors and non-hodgkin’s lymphoma. American Society of Clinical Oncology Annual Meeting Proceedings Part I25(Suppl. 18), 35002 (2007).
  • Duvic M, Talpur R, Ni X et al. Phase 2 trial of oral vorinostat (suberoylanilide hydroxamic acid, SAHA) for refractory cutaneous T-cell lymphoma (CTCL). Blood109(1), 31–39 (2007).
  • O’Connor OA, Heaney ML, Schwartz L et al. Clinical experience with intravenous and oral formulations of the novel histone deacetylase inhibitor suberoylanilide hydroxamic acid in patients with advanced hematologic malignancies. J. Clin. Oncol.24(1), 166–173 (2006).
  • Younes A, Fanale M, Pro B et al. A Phase II study of a novel oral isotype-selective histone deacetylase (HDAC) inhibitor in patients with relapsed or refractory Hodgkin lymphoma. American Society of Clinical Oncology Annual Meeting Proceedings Part I25(Suppl. 18), 8000 (2007).
  • Garcia-Manero G, Yang H, Sanchez-Gonzalez B et al. Final results of a Phase I study of the histone deacetylase inhibitor vorinostat (suberoyanilide hydroxamic acid, SAHA), in patients with leukemia and myelodysplastic syndrome. American Society of Hematology Annual Meeting Abstracts106, 2801 (2007).
  • Byrd JC, Marcucci G, Parthun MR et al. A Phase 1 and pharmacodynamic study of depsipeptide (FK228) in chronic lymphocytic leukemia and acute myeloid leukemia. Blood105(3), 959–967 (2005).
  • Niesvizky R, Ely S, Diliberto M et al. Multicenter Phase II trial of the histone deacetylase inhibitor depsipeptide (FK228) for the treatment of relapsed or refractory multiple myeloma (MM). American Society of Hematology Annual Meeting Abstracts106(11), 2574 (2005).
  • Gojo I, Jiemjit A, Trepel JB et al. Phase 1 and pharmacologic study of MS-275, a histone deacetylase inhibitor, in adults with refractory and relapsed acute leukemias. Blood109(7), 2781–2790 (2007).
  • Gimsing P, Wu F, Qian X et al. Activity of the histone deacetylase (HDAC) inhibitor PXD101 in preclinical studies and in a Phase I study in patients with advanced haematological tumors. American Society of Hematology Annual Meeting Abstracts106, 3337 (2005).
  • Kaminskas E, Farrell A, Abraham S et al. Approval summary: azacitidine for treatment of myelodysplastic syndrome subtypes. Clin. Cancer Res.11, 3604–3608 (2005).
  • Silverman LR, Holland JF, Weinberg RS et al. Effects of treatment with 5-azacytidine on the in vivo and in vitro hematopoiesis in patients with myelodysplastic syndromes. Leukemia7(Suppl. 1), 21–29 (1993).
  • Silverman LR, Demakos EP, Peterson BL et al. Randomized controlled trial of azacitidine in patients with the myelodysplastic syndrome: a study of the Cancer and Leukemia Group B. J. Clin. Oncol.20(10), 2429–2440 (2002).
  • Kornblith AB, Herndon JE II, Silverman LR et al. Impact of azacytidine on the quality of life of patients with myelodysplastic syndrome treated in a randomized Phase III trial: a Cancer and Leukemia Group B study. J. Clin. Oncol.20(10), 2441–2452 (2002).
  • Lyons RM, Cosgriff T, Modi S et al. Tolerability and hematologic improvement assessed using three alternative dosing schedules of azacitidine in patients with myelodysplastic syndromes. American Society of Clinical Oncology Annual Meeting Proceedings Part I25(Suppl. 18), 7083 (2007).
  • Kantarjian H, Issa JP, Rosenfeld CS et al. Decitabine improves patient outcomes in myelodysplastic syndromes: results of a Phase III randomized study. Cancer106(8), 1794–1803 (2006).
  • Cheson BD, Greenberg PL, Bennett JM et al. Clinical application and proposal for modification of the International Working Group (IWG) response criteria in myelodysplasia. Blood108(2), 419–425 (2006).
  • Kantarjian H, Oki Y, Garcia-Manero G et al. Results of a randomized study of 3 schedules of low-dose decitabine in higher-risk myelodysplastic syndrome and chronic myelomonocytic leukemia. Blood109(1), 52–57 (2007).
  • Claus R, Wijermans P, Rüter B et al. Preferential Cytogenetic response to low dose decitabine in MDS with chromosome 7 abnormalities. European Haematology Association Annual Meeting (2007) (Abstract 0233).
  • Scott SA, Lakshimikuttysamma A, Sheridan DP et al. Zebularine inhibits human acute myeloid leukemia cell growth in vitro in association with p15INK4B demethylation and reexpression. Exp. Hematol.35(2), 263–273 (2007).
  • Galm O, Herman JG, Baylin SB. The fundamental role of epigenetics in hematopoietic malignancies. Blood Rev.20(1), 1–13 (2006).
  • Cameron EE, Bachman KE, Myohanen S, Herman JG, Baylin SB. Synergy of demethylation and histone deacetylase inhibition in the re-expression of genes silenced in cancer. Nat. Genet.21(1), 103–107 (1999).
  • Gore SD, Baylin S, Sugar E et al. Combined DNA methyltransferase and histone deacetylase inhibition in the treatment of myeloid neoplasms. Cancer Res.66(12), 6361–6369 (2006).
  • Maslak P, Chanel S, Camacho LH et al. Pilot study of combination transcriptional modulation therapy with sodium phenylbutyrate and 5-azacytidine in patients with acute myeloid leukemia or myelodysplastic syndrome. Leukemia20(2), 212–217 (2006).
  • Garcia-Manero G, Kantarjian HM, Sanchez-Gonzalez B et al. Phase 1/2 study of the combination of 5-aza-2´-deoxycytidine with valproic acid in patients with leukemia. Blood108(10), 3271–3279 (2006).
  • Raffoux E, de Labarthe A, Gardin C et al. Sequential treatment with 5-azacytidine, valproic acid (VPA), and all-trans retinoic acid (ATRA) in patients with high-risk acute myeloid leukemia (AML). American Society of Hematology Annual Meeting Abstracts106, 2798 (2006).
  • Soriano AO, Yang H, Tong W et al. Significant clinical activity of the combination of 5-azacytidine, valproic acid and all-trans retinoic (ATRA) acid in leukemia: results of a Phase I/II study. American Society of Hematology Annual Meeting Abstracts108, 160 (2006).
  • Gore SD, Jiemjit A, Silverman LB et al. Combined methyltransferase/histone deacetylase inhibition with 5-azacitidine and MS-275 in patients with MDS, CMMoL and AML: clinical response, histone acetylation and DNA damage. American Society of Hematology Annual Meeting Abstracts108, 517 (2006).
  • Blum W, Klisovic RB, Hackanson B et al. Clinical, pharmacokinetic, and pharmacodynamic results of a Phase I study of decitabine alone or in combination with valproic acid in acute myeloid leukemia. J. Clin. Oncol.25(25), 3884–3891 (2007).
  • Hutchins LF, Unger JM, Crowley JJ, Coltman CA Jr, Albain KS. Underrepresentation of patients 65 years of age or older in cancer-treatment trials. N. Engl. J. Med.341(27), 2061–2067 (1999).
  • Kantarjian H, O’Brien S, Cortes J et al. Results of intensive chemotherapy in 998 patients age 65 years or older with acute myeloid leukemia or high-risk myelodysplastic syndrome: predictive prognostic models for outcome. Cancer106(5), 1090–1098 (2006).
  • Frohling S, Schlenk RF, Kayser S et al. Cytogenetics and age are major determinants of outcome in intensively treated acute myeloid leukemia patients older than 60 years: results from AMLSG trial AML HD98-B. Blood108(10), 3280–3288 (2006).
  • Garcia-Manero G, Yang AS, Klimek V et al. Phase I/II study of a novel oral isotype-selective histone deacetylase (HDAC) inhibitor MGCD0103 in combination with azacitidine in patients (pts) with high-risk myelodysplastic syndrome (MDS) or acute myelogenous leukemia (AML). American Society of Clinical Oncology Annual Meeting Proceedings Part I25(Suppl. 18), 7062 (2007).
  • Zhou DC, Kim SH, Ding W et al. Frequent mutations in the ligand-binding domain of PML–RARα 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. Blood99(4), 1356–1363 (2002).
  • Kitamura K, Hoshi S, Koike M et al. Histone deacetylase inhibitor but not arsenic trioxide differentiates acute promyelocytic leukaemia cells with t(11;17) in combination with all-trans retinoic acid. Br. J. Haematol.108(4), 696–702 (2000).
  • Klisovic MI, Maghraby EA, Parthun MR et al. Depsipeptide (FR 901228) promotes histone acetylation, gene transcription, apoptosis and its activity is enhanced by DNA methyltransferase inhibitors in AML1/ETO-positive leukemic cells. Leukemia17(2), 350–358 (2003).
  • Tabe Y, Jin L, Contractor R et al. 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 (2007).
  • Liu S, Klisovic RB, Vukosavljevic T et al. Targeting AML1/ETO-histone deacetylase repressor complex: a novel mechanism for valproic acid-mediated gene expression and cellular differentiation in AML1/ETO-positive acute myeloid leukemia cells. J. Pharmacol. Exp. Ther.321(3), 953–960 (2007).
  • Odenike OM, Alkan S, Sher D et al. The histone deacetylase inhibitor depsipeptide has differential activity in specific cytogenetic subsets of acute myeloid leukemia (AML). Blood104(11), A79 (2004).
  • Kuendgen A, Schmid M, Schlenk R et al. The histone deacetylase (HDAC) inhibitor valproic acid as monotherapy or in combination with all-trans retinoic acid in patients with acute myeloid leukemia. Cancer106(1), 112–119 (2006).
  • Bug G, Ritter M, Wassmann B et al. Clinical trial of valproic acid and all-trans retinoic acid in patients with poor-risk acute myeloid leukemia. Cancer104(12), 2717–2725 (2005).
  • Sanchez-Gonzalez B, Yang H, Bueso-Ramos C et al. Antileukemia activity of the combination of an anthracycline with a histone deacetylase inhibitor. Blood108(4), 1174–1182 (2006).
  • Kano Y, Akutsu M, Tsunoda S et al. Cytotoxic effects of histone deacetylase inhibitor FK228 (depsipeptide, formally named FR901228) in combination with conventional anti-leukemia/lymphoma agents against human leukemia/lymphoma cell lines. Invest. New Drugs25(1), 31–40 (2007).
  • Munster P, Marchion D, Bicaku E et al. Phase I trial of histone deacetylase inhibition by valproic acid followed by the topoisomerase II inhibitor epirubicin in advanced solid tumors: a clinical and translational study. J. Clin. Oncol.25(15), 1979–1985 (2007).
  • Pei XY, Dai Y, Grant S. Synergistic induction of oxidative injury and apoptosis in human multiple myeloma cells by the proteasome inhibitor bortezomib and histone deacetylase inhibitors. Clin. Cancer Res.10(11), 3839–3852 (2004).
  • Catley L, Weisberg E, Kiziltepe T et al. Aggresome induction by proteasome inhibitor bortezomib and α-tubulin hyperacetylation by tubulin deacetylase (TDAC) inhibitor LBH589 are synergistic in myeloma cells. Blood108(10), 3441–3449 (2006).
  • Kantarjian H, Pasquini R, Hamerschlak N et al. Dasatinib or high-dose imatinib for chronic-phase chronic myeloid leukemia after failure of first-line imatinib: a randomized Phase 2 trial. Blood109(12), 5143–5150 (2007).
  • Kawano T, Horiguchi-Yamada J, Iwase S et al. Depsipeptide enhances imatinib mesylate-induced apoptosis of Bcr–Abl-positive cells and ectopic expression of cyclin D1, c-Myc or active MEK abrogates this effect. Anticancer Res.24(5A), 2705–2712 (2004).
  • Yu C, Rahmani M, Almenara J et al. Histone deacetylase inhibitors promote STI571-mediated apoptosis in STI571-sensitive and -resistant Bcr/Abl+ human myeloid leukemia cells. Cancer Res.63(9), 2118–2126 (2003).
  • Nimmanapalli R, Fuino L, Stobaugh C, Richon V, Bhalla K. Cotreatment with the histone deacetylase inhibitor suberoylanilide hydroxamic acid (SAHA) enhances imatinib-induced apoptosis of Bcr–Abl-positive human acute leukemia cells. Blood101(8), 3236–3239 (2003).
  • Fiskus W, Pranpat M, Bali P et al. Combined effects of novel tyrosine kinase inhibitor AMN107 and histone deacetylase inhibitor LBH589 against Bcr–Abl-expressing human leukemia cells. Blood108(2), 645–652 (2006).
  • Yu C, Rahmani M, Conrad D et al. The proteasome inhibitor bortezomib interacts synergistically with histone deacetylase inhibitors to induce apoptosis in Bcr/Abl+ cells sensitive and resistant to STI571. Blood102(10), 3765–3774 (2003).
  • Konstantinopoulos PA, Papavassiliou AG. 17-AAG: mechanisms of antitumour activity. Expert Opin. Investig. Drugs14(12), 1471–1474 (2005).
  • Budillon A, Bruzzese F, Di Gennaro E, Caraglia M. Multiple-target drugs: inhibitors of heat shock protein 90 and of histone deacetylase. Curr. Drug Targets6(3), 337–351 (2005).
  • Rahmani M, Yu C, Dai Y et al. Coadministration of the heat shock protein 90 antagonist 17-allylamino-17-demethoxygeldanamycin with suberoylanilide hydroxamic acid or sodium butyrate synergistically induces apoptosis in human leukemia cells. Cancer Res.63(23), 8420–8427 (2003).
  • George P, Bali P, Annavarapu S et al. Combination of the histone deacetylase inhibitor LBH589 and the hsp90 inhibitor 17-AAG is highly active against human CML-BC cells and AML cells with activating mutation of FLT-3. Blood105(4), 1768–1776 (2005).
  • Huang HC, Liu YC, Liu SH et al. Geldanamycin inhibits trichostatin A-induced cell death and histone H4 hyperacetylation in COS-7 cells. Life Sci.70(15), 1763–1775 (2002).
  • Byrd JC, Kitada S, Flinn IW et al. The mechanism of tumor cell clearance by rituximab in vivo in patients with B-cell chronic lymphocytic leukemia: evidence of caspase activation and apoptosis induction. Blood99(3), 1038–1043 (2002).
  • Bolden JE, Peart MJ, Johnstone RW. Anticancer activities of histone deacetylase inhibitors. Nat. Rev. Drug Discov.5(9), 769–784 (2006).
  • Zhao W, Wang L, Liu Y et al. Histone deacetylase inhibitor promotes rituximab-induced apoptosis in non-hodgkin’s B-lymphoma cells by NF-κB-Mediated Bcl-2/Bcl-XL downregulation and c-Myc degradation. American Society of Hematology Annual Meeting Abstracts108, 2526 (2006).
  • Dasmahapatra G, Almenara JA, Grant S. Flavopiridol and histone deacetylase inhibitors promote mitochondrial injury and cell death in human leukemia cells that overexpress Bcl-2. Mol. Pharmacol.69(1), 288–298 (2006).
  • Piekarz RL, Robey RW, Zhan Z et al. T-cell lymphoma as a model for the use of histone deacetylase inhibitors in cancer therapy: impact of depsipeptide on molecular markers, therapeutic targets, and mechanisms of resistance. Blood103(12), 4636–4643 (2004).
  • Inoue S, MacFarlane M, Harper N et al. Histone deacetylase inhibitors potentiate TNF-related apoptosis-inducing ligand (TRAIL)-induced apoptosis in lymphoid malignancies. Cell Death Differ.11(Suppl. 2), S193–S206 (2004).
  • Rosato RR, Almenara JA, Dai Y et al. Simultaneous activation of the intrinsic and extrinsic pathways by histone deacetylase (HDAC) inhibitors and tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) synergistically induces mitochondrial damage and apoptosis in human leukemia cells. Mol. Cancer Ther.2(12), 1273–1284 (2003).
  • Musto P, Falcone A, Sanpaolo G et al. Thalidomide abolishes transfusion-dependence in selected patients with myelodysplastic syndromes. Haematologica87(8), 884–886 (2002).
  • Westervelt P, Amirifeli S, Mehdi M et al. Low dose Vidaza and thalidomide is an effective combination for patients with myelodysplastic syndromes (MDS) and acute myeloid leukemia (AML). American Society of Clinical Oncology Annual Meeting Proceedings Part I24(Suppl. 18), 6570 (2006).
  • Kenealy MK, Seymour JF. Treatment of myelodysplastic syndromes with azacitidine and thalidomide. Leukemia Res.31(Suppl. 1), S7–S8 (2007).
  • Garcia-Manero G, Gore SD. Future directions for the use of hypomethylating agents. Semin. Hematol.42(3 Suppl. 2), S50–S59 (2005).
  • Balaian L, Ball ED. 5-azacytidine potentiates the anti-proliferative activity of anti-CD33 monoclonal antibodies (mAb) in acute myeloid leukemia in part through induction of Syk and SHP-1 expression. American Society of Hematology Annual Meeting Abstracts106, 2459 (2005).
  • Boula A, Voulgarelis M, Giannouli S et al. Effect of cA2 anti-tumor necrosis factor-α antibody therapy on hematopoiesis of patients with myelodysplastic syndromes. Clin. Cancer Res.12(10), 3099–3108 (2006).
  • Kantarjian HM, O’Brien S, Cortes J et al. Results of decitabine (5-aza-2´deoxycytidine) therapy in 130 patients with chronic myelogenous leukemia. Cancer98(3), 522–528 (2003).
  • Oki Y, Kantarjian HM, Gharibyan V et al. Phase II study of low-dose decitabine in combination with imatinib mesylate in patients with accelerated or myeloid blastic phase of chronic myelogenous leukemia. Cancer109(5), 899–906 (2007).
  • Huang Y, Tan M, Gosink M, Wang KK, Sun Y. Histone deacetylase 5 is not a p53 target gene, but its overexpression inhibits tumor cell growth and induces apoptosis. Cancer Res.62(10), 2913–2922 (2002).
  • Kawaguchi Y, Kovacs JJ, McLaurin A et al. The deacetylase HDAC6 regulates aggresome formation and cell viability in response to misfolded protein stress. Cell115(6), 727–738 (2003).
  • Qian DZ, Kachhap SK, Collis SJ et al. Class II Histone deacetylases are associated with VHL-independent regulation of hypoxia-inducible factor 1α. Cancer Res.66(17), 8814–8821 (2006).
  • Bali P, Pranpat M, Bradner J et al. Inhibition of histone deacetylase 6 acetylates and disrupts the chaperone function of heat shock protein 90: a novel basis for antileukemia activity of histone deacetylase inhibitors. J. Biol. Chem.280(29), 26729–26734 (2005).
  • Rasheed WK, Johnstone RW, Prince HM. Histone deacetylase inhibitors in cancer therapy. Expert Opin. Investig. Drugs.16(5), 659–678 (2007).

Reprints and Corporate Permissions

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

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

Academic Permissions

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

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

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