메뉴 건너뛰기




Volumn 88, Issue 9, 2014, Pages 1651-1668

Epigenetic drugs against cancer: An evolving landscape

Author keywords

Cancer; Chromatin modulation; Drug discovery; Epigenetics

Indexed keywords

AGK 2; ANTINEOPLASTIC AGENT; BUTYRIC ACID DERIVATIVE; CAMBINOL; DNA METHYLTRANSFERASE INHIBITOR; ENTINOSTAT; EPIGENETIC DRUG; HISTONE; HISTONE ACETYLTRANSFERASE INHIBITOR; HISTONE DEACETYLASE INHIBITOR; HISTONE METHYLTRANSFERASE INHIBITOR; HISTONE READER; INDOLE; JUMONJI C CONTAINING DEMETHYLASE INHIBITOR; LYSINE SPECIFIC DEMETHYLASE INHIBITOR; MOCETINOSTAT; NICOTINAMIDE ADENINE DINUCLEOTIDE; NICOTINAMIDE DERIVATIVE; PANOBINOSTAT; RESVERATROL; ROMIDEPSIN; SIRTUIN; SPLITOMICIN; SRT 1460; SRT 1720; SRT 2183; SRT1460; SRT1720; SRT2183; SURAMIN; TRICHOSTATIN A; UNCLASSIFIED DRUG; VALPROIC ACID; VORINOSTAT; NEW DRUG; TUMOR PROTEIN;

EID: 84906231124     PISSN: 03405761     EISSN: 14320738     Source Type: Journal    
DOI: 10.1007/s00204-014-1315-6     Document Type: Review
Times cited : (51)

References (193)
  • 1
    • 36049028058 scopus 로고    scopus 로고
    • New nomenclature for chro-matin- modifying enzymes
    • doi:10.1016/j. cell.2007.10.039
    • Allis CD, Berger SL, Cote J et al (2007) New nomenclature for chro-matin- modifying enzymes. Cell 131(4):633-636. doi:10.1016/j. cell.2007.10.039
    • (2007) Cell , vol.131 , Issue.4 , pp. 633-636
    • Allis, C.D.1    Berger, S.L.2    Cote, J.3
  • 2
    • 84861048806 scopus 로고    scopus 로고
    • MG98, a second-generation DNMT1 inhibitor, in the treatment of advanced renal cell carcinoma
    • doi:10.3109/07357907.2012.675381
    • Amato RJ, Stephenson J, Hotte S et al (2012) MG98, a second-generation DNMT1 inhibitor, in the treatment of advanced renal cell carcinoma. Cancer Invest 30(5):415-421. doi:10.3109/07357907.2012.675381
    • (2012) Cancer Invest , vol.30 , Issue.5 , pp. 415-421
    • Amato, R.J.1    Stephenson, J.2    Hotte, S.3
  • 3
    • 84883017134 scopus 로고    scopus 로고
    • Phase I trial of a new schedule of romidepsin in patients with advanced cancers
    • doi:10.1158/1078-0432.CR-13-0095
    • Amiri-Kordestani L, Luchenko V, Peer CJ et al (2013) Phase I trial of a new schedule of romidepsin in patients with advanced cancers. Clin Cancer Res 19(16):4499-4507. doi:10.1158/1078-0432.CR-13-0095
    • (2013) Clin Cancer Res , vol.19 , Issue.16 , pp. 4499-4507
    • Amiri-Kordestani, L.1    Luchenko, V.2    Peer, C.J.3
  • 4
    • 84896925980 scopus 로고    scopus 로고
    • Resveratrol-sulfates provide an intracellular reservoir for generation of parent resveratrol, which induces autophagy in cancer cells
    • doi:10.4161/auto.27593
    • Andreadi C, Britton RG, Patel KR, Brown K (2014) Resveratrol-sulfates provide an intracellular reservoir for generation of parent resveratrol, which induces autophagy in cancer cells. Autophagy 10(3):524-525. doi:10.4161/auto. 27593
    • (2014) Autophagy , vol.10 , Issue.3 , pp. 524-525
    • Andreadi, C.1    Britton, R.G.2    Patel, K.R.3    Brown, K.4
  • 6
    • 79959866409 scopus 로고    scopus 로고
    • Nicotinamide blocks proliferation and induces apoptosis of chronic lymphocytic leukemia cells through activation of the p53/miR-34a/SIRT1 tumor suppressor network
    • doi:10.1158/0008-5472.CAN-10-4452
    • Audrito V, Vaisitti T, Rossi D et al (2011) Nicotinamide blocks proliferation and induces apoptosis of chronic lymphocytic leukemia cells through activation of the p53/miR-34a/SIRT1 tumor suppressor network. Cancer Res 71(13):4473-4483. doi:10.1158/0008-5472.CAN-10-4452
    • (2011) Cancer Res , vol.71 , Issue.13 , pp. 4473-4483
    • Audrito, V.1    Vaisitti, T.2    Rossi, D.3
  • 8
    • 10944243759 scopus 로고    scopus 로고
    • Curcumin, a novel p300/CREB-binding protein-specific inhibitor of acetyltransferase, represses the acetylation of histone/nonhistone proteins and histone acetyltransferase-dependent chromatin transcription
    • DOI 10.1074/jbc.M409024200
    • Balasubramanyam K, Varier RA, Altaf M et al (2004) Curcumin, a novel p300/CREB-binding protein-specific inhibitor of acetyltransferase, represses the acetylation of histone/nonhistone proteins and histone acetyltransferase- dependent chromatin transcription. J Biol Chem 279(49):51163-51171. doi:10.1074/jbc.M409024200 (Pubitemid 40017859)
    • (2004) Journal of Biological Chemistry , vol.279 , Issue.49 , pp. 51163-51171
    • Balasubramanyam, K.1    Varier, R.A.2    Altaf, M.3    Swaminathan, V.4    Siddappa, N.B.5    Ranga, U.6    Kundu, T.K.7
  • 9
    • 79952534189 scopus 로고    scopus 로고
    • Regulation of chromatin by histone modifications
    • doi:10.1038/cr.2011.22
    • Bannister AJ, Kouzarides T (2011) Regulation of chromatin by histone modifications. Cell Res 21(3):381-395. doi:10.1038/cr.2011.22
    • (2011) Cell Res , vol.21 , Issue.3 , pp. 381-395
    • Bannister, A.J.1    Kouzarides, T.2
  • 11
    • 84863453769 scopus 로고    scopus 로고
    • SIRT7 links H3K18 deacetylation to maintenance of oncogenic transformation
    • doi:10.1038/nature11043
    • Barber MF, Michishita-Kioi E, Xi Y et al (2012) SIRT7 links H3K18 deacetylation to maintenance of oncogenic transformation. Nature 487(7405):114-118. doi:10.1038/nature11043
    • (2012) Nature , vol.487 , Issue.7405 , pp. 114-118
    • Barber, M.F.1    Michishita-Kioi, E.2    Xi, Y.3
  • 12
    • 84871671268 scopus 로고    scopus 로고
    • Highly ligand efficient and selective N-2- (Thioethyl)picolinamide histone deacetylase inhibitors inspired by the natural product psammaplin A
    • doi:10.1002/cmdc.201200450
    • Baud MG, Haus P, Leiser T, Meyer-Almes FJ, Fuchter MJ (2013) Highly ligand efficient and selective N-2- (Thioethyl)picolinamide histone deacetylase inhibitors inspired by the natural product psammaplin A. ChemMedChem 8(1):149-156. doi:10.1002/cmdc.201200450
    • (2013) ChemMedChem , vol.8 , Issue.1 , pp. 149-156
    • Baud, M.G.1    Haus, P.2    Leiser, T.3    Meyer-Almes, F.J.4    Fuchter, M.J.5
  • 15
    • 78751512281 scopus 로고    scopus 로고
    • Combination therapy with vidaza and entinostat suppresses tumor growth and reprograms the epigenome in an orthotopic lung cancer model
    • doi:10.1158/0008-5472.CAN-10-3184
    • Belinsky SA, Grimes MJ, Picchi MA et al (2011) Combination therapy with vidaza and entinostat suppresses tumor growth and reprograms the epigenome in an orthotopic lung cancer model. Cancer Res 71(2):454-462. doi:10.1158/0008-5472. CAN-10-3184
    • (2011) Cancer Res , vol.71 , Issue.2 , pp. 454-462
    • Belinsky, S.A.1    Grimes, M.J.2    Picchi, M.A.3
  • 16
    • 84935029387 scopus 로고    scopus 로고
    • Targeting histone deacetylases in diseases: Where are we?
    • doi:10.1089/ars.2013.5776
    • Benedetti R, Conte M, Altucci L (2014) Targeting histone deacetylases in diseases: Where are we? Antioxid Redox Signal. doi:10.1089/ars.2013.5776
    • (2014) Antioxid Redox Signal
    • Benedetti, R.1    Conte, M.2    Altucci, L.3
  • 17
    • 4544318283 scopus 로고    scopus 로고
    • Design, synthesis, and biological evaluation of a small-molecule inhibitor of the histone acetyltransferase Gcn5
    • DOI 10.1002/anie.200453879
    • Biel M, Kretsovali A, Karatzali E, Papamatheakis J, Giannis A (2004) Design, synthesis, and biological evaluation of a small-molecule inhibitor of the histone acetyltransferase Gcn5. Angew Chem 43(30):3974-3976. doi:10.1002/anie.200453879 (Pubitemid 39257485)
    • (2004) Angewandte Chemie - International Edition , vol.43 , Issue.30 , pp. 3974-3976
    • Biel, M.1    Kretsovali, A.2    Karatzali, E.3    Papamatheakis, J.4    Giannis, A.5
  • 18
    • 77954848597 scopus 로고    scopus 로고
    • Curcumin, resveratrol and flavonoids as anti-inflammatory, cyto- and DNA-protective dietary compounds
    • doi:10.1016/j.tox.2009.11.008
    • Bisht K, Wagner KH, Bulmer AC (2010) Curcumin, resveratrol and flavonoids as anti-inflammatory, cyto- and DNA-protective dietary compounds. Toxicology 278(1):88-100. doi:10.1016/j.tox.2009.11.008
    • (2010) Toxicology , vol.278 , Issue.1 , pp. 88-100
    • Bisht, K.1    Wagner, K.H.2    Bulmer, A.C.3
  • 19
    • 84887057701 scopus 로고    scopus 로고
    • The promise and failures of epigenetic therapies for cancer treatment
    • doi:10.1016/j.ctrv.2013.05.009
    • Bojang P Jr, Ramos KS (2014) The promise and failures of epigenetic therapies for cancer treatment. Cancer Treat Rev 40(1):153-169. doi:10.1016/j.ctrv.2013.05.009
    • (2014) Cancer Treat Rev , vol.40 , Issue.1 , pp. 153-169
    • Bojang Jr., P.1    Ramos, K.S.2
  • 20
    • 84905238187 scopus 로고    scopus 로고
    • Histone deacetylase inhibitor (HDACI) mechanisms of action: Emerging insights
    • doi:10.1016/j.pharmthera.2014.04.004
    • Bose P, Dai Y, Grant S (2014) Histone deacetylase inhibitor (HDACI) mechanisms of action: emerging insights. Pharmacol Ther. doi:10.1016/j. pharmthera.2014.04.004
    • (2014) Pharmacol Ther
    • Bose, P.1    Dai, Y.2    Grant, S.3
  • 21
    • 0020541275 scopus 로고
    • Incorporation of 5-aza-2'-deoxycytidine-5'-triphosphate into DNA. Interactions with mammalian DNA polymerase alpha and DNA methylase
    • Bouchard J, Momparler RL (1983) Incorporation of 5-Aza-2′- deoxycytidine-5′-triphosphate into DNA. Interactions with mammalian DNA polymerase alpha and DNA methylase. Mol Pharmacol 24(1):109-114 (Pubitemid 13075113)
    • (1983) Molecular Pharmacology , vol.24 , Issue.1 , pp. 109-114
    • Bouchard, J.1    Momparler, R.L.2
  • 22
    • 77954204335 scopus 로고    scopus 로고
    • Virtual ligand screening of the p300/CBP histone acetyltransferase: Identification of a selective small molecule inhibitor
    • doi:10.1016/j.chembiol.2010.03.006
    • Bowers EM, Yan G, Mukherjee C et al (2010) Virtual ligand screening of the p300/CBP histone acetyltransferase: identification of a selective small molecule inhibitor. Chem Biol 17(5):471-482. doi:10.1016/j.chembiol.2010.03.006
    • (2010) Chem Biol , vol.17 , Issue.5 , pp. 471-482
    • Bowers, E.M.1    Yan, G.2    Mukherjee, C.3
  • 23
    • 22244435605 scopus 로고    scopus 로고
    • Epigenetic reactivation of tumor suppressor genes by a novel small-molecule inhibitor of human DNA methyltransferases
    • DOI 10.1158/0008-5472.CAN-04-2957
    • Brueckner B, Garcia Boy R, Siedlecki P et al (2005) Epigenetic reactivation of tumor suppressor genes by a novel small-molecule inhibitor of human DNA methyltransferases. Cancer Res 65(14):6305-6311. doi:10.1158/0008- 5472.CAN-04-2957 (Pubitemid 40994416)
    • (2005) Cancer Research , vol.65 , Issue.14 , pp. 6305-6311
    • Brueckner, B.1    Boy, R.G.2    Siedlecki, P.3    Musch, T.4    Kliem, H.C.5    Zielenkiewicz, P.6    Suhai, S.7    Wiessler, M.8    Lyko, F.9
  • 24
    • 84876717815 scopus 로고    scopus 로고
    • Rejuvenating sirtuins: The rise of a new family of cancer drug targets
    • Bruzzone S, Parenti MD, Grozio A et al (2013) Rejuvenating sirtuins: the rise of a new family of cancer drug targets. Curr Pharm Des 19(4):614-623
    • (2013) Curr Pharm des , vol.19 , Issue.4 , pp. 614-623
    • Bruzzone, S.1    Parenti, M.D.2    Grozio, A.3
  • 25
    • 0032948005 scopus 로고    scopus 로고
    • Synergy of demethylation and histone deacetylase inhibition in the re- expression of genes silenced in cancer
    • DOI 10.1038/5047
    • Cameron E, Bachman KE, Myohanen S, Herman JG, Baylin SB (1999) Synergy of demethylation and histone deacetylase inhibition in the re-expression of genes silenced in cancer. Nat Genet 21(1):103-107. doi:10.1038/5047 (Pubitemid 29036293)
    • (1999) Nature Genetics , vol.21 , Issue.1 , pp. 103-107
    • Cameron, E.E.1    Bachman, K.E.2    Myohanen, S.3    Herman, J.G.4    Baylin, S.B.5
  • 26
    • 79955932718 scopus 로고    scopus 로고
    • Potential beneficial effects of butyrate in intestinal and extraintestinal diseases
    • doi:10.3748/wjg.v17.i12.1519
    • Canani RB, Costanzo MD, Leone L, Pedata M, Meli R, Calignano A (2011) Potential beneficial effects of butyrate in intestinal and extraintestinal diseases. World J Gastroenterol 17(12):1519-1528. doi:10.3748/wjg.v17.i12.1519
    • (2011) World J Gastroenterol , vol.17 , Issue.12 , pp. 1519-1528
    • Canani, R.B.1    Costanzo, M.D.2    Leone, L.3    Pedata, M.4    Meli, R.5    Calignano, A.6
  • 28
    • 79960821550 scopus 로고    scopus 로고
    • Synergistic interactions between HDAC and sirtuin inhibitors in human leukemia cells
    • doi:10.1371/journal.pone.0022739
    • Cea M, Soncini D, Fruscione F et al (2011) Synergistic interactions between HDAC and sirtuin inhibitors in human leukemia cells. PLoS ONE 6(7):e22739. doi:10.1371/journal.pone.0022739
    • (2011) PLoS ONE , vol.6 , Issue.7
    • Cea, M.1    Soncini, D.2    Fruscione, F.3
  • 29
    • 77953292895 scopus 로고    scopus 로고
    • Sirtuins inhibitors: The approach to affinity and selectivity
    • doi:10.1016/j.bbapap.2009.11.010
    • Cen Y (2010) Sirtuins inhibitors: the approach to affinity and selectivity. Biochim Biophys Acta 1804(8):1635-1644. doi:10.1016/j.bbapap.2009. 11.010
    • (2010) Biochim Biophys Acta , vol.1804 , Issue.8 , pp. 1635-1644
    • Cen, Y.1
  • 30
    • 84897024898 scopus 로고    scopus 로고
    • Potent antimyeloma activity of the novel bromodomain inhibitors I-BET151 and I-BET762
    • doi:10.1182/blood-2013-01-478420
    • Chaidos A, Caputo V, Gouvedenou K et al (2014) Potent antimyeloma activity of the novel bromodomain inhibitors I-BET151 and I-BET762. Blood 123(5):697-705. doi:10.1182/blood-2013-01-478420
    • (2014) Blood , vol.123 , Issue.5 , pp. 697-705
    • Chaidos, A.1    Caputo, V.2    Gouvedenou, K.3
  • 31
    • 84555207349 scopus 로고    scopus 로고
    • Dnmt3a is essential for hematopoietic stem cell differentiation
    • doi:10.1038/ng.1009
    • Challen GA, Sun D, Jeong M et al (2012) Dnmt3a is essential for hematopoietic stem cell differentiation. Nat Genet 44(1):23-31. doi:10.1038/ng.1009
    • (2012) Nat Genet , vol.44 , Issue.1 , pp. 23-31
    • Challen, G.A.1    Sun, D.2    Jeong, M.3
  • 32
    • 62049083789 scopus 로고    scopus 로고
    • Structural basis for G9alike protein lysine methyltransferase inhibition by BIX-01294
    • doi:10.1038/nsmb.1560
    • Chang Y, Zhang X, Horton JR et al (2009) Structural basis for G9alike protein lysine methyltransferase inhibition by BIX-01294. Nat Struct Mol Biol 16(3):312-317. doi:10.1038/nsmb.1560
    • (2009) Nat Struct Mol Biol , vol.16 , Issue.3 , pp. 312-317
    • Chang, Y.1    Zhang, X.2    Horton, J.R.3
  • 33
    • 84863130872 scopus 로고    scopus 로고
    • DNA methyltransferase inhibitor, zebularine, delays tumor growth and induces apoptosis in a genetically engineered mouse model of breast cancer
    • doi:10.1158/1535-7163.MCT-11-0458
    • Chen M, Shabashvili D, Nawab A et al (2012) DNA methyltransferase inhibitor, zebularine, delays tumor growth and induces apoptosis in a genetically engineered mouse model of breast cancer. Mol Cancer Ther 11(2):370-382. doi:10.1158/1535-7163.MCT-11-0458
    • (2012) Mol Cancer Ther , vol.11 , Issue.2 , pp. 370-382
    • Chen, M.1    Shabashvili, D.2    Nawab, A.3
  • 35
    • 84874041667 scopus 로고    scopus 로고
    • Chaetocin is a nonspecific inhibitor of histone lysine methyltransferases
    • doi:10.1038/nchembio.1187
    • Cherblanc FL, Chapman KL, Brown R, Fuchter MJ (2013) Chaetocin is a nonspecific inhibitor of histone lysine methyltransferases. Nat Chem Biol 9(3):136-137. doi:10.1038/nchembio.1187
    • (2013) Nat Chem Biol , vol.9 , Issue.3 , pp. 136-137
    • Cherblanc, F.L.1    Chapman, K.L.2    Brown, R.3    Fuchter, M.J.4
  • 36
    • 77956246016 scopus 로고    scopus 로고
    • The histone deacetylase inhibitor MGCD0103 has both deacetylase and microtubule inhibitory activity
    • doi:10.1124/mol.110.065169
    • Chia K, Beamish H, Jafferi K, Gabrielli B (2010) The histone deacetylase inhibitor MGCD0103 has both deacetylase and microtubule inhibitory activity. Mol Pharmacol 78(3):436-443. doi:10.1124/mol.110.065169
    • (2010) Mol Pharmacol , vol.78 , Issue.3 , pp. 436-443
    • Chia, K.1    Beamish, H.2    Jafferi, K.3    Gabrielli, B.4
  • 37
    • 84867325687 scopus 로고    scopus 로고
    • Characterisation of a Tip60 specific inhibitor, NU9056, in prostate cancer
    • doi:10.1371/journal.pone.0045539
    • Coffey K, Blackburn TJ, Cook S et al (2012) Characterisation of a Tip60 specific inhibitor, NU9056, in prostate cancer. PLoS ONE 7(10):e45539. doi:10.1371/journal.pone.0045539
    • (2012) PLoS ONE , vol.7 , Issue.10
    • Coffey, K.1    Blackburn, T.J.2    Cook, S.3
  • 38
    • 84872942313 scopus 로고    scopus 로고
    • Differential effects of garcinol and curcumin on histone and p53 modifications in tumour cells
    • doi:10.1186/1471-2407-13-37
    • Collins HM, Abdelghany MK, Messmer M et al (2013) Differential effects of garcinol and curcumin on histone and p53 modifications in tumour cells. BMC Cancer 13:37. doi:10.1186/1471-2407-13-37
    • (2013) BMC Cancer , vol.13 , pp. 37
    • Collins, H.M.1    Abdelghany, M.K.2    Messmer, M.3
  • 39
    • 84867550050 scopus 로고    scopus 로고
    • Molecular pathways: The complexity of the epigenome in cancer and recent clinical advances
    • doi:10.1158/1078-0432.CR-12-2037
    • Conte M, Altucci L (2012) Molecular pathways: the complexity of the epigenome in cancer and recent clinical advances. Clin Cancer Res 18(20):5526-5534. doi:10.1158/1078-0432.CR-12-2037
    • (2012) Clin Cancer Res , vol.18 , Issue.20 , pp. 5526-5534
    • Conte, M.1    Altucci, L.2
  • 40
    • 79960058024 scopus 로고    scopus 로고
    • Selective killing of mixed lineage leukemia cells by a potent smallmolecule DOT1L inhibitor
    • doi:10.1016/j.ccr.2011.06.009
    • Daigle SR, Olhava EJ, Therkelsen CA et al (2011) Selective killing of mixed lineage leukemia cells by a potent smallmolecule DOT1L inhibitor. Cancer Cell 20(1):53-65. doi:10.1016/j.ccr.2011.06.009
    • (2011) Cancer Cell , vol.20 , Issue.1 , pp. 53-65
    • Daigle, S.R.1    Olhava, E.J.2    Therkelsen, C.A.3
  • 41
    • 84886864184 scopus 로고    scopus 로고
    • Potent inhibition of DOT1L as treatment of ML-fusion leukemia
    • doi:10.1182/blood-2013-04-497644
    • Daigle SR, Olhava EJ, Therkelsen CA et al (2013) Potent inhibition of DOT1L as treatment of ML-fusion leukemia. Blood 122(6):1017-1025. doi:10.1182/blood-2013-04-497644
    • (2013) Blood , vol.122 , Issue.6 , pp. 1017-1025
    • Daigle, S.R.1    Olhava, E.J.2    Therkelsen, C.A.3
  • 42
    • 75149188170 scopus 로고    scopus 로고
    • Systematic sequencing of renal carcinoma reveals inactivation of histone modifying genes
    • doi:10.1038/nature08672
    • Dalgliesh GL, Furge K, Greenman C et al (2010) Systematic sequencing of renal carcinoma reveals inactivation of histone modifying genes. Nature 463(7279):360-363. doi:10.1038/nature08672
    • (2010) Nature , vol.463 , Issue.7279 , pp. 360-363
    • Dalgliesh, G.L.1    Furge, K.2    Greenman, C.3
  • 44
    • 80054984945 scopus 로고    scopus 로고
    • Inhibition of BET recruitment to chromatin as an effective treatment for MLfusion leukaemia
    • doi:10.1038/ nature10509
    • Dawson MA, Prinjha RK, Dittmann A et al (2011) Inhibition of BET recruitment to chromatin as an effective treatment for MLfusion leukaemia. Nature 478(7370):529-533. doi:10.1038/ nature10509
    • (2011) Nature , vol.478 , Issue.7370 , pp. 529-533
    • Dawson, M.A.1    Prinjha, R.K.2    Dittmann, A.3
  • 45
    • 84865005165 scopus 로고    scopus 로고
    • Targeting epigenetic readers in cancer
    • doi:10.1056/NEJMra1112635
    • Dawson MA, Kouzarides T, Huntly BJ (2012) Targeting epigenetic readers in cancer. N Engl J Med 367(7):647-657. doi:10.1056/NEJMra1112635
    • (2012) N Engl J Med , vol.367 , Issue.7 , pp. 647-657
    • Dawson, M.A.1    Kouzarides, T.2    Huntly, B.J.3
  • 46
    • 84893735484 scopus 로고    scopus 로고
    • Recurrent mutations, including NPM1c, activate a BRD4-dependent core transcriptional program in acute myeloid leukemia
    • doi:10.1038/leu.2013.338
    • Dawson MA, Gudgin EJ, Horton SJ et al (2014) Recurrent mutations, including NPM1c, activate a BRD4-dependent core transcriptional program in acute myeloid leukemia. Leukemia 28(2):311-320. doi:10.1038/leu.2013.338
    • (2014) Leukemia , vol.28 , Issue.2 , pp. 311-320
    • Dawson, M.A.1    Gudgin, E.J.2    Horton, S.J.3
  • 47
    • 80052955256 scopus 로고    scopus 로고
    • BET bromodomain inhibition as a therapeutic strategy to target c-Myc
    • doi:10.1016/j.cell.2011.08.017
    • Delmore JE, Issa GC, Lemieux ME et al (2011) BET bromodomain inhibition as a therapeutic strategy to target c-Myc. Cell 146(6):904-917. doi:10.1016/j.cell.2011.08.017
    • (2011) Cell , vol.146 , Issue.6 , pp. 904-917
    • Delmore, J.E.1    Issa, G.C.2    Lemieux, M.E.3
  • 48
    • 47749085845 scopus 로고    scopus 로고
    • Long-term effects of resveratrol supplementation on suppression of atherogenic lesion formation and cholesterol synthesis in apo E-deficient mice
    • doi:10.1016/j.bbrc.2008.06.113
    • Do GM, Kwon EY, Kim HJ et al (2008) Long-term effects of resveratrol supplementation on suppression of atherogenic lesion formation and cholesterol synthesis in apo E-deficient mice. Biochem Biophys Res Commun 374(1):55-59. doi:10.1016/j.bbrc.2008.06.113
    • (2008) Biochem Biophys Res Commun , vol.374 , Issue.1 , pp. 55-59
    • Do, G.M.1    Kwon, E.Y.2    Kim, H.J.3
  • 49
    • 35548988945 scopus 로고    scopus 로고
    • Vorinostat in cutaneous T-cell lymphoma
    • DOI 10.1358/dot.2007.43.9.1112980
    • Duvic M, Vu J (2007) Vorinostat in cutaneous T-cell lymphoma. Drugs Today 43(9):585-599. doi:10.1358/dot.2007.43.9.1112980 (Pubitemid 350004865)
    • (2007) Drugs of Today , vol.43 , Issue.9 , pp. 585-599
    • Duvic, M.1    Vu, J.2
  • 50
    • 84905679330 scopus 로고    scopus 로고
    • Disruption of autophagy by the histone deacetylase inhibitor MGCD0103 and its therapeutic implication in B-cell chronic lymphocytic leukemia
    • doi:10.1038/leu.2014.19
    • El-Khoury V, Pierson S, Szwarcbart E et al (2014) Disruption of autophagy by the histone deacetylase inhibitor MGCD0103 and its therapeutic implication in B-cell chronic lymphocytic leukemia. Leukemia. doi:10.1038/leu.2014.19
    • (2014) Leukemia
    • El-Khoury, V.1    Pierson, S.2    Szwarcbart, E.3
  • 51
    • 51649126841 scopus 로고    scopus 로고
    • Histone deacetylase inhibitor panobinostat induces clinical responses with associated alterations in gene expression profiles in cutaneous T-cell lymphoma
    • doi:10.1158/1078-0432.CR-07-4262
    • Ellis L, Pan Y, Smyth GK et al (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.CR-07-4262
    • (2008) Clin Cancer Res , vol.14 , Issue.14 , pp. 4500-4510
    • Ellis, L.1    Pan, Y.2    Smyth, G.K.3
  • 52
    • 0021772472 scopus 로고
    • Differential inhibition of histone and polyamine acetylases by multisubstrate analogues
    • Erwin BG, Persson L, Pegg AE (1984) Differential inhibition of histone and polyamine acetylases by multisubstrate analogues. Biochemistry 23(18):4250-4255
    • (1984) Biochemistry , vol.23 , Issue.18 , pp. 4250-4255
    • Erwin, B.G.1    Persson, L.2    Pegg, A.E.3
  • 53
    • 33947315736 scopus 로고    scopus 로고
    • Cancer epigenomics: DNA methylomes and histone-modification maps
    • doi:10.1038/nrg2005
    • Esteller M (2007) Cancer epigenomics: DNA methylomes and histone-modification maps. Nat Rev Genet 8(4):286-298. doi:10.1038/nrg2005
    • (2007) Nat Rev Genet , vol.8 , Issue.4 , pp. 286-298
    • Esteller, M.1
  • 54
    • 40849139208 scopus 로고    scopus 로고
    • Epigenetics in cancer
    • doi:10.1056/NEJMra072067
    • Esteller M (2008) Epigenetics in cancer. N Engl J Med 358(11):1148- 1159. doi:10.1056/NEJMra072067
    • (2008) N Engl J Med , vol.358 , Issue.11 , pp. 1148-1159
    • Esteller, M.1
  • 55
    • 2342624735 scopus 로고    scopus 로고
    • The study of 5-Aza-2′-deoxycytidine on transcription regulation of p16/CDKN2 gene demethylation in RKO human colorectal cell line
    • Fang XM, Sun LF, Peng JP, Dong Q, Zheng S (2003) The study of 5-Aza-2′-deoxycytidine on transcription regulation of p16/CDKN2 gene demethylation in RKO human colorectal cell line. Zhonghua yi xue za zhi 83(23):2077-2082
    • (2003) Zhonghua Yi Xue Za Zhi , vol.83 , Issue.23 , pp. 2077-2082
    • Fang, X.M.1    Sun, L.F.2    Peng, J.P.3    Dong, Q.4    Zheng, S.5
  • 56
    • 78650847770 scopus 로고    scopus 로고
    • Selective inhibition of BET bromodomains
    • doi:10.1038/nature09504
    • Filippakopoulos P, Qi J, Picaud S et al (2010) Selective inhibition of BET bromodomains. Nature 468(7327):1067-1073. doi:10.1038/nature09504
    • (2010) Nature , vol.468 , Issue.7327 , pp. 1067-1073
    • Filippakopoulos, P.1    Qi, J.2    Picaud, S.3
  • 57
    • 67949102053 scopus 로고    scopus 로고
    • Recent progress in the biology and physiology of sirtuins
    • doi:10.1038/nature08197
    • Finkel T, Deng CX, Mostoslavsky R (2009) Recent progress in the biology and physiology of sirtuins. Nature 460(7255):587-591. doi:10.1038/nature08197
    • (2009) Nature , vol.460 , Issue.7255 , pp. 587-591
    • Finkel, T.1    Deng, C.X.2    Mostoslavsky, R.3
  • 58
    • 16344368814 scopus 로고    scopus 로고
    • Histone demethylation catalysed by LSD1 is a flavin-dependent oxidative process
    • DOI 10.1016/j.febslet.2005.03.015
    • Forneris F, Binda C, Vanoni MA, Mattevi A, Battaglioli E (2005) Histone demethylation catalysed by LSD1 is a flavin-dependent oxidative process. FEBS Lett 579(10):2203-2207. doi:10.1016/j.febslet.2005.03.015 (Pubitemid 40469693)
    • (2005) FEBS Letters , vol.579 , Issue.10 , pp. 2203-2207
    • Forneris, F.1    Binda, C.2    Vanoni, M.A.3    Mattevi, A.4    Battaglioli, E.5
  • 59
    • 84979581717 scopus 로고    scopus 로고
    • The class I-specific HDAC inhibitor MS-275 modulates the differentiation potential of mouse embryonic stem cells
    • doi:10.1242/bio.20135587
    • Franci G, Casalino L, Petraglia F et al (2013) The class I-specific HDAC inhibitor MS-275 modulates the differentiation potential of mouse embryonic stem cells. Biol Open 2(10):1070-1077. doi:10.1242/bio.20135587
    • (2013) Biol Open , vol.2 , Issue.10 , pp. 1070-1077
    • Franci, G.1    Casalino, L.2    Petraglia, F.3
  • 60
    • 84894797638 scopus 로고    scopus 로고
    • Histone deacetylase inhibition in the treatment of acute myeloid leukemia: The effects of valproic acid on leukemic cells, and the clinical and experimental evidence for combining valproic acid with other antileukemic agents
    • doi:10.1186/1868-7083-5-12
    • Fredly H, Gjertsen BT, Bruserud O (2013) Histone deacetylase inhibition in the treatment of acute myeloid leukemia: the effects of valproic acid on leukemic cells, and the clinical and experimental evidence for combining valproic acid with other antileukemic agents. Clin Epigenet 5(1):12. doi:10.1186/1868-7083-5-12
    • (2013) Clin Epigenet , vol.5 , Issue.1 , pp. 12
    • Fredly, H.1    Gjertsen, B.T.2    Bruserud, O.3
  • 62
    • 84862554715 scopus 로고    scopus 로고
    • Myelodysplastic syndromes: 2012 update on diagnosis, risk-stratification, and management
    • doi:10.1002/ajh.23264
    • Garcia-Manero G (2012) Myelodysplastic syndromes: 2012 update on diagnosis, risk-stratification, and management. Am J Hematol 87(7):692-701. doi:10.1002/ajh.23264
    • (2012) Am J Hematol , vol.87 , Issue.7 , pp. 692-701
    • Garcia-Manero, G.1
  • 63
    • 20844444898 scopus 로고    scopus 로고
    • 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
    • DOI 10.1182/blood-2004-09-3413
    • George P, Bali P, Annavarapu S et al (2005) 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. Blood 105(4):1768-1776. doi:10.1182/blood-2004-09-3413 (Pubitemid 40223701)
    • (2005) Blood , vol.105 , Issue.4 , pp. 1768-1776
    • George, P.1    Bali, P.2    Annavarapu, S.3    Scuto, A.4    Fiskus, W.5    Guo, F.6    Sigua, C.7    Sondarva, G.8    Moscinski, L.9    Atadja, P.10    Bhalla, K.11
  • 65
    • 0036861629 scopus 로고    scopus 로고
    • DNA methyltransferase inhibitors-state of the art
    • Goffin J, Eisenhauer E (2002) DNA methyltransferase inhibitors-state of the art. Ann Oncol 13(11):1699-1716
    • (2002) Ann Oncol , vol.13 , Issue.11 , pp. 1699-1716
    • Goffin, J.1    Eisenhauer, E.2
  • 67
    • 84903700216 scopus 로고    scopus 로고
    • Anti-tumoral effect of the non-nucleoside DNMT inhibitor RG108 in human prostate cancer cells
    • Graca I, Sousa EJ, Baptista T et al (2014) Anti-tumoral effect of the non-nucleoside DNMT inhibitor RG108 in human prostate cancer cells. Curr Pharm Des 20(11):1803-1811
    • (2014) Curr Pharm des , vol.20 , Issue.11 , pp. 1803-1811
    • Graca, I.1    Sousa, E.J.2    Baptista, T.3
  • 69
    • 78649327715 scopus 로고    scopus 로고
    • Biological rationale for the use of DNA methyltransferase inhibitors as new strategy for modulation of tumor response to chemotherapy and radiation
    • doi:10.1186/1476-4598-9-305
    • Gravina GL, Festuccia C, Marampon F et al (2010) Biological rationale for the use of DNA methyltransferase inhibitors as new strategy for modulation of tumor response to chemotherapy and radiation. Mol Cancer 9:305. doi:10.1186/1476-4598-9-305
    • (2010) Mol Cancer , vol.9 , pp. 305
    • Gravina, G.L.1    Festuccia, C.2    Marampon, F.3
  • 70
    • 50349086621 scopus 로고    scopus 로고
    • The potential role of histone deacetylase inhibitors in the treatment of non-small-cell lung cancer
    • doi:10.1016/j.cr itrevonc.2008.03.002
    • Gridelli C, Rossi A, Maione P (2008) The potential role of histone deacetylase inhibitors in the treatment of non-small-cell lung cancer. Crit Rev Oncol Hematol 68(1):29-36. doi:10.1016/j.cr itrevonc.2008.03.002
    • (2008) Crit Rev Oncol Hematol , vol.68 , Issue.1 , pp. 29-36
    • Gridelli, C.1    Rossi, A.2    Maione, P.3
  • 71
    • 78851469815 scopus 로고    scopus 로고
    • Chemosensitization of tumors by resveratrol
    • doi:10.1111/j.1749-6632.2010.05852.x
    • Gupta SC, Kannappan R, Reuter S, Kim JH, Aggarwal BB (2011) Chemosensitization of tumors by resveratrol. Ann N Y Acad Sci 1215:150-160. doi:10.1111/j.1749-6632.2010.05852.x
    • (2011) Ann N Y Acad Sci , vol.1215 , pp. 150-160
    • Gupta, S.C.1    Kannappan, R.2    Reuter, S.3    Kim, J.H.4    Aggarwal, B.B.5
  • 72
    • 84863115185 scopus 로고    scopus 로고
    • Discovery of curcumin, a component of golden spice, and its miraculous biological activities
    • doi:10.1111/j.1440-1681.2011.05648.x
    • Gupta SC, Patchva S, Koh W, Aggarwal BB (2012) Discovery of curcumin, a component of golden spice, and its miraculous biological activities. Clin Exp Pharmacol Physiol 39(3):283-299. doi:10.1111/j.1440-1681.2011.05648.x
    • (2012) Clin Exp Pharmacol Physiol , vol.39 , Issue.3 , pp. 283-299
    • Gupta, S.C.1    Patchva, S.2    Koh, W.3    Aggarwal, B.B.4
  • 73
    • 84859837026 scopus 로고    scopus 로고
    • The histone demethylase KDM1A sustains the oncogenic potential of MLAF9 leukemia stem cells
    • doi:10.1016/j.ccr.2012.03.014
    • Harris WJ, Huang X, Lynch JT et al (2012) The histone demethylase KDM1A sustains the oncogenic potential of MLAF9 leukemia stem cells. Cancer Cell 21(4):473-487. doi:10.1016/j.ccr.2012.03.014
    • (2012) Cancer Cell , vol.21 , Issue.4 , pp. 473-487
    • Harris, W.J.1    Huang, X.2    Lynch, J.T.3
  • 74
    • 79958769268 scopus 로고    scopus 로고
    • Epigenetic aberrations during oncogenesis
    • doi:10.1007/ s00018-010-0624-z
    • Hatziapostolou M, Iliopoulos D (2011) Epigenetic aberrations during oncogenesis. Cell Mol Life Sci 68(10):1681-1702. doi:10.1007/ s00018-010-0624-z
    • (2011) Cell Mol Life Sci , vol.68 , Issue.10 , pp. 1681-1702
    • Hatziapostolou, M.1    Iliopoulos, D.2
  • 75
    • 84886808679 scopus 로고    scopus 로고
    • Chromatin proteins and modifications as drug targets
    • doi:10.1038/nature12751
    • Helin K, Dhanak D (2013) Chromatin proteins and modifications as drug targets. Nature 502(7472):480-488. doi:10.1038/nature12751
    • (2013) Nature , vol.502 , Issue.7472 , pp. 480-488
    • Helin, K.1    Dhanak, D.2
  • 76
    • 33646254136 scopus 로고    scopus 로고
    • Antitumor activity of a small-molecule inhibitor of human silent information regulator 2 enzymes
    • doi:10.1158/0008-5472.CAN-05-3617
    • Heltweg B, Gatbonton T, Schuler AD et al (2006) Antitumor activity of a small-molecule inhibitor of human silent information regulator 2 enzymes. Cancer Res 66(8):4368-4377. doi:10.1158/0008-5472.CAN-05-3617
    • (2006) Cancer Res , vol.66 , Issue.8 , pp. 4368-4377
    • Heltweg, B.1    Gatbonton, T.2    Schuler, A.D.3
  • 77
    • 12344267827 scopus 로고    scopus 로고
    • Simultaneous detection of six stone fruit viruses by non-isotopic molecular hybridization using a unique riboprobe or 'polyprobe'
    • DOI 10.1016/j.jviromet.2004.11.003, PII S0166093404003362
    • Herranz MC, Sanchez-Navarro JA, Aparicio F, Pallas V (2005) Simultaneous detection of six stone fruit viruses by non-isotopic molecular hybridization using a unique riboprobe or 'polyprobe'. J Virol Methods 124(1-2):49-55. doi:10.1016/j.jviromet.2004.11.003 (Pubitemid 40138780)
    • (2005) Journal of Virological Methods , vol.124 , Issue.1-2 , pp. 49-55
    • Herranz, M.C.1    Sanchez-Navarro, J.A.2    Aparicio, F.3    Pallas, V.4
  • 78
    • 31544439102 scopus 로고    scopus 로고
    • The novel DNA methylation inhibitor zebularine is effective against the development of murine T-cell lymphoma
    • DOI 10.1182/blood-2005-05-2033
    • Herranz M, Martin-Caballero J, Fraga MF et al (2006) The novel DNA methylation inhibitor zebularine is effective against the development of murine T-cell lymphoma. Blood 107(3):1174-1177. doi:10.1182/blood-2005-05-2033 (Pubitemid 43156320)
    • (2006) Blood , vol.107 , Issue.3 , pp. 1174-1177
    • Herranz, M.1    Martin-Caballero, J.2    Fraga, M.F.3    Ruiz-Cabello, J.4    Flores, J.M.5    Desco, M.6    Marquez, V.7    Esteller, M.8
  • 80
    • 84889573275 scopus 로고    scopus 로고
    • Histone lysine demethylases as targets for anticancer therapy
    • doi:10.1038/nrd4154
    • Hojfeldt JW, Agger K, Helin K (2013) Histone lysine demethylases as targets for anticancer therapy. Nat Rev Drug Discovery 12(12):917-930. doi:10.1038/nrd4154
    • (2013) Nat Rev Drug Discovery , vol.12 , Issue.12 , pp. 917-930
    • Hojfeldt, J.W.1    Agger, K.2    Helin, K.3
  • 82
    • 35948981189 scopus 로고    scopus 로고
    • Solution structure of the second bromodomain of Brd2 and its specific interaction with acetylated histone tails
    • doi:10.1186/1472-6807-7-57
    • Huang H, Zhang J, Shen W et al (2007) Solution structure of the second bromodomain of Brd2 and its specific interaction with acetylated histone tails. BMC Struct Biol 7:57. doi:10.1186/1472-6807-7-57
    • (2007) BMC Struct Biol , vol.7 , pp. 57
    • Huang, H.1    Zhang, J.2    Shen, W.3
  • 83
    • 33947214077 scopus 로고    scopus 로고
    • Chaetocin: A promising new antimyeloma agent with in vitro and in vivo activity mediated via imposition of oxidative stress
    • DOI 10.1182/blood-2006-07-027326
    • Isham CR, Tibodeau JD, Jin W, Xu R, Timm MM, Bible KC (2007) Chaetocin: a promising new antimyeloma agent with in vitro and in vivo activity mediated via imposition of oxidative stress. Blood 109(6):2579-2588. doi:10.1182/blood-2006- 07-027326 (Pubitemid 46425905)
    • (2007) Blood , vol.109 , Issue.6 , pp. 2579-2588
    • Isham, C.R.1    Tibodeau, J.D.2    Jin, W.3    Xu, R.4    Timm, M.M.5    Bible, K.C.6
  • 84
    • 80052461558 scopus 로고    scopus 로고
    • Tet proteins can convert 5-methylcytosine to 5-formylcytosine and 5-carboxylcytosine
    • doi:10.1126/science.1210597
    • Ito S, Shen L, Dai Q et al (2011) Tet proteins can convert 5-methylcytosine to 5-formylcytosine and 5-carboxylcytosine. Science 333(6047):1300-1303. doi:10.1126/science.1210597
    • (2011) Science , vol.333 , Issue.6047 , pp. 1300-1303
    • Ito, S.1    Shen, L.2    Dai, Q.3
  • 86
    • 84255200412 scopus 로고    scopus 로고
    • 5-Hydroxymethylcytosine is strongly depleted in human cancers but its levels do not correlate with IDH1 mutations
    • doi:10.1158/0008-5472.CAN-11-2023
    • Jin SG, Jiang Y, Qiu R et al (2011) 5-Hydroxymethylcytosine is strongly depleted in human cancers but its levels do not correlate with IDH1 mutations. Cancer Res 71(24):7360-7365. doi:10.1158/0008-5472.CAN-11-2023
    • (2011) Cancer Res , vol.71 , Issue.24 , pp. 7360-7365
    • Jin, S.G.1    Jiang, Y.2    Qiu, R.3
  • 87
    • 33847065486 scopus 로고    scopus 로고
    • The Epigenomics of Cancer
    • DOI 10.1016/j.cell.2007.01.029, PII S0092867407001274
    • Jones PA, Baylin SB (2007) The epigenomics of cancer. Cell 128(4):683-692. doi:10.1016/j.cell.2007.01.029 (Pubitemid 46273572)
    • (2007) Cell , vol.128 , Issue.4 , pp. 683-692
    • Jones, P.A.1    Baylin, S.B.2
  • 88
    • 70349323610 scopus 로고    scopus 로고
    • SIRT1 controls circadian clock circuitry and promotes cell survival: A connection with age-related neoplasms
    • doi:10.1096/fj.09-129148
    • Jung-Hynes B, Ahmad N (2009) SIRT1 controls circadian clock circuitry and promotes cell survival: a connection with age-related neoplasms. FASEB J 23(9):2803-2809. doi:10.1096/fj.09-129148
    • (2009) FASEB J , vol.23 , Issue.9 , pp. 2803-2809
    • Jung-Hynes, B.1    Ahmad, N.2
  • 89
    • 0028151343 scopus 로고
    • Toxicity of 5-aza-2′-deoxycytidine to mammalian cells is mediated primarily by covalent trapping of DNA methyltransferase rather than DNA demethylation
    • Juttermann R, Li E, Jaenisch R (1994) Toxicity of 5-aza-2′- deoxycytidine to mammalian cells is mediated primarily by covalent trapping of DNA methyltransferase rather than DNA demethylation. Proc Natl Acad Sci USA 91(25):11797-11801
    • (1994) Proc Natl Acad Sci USA , vol.91 , Issue.25 , pp. 11797-11801
    • Juttermann, R.1    Li, E.2    Jaenisch, R.3
  • 90
    • 80053214606 scopus 로고    scopus 로고
    • Failure of hypomethylating agent-based therapy in myelodysplastic syndromes
    • doi:10.1053/j.seminoncol.2011.04.011
    • Kadia TM, Jabbour E, Kantarjian H (2011) Failure of hypomethylating agent-based therapy in myelodysplastic syndromes. Semin Oncol 38(5):682-692. doi:10.1053/j.seminoncol.2011.04.011
    • (2011) Semin Oncol , vol.38 , Issue.5 , pp. 682-692
    • Kadia, T.M.1    Jabbour, E.2    Kantarjian, H.3
  • 91
    • 84899486027 scopus 로고    scopus 로고
    • Romidepsin induces cell cycle arrest, apoptosis, histone hyperacetylation and reduces matrix metalloproteinases 2 and 9 expression in bortezomib sensitized non-small cell lung cancer cells
    • doi:10.1016/j.biopha.2014.01.002
    • Karthik S, Sankar R, Varunkumar K, Ravikumar V (2014) Romidepsin induces cell cycle arrest, apoptosis, histone hyperacetylation and reduces matrix metalloproteinases 2 and 9 expression in bortezomib sensitized non-small cell lung cancer cells. Biomed Pharmacother 68(3):327-334. doi:10.1016/j.biopha.2014. 01.002
    • (2014) Biomed Pharmacother , vol.68 , Issue.3 , pp. 327-334
    • Karthik, S.1    Sankar, R.2    Varunkumar, K.3    Ravikumar, V.4
  • 92
    • 84855471990 scopus 로고    scopus 로고
    • HDAC inhibitors in cancer biology: Emerging mechanisms and clinical applications
    • doi:10.1038/icb.2011.100
    • Khan O, La Thangue NB (2012) HDAC inhibitors in cancer biology: emerging mechanisms and clinical applications. Immunol Cell Biol 90(1):85-94. doi:10.1038/icb.2011.100
    • (2012) Immunol Cell Biol , vol.90 , Issue.1 , pp. 85-94
    • Khan, O.1    La Thangue, N.B.2
  • 93
    • 32944469082 scopus 로고    scopus 로고
    • A decade of histone acetylation: Marking eukaryotic chromosomes with specific codes
    • DOI 10.1093/jb/mvi184
    • Kimura A, Matsubara K, Horikoshi M (2005) A decade of histone acetylation: marking eukaryotic chromosomes with specific codes. J Biochem 138(6):647-662. doi:10.1093/jb/mvi184 (Pubitemid 43258858)
    • (2005) Journal of Biochemistry , vol.138 , Issue.6 , pp. 647-662
    • Kimura, A.1    Matsubara, K.2    Horikoshi, M.3
  • 94
    • 79957623760 scopus 로고    scopus 로고
    • Mutations in DNMT1 cause hereditary sensory neuropathy with dementia and hearing loss
    • doi:10.1038/ng.830
    • Klein CJ, Botuyan MV, Wu Y et al (2011) Mutations in DNMT1 cause hereditary sensory neuropathy with dementia and hearing loss. Nat Genet 43(6):595-600. doi:10.1038/ng.830
    • (2011) Nat Genet , vol.43 , Issue.6 , pp. 595-600
    • Klein, C.J.1    Botuyan, M.V.2    Wu, Y.3
  • 96
    • 84877324084 scopus 로고    scopus 로고
    • Durable tumor regression in genetically altered malignant rhabdoid tumors by inhibition of methyltransferase EZH2
    • doi:10.1073/pnas.1303800110
    • Knutson SK, Warholic NM, Wigle TJ et al (2013) Durable tumor regression in genetically altered malignant rhabdoid tumors by inhibition of methyltransferase EZH2. Proc Natl Acad Sci USA 110(19):7922-7927. doi:10.1073/pnas.1303800110
    • (2013) Proc Natl Acad Sci USA , vol.110 , Issue.19 , pp. 7922-7927
    • Knutson, S.K.1    Warholic, N.M.2    Wigle, T.J.3
  • 97
    • 84860215207 scopus 로고    scopus 로고
    • Molecular mechanisms and potential functions of histone demethylases
    • doi:10.1038/nrm3327
    • Kooistra SM, Helin K (2012) Molecular mechanisms and potential functions of histone demethylases. Nat Rev Mol Cell Biol 13(5):297-311. doi:10.1038/nrm3327
    • (2012) Nat Rev Mol Cell Biol , vol.13 , Issue.5 , pp. 297-311
    • Kooistra, S.M.1    Helin, K.2
  • 98
    • 33847076849 scopus 로고    scopus 로고
    • Chromatin modifications and their function
    • doi:10.1016/j.cell.2007.02.005
    • Kouzarides T (2007) Chromatin modifications and their function. Cell 128(4):693-705. doi:10.1016/j.cell.2007.02.005
    • (2007) Cell , vol.128 , Issue.4 , pp. 693-705
    • Kouzarides, T.1
  • 99
    • 84865120905 scopus 로고    scopus 로고
    • A selective jumonji H3K27 demethylase inhibitor modulates the proinflammatory macrophage response
    • doi:10.1038/nature11262
    • Kruidenier L, Chung CW, Cheng Z et al (2012) A selective jumonji H3K27 demethylase inhibitor modulates the proinflammatory macrophage response. Nature 488(7411):404-408. doi:10.1038/nature11262
    • (2012) Nature , vol.488 , Issue.7411 , pp. 404-408
    • Kruidenier, L.1    Chung, C.W.2    Cheng, Z.3
  • 101
    • 84871822817 scopus 로고    scopus 로고
    • Vorinostat, an HDAC inhibitor attenuates epidermoid squamous cell carcinoma growth by dampening mTOR signaling pathway in a human xenograft murine model
    • doi:10.1016/j.taap.2012.11.002
    • Kurundkar D, Srivastava RK, Chaudhary SC et al (2013) Vorinostat, an HDAC inhibitor attenuates epidermoid squamous cell carcinoma growth by dampening mTOR signaling pathway in a human xenograft murine model. Toxicol Appl Pharmacol 266(2):233-244. doi:10.1016/j.taap.2012.11.002
    • (2013) Toxicol Appl Pharmacol , vol.266 , Issue.2 , pp. 233-244
    • Kurundkar, D.1    Srivastava, R.K.2    Chaudhary, S.C.3
  • 102
    • 0023130877 scopus 로고
    • Turmeric and curcumin as topical agents in cancer therapy
    • Kuttan R, Sudheeran PC, Josph CD (1987) Turmeric and curcumin as topical agents in cancer therapy. Tumori 73(1):29-31 (Pubitemid 17034485)
    • (1987) Tumori , vol.73 , Issue.1 , pp. 29-31
    • Kuttan, R.1    Sudheeran, P.C.2    Josph, C.D.3
  • 103
    • 0033714888 scopus 로고    scopus 로고
    • HATs off: Selective synthetic inhibitors of the histone acetyltransferases p300 and PCAF
    • Lau OD, Kundu TK, Soccio RE et al (2000) HATs off: selective synthetic inhibitors of the histone acetyltransferases p300 and PCAF. Mol Cell 5(3):589-595
    • (2000) Mol Cell , vol.5 , Issue.3 , pp. 589-595
    • Lau, O.D.1    Kundu, T.K.2    Soccio, R.E.3
  • 104
    • 28844497663 scopus 로고    scopus 로고
    • Procainamide is a specific inhibitor of DNA methyltransferase 1
    • DOI 10.1074/jbc.M505593200
    • Lee BH, Yegnasubramanian S, Lin X, Nelson WG (2005a) Procainamide is a specific inhibitor of DNA methyltransferase 1. J Biol Chem 280(49):40749-40756. doi:10.1074/jbc.M505593200 (Pubitemid 41780565)
    • (2005) Journal of Biological Chemistry , vol.280 , Issue.49 , pp. 40749-40756
    • Lee, B.H.1    Yegnasubramanian, S.2    Lin, X.3    Nelson, W.G.4
  • 105
    • 25144507259 scopus 로고    scopus 로고
    • Mechanisms for the inhibition of DNA methyltransferases by tea catechins and bioflavonoids
    • doi:10.1124/mol.104.008367
    • Lee WJ, Shim JY, Zhu BT (2005b) Mechanisms for the inhibition of DNA methyltransferases by tea catechins and bioflavonoids. Mol Pharmacol 68(4):1018-1030. doi:10.1124/mol.104.008367
    • (2005) Mol Pharmacol , vol.68 , Issue.4 , pp. 1018-1030
    • Lee, W.J.1    Shim, J.Y.2    Zhu, B.T.3
  • 106
    • 77957091318 scopus 로고    scopus 로고
    • Histone deacetylase inhibitor induces DNA damage, which normal but not transformed cells can repair
    • doi:10.1073/pnas.1008522107
    • Lee JH, Choy ML, Ngo L, Foster SS, Marks PA (2010) Histone deacetylase inhibitor induces DNA damage, which normal but not transformed cells can repair. Proc Natl Acad Sci USA 107(33):14639-14644. doi:10.1073/pnas.1008522107
    • (2010) Proc Natl Acad Sci USA , vol.107 , Issue.33 , pp. 14639-14644
    • Lee, J.H.1    Choy, M.L.2    Ngo, L.3    Foster, S.S.4    Marks, P.A.5
  • 107
    • 83755195245 scopus 로고    scopus 로고
    • Role of checkpoint kinase 1 (Chk1) in the mechanisms of resistance to histone deacetylase inhibitors
    • doi:10.1073/pnas.1117544108
    • Lee JH, Choy ML, Ngo L, Venta-Perez G, Marks PA (2011) Role of checkpoint kinase 1 (Chk1) in the mechanisms of resistance to histone deacetylase inhibitors. Proc Natl Acad Sci USA 108(49):19629-19634. doi:10.1073/pnas. 1117544108
    • (2011) Proc Natl Acad Sci USA , vol.108 , Issue.49 , pp. 19629-19634
    • Lee, J.H.1    Choy, M.L.2    Ngo, L.3    Venta-Perez, G.4    Marks, P.A.5
  • 108
    • 0036902962 scopus 로고    scopus 로고
    • DNA methylation and demethylating drugs in myelodysplastic syndromes and secondary leukemias
    • Leone G, Teofili L, Voso MT, Lubbert M (2002) DNA methylation and demethylating drugs in myelodysplastic syndromes and secondary leukemias. Haematologica 87(12):1324-1341 (Pubitemid 36005306)
    • (2002) Haematologica , vol.87 , Issue.12 , pp. 1324-1341
    • Leone, G.1    Teofili, L.2    Voso, M.T.3    Lubbert, M.4
  • 109
    • 78649906060 scopus 로고    scopus 로고
    • DNMT3A mutations in acute myeloid leukemia
    • doi:10.1056/NEJMoa1005143
    • Ley TJ, Ding L, Walter MJ et al (2010) DNMT3A mutations in acute myeloid leukemia. N Engl J Med 363(25):2424-2433. doi:10.1056/NEJMoa1005143
    • (2010) N Engl J Med , vol.363 , Issue.25 , pp. 2424-2433
    • Ley, T.J.1    Ding, L.2    Walter, M.J.3
  • 110
    • 73249124141 scopus 로고    scopus 로고
    • Discovery of a 2,4-diamino-7-aminoalkoxyquinazoline as a potent and selective inhibitor of histone lysine methyltransferase G9a
    • doi:10.1021/Jm901543m
    • Liu F, Chen X, Allali-Hassani A et al (2009) Discovery of a 2,4-diamino-7-aminoalkoxyquinazoline as a potent and selective inhibitor of histone lysine methyltransferase G9a. J Med Chem 52(24):7950-7953. doi:10.1021/Jm901543m
    • (2009) J Med Chem , vol.52 , Issue.24 , pp. 7950-7953
    • Liu, F.1    Chen, X.2    Allali-Hassani, A.3
  • 111
    • 77955363182 scopus 로고    scopus 로고
    • Protein lysine methyltransferase G9a inhibitors: Design, synthesis, and structure activity relationships of 2,4-diamino-7-aminoalkoxy-quinazolines
    • doi:10.1021/jm100478y
    • Liu F, Chen X, Allali-Hassani A et al (2010) Protein lysine methyltransferase G9a inhibitors: design, synthesis, and structure activity relationships of 2,4-diamino-7-aminoalkoxy-quinazolines. J Med Chem 53(15):5844-5857. doi:10.1021/jm100478y
    • (2010) J Med Chem , vol.53 , Issue.15 , pp. 5844-5857
    • Liu, F.1    Chen, X.2    Allali-Hassani, A.3
  • 112
    • 84887923349 scopus 로고    scopus 로고
    • Discovery of an in vivo chemical probe of the lysine methyltransferases G9a and GLP
    • doi:10.1021/jm401480r
    • Liu F, Barsyte-Lovejoy D, Li F et al (2013) Discovery of an in vivo chemical probe of the lysine methyltransferases G9a and GLP. J Med Chem 56(21):8931-8942. doi:10.1021/jm401480r
    • (2013) J Med Chem , vol.56 , Issue.21 , pp. 8931-8942
    • Liu, F.1    Barsyte-Lovejoy, D.2    Li, F.3
  • 113
    • 84886815629 scopus 로고    scopus 로고
    • Preclinical characterization of a potent and selective inhibitor of the histone demethylase KDM1A for ML leukemia
    • Maes T, Tirapu I, Mascaro C et al (2013) Preclinical characterization of a potent and selective inhibitor of the histone demethylase KDM1A for ML leukemia. J Clin Oncol 31(15):e13543
    • (2013) J Clin Oncol , vol.31 , Issue.15
    • Maes, T.1    Tirapu, I.2    Mascaro, C.3
  • 114
    • 34250316675 scopus 로고    scopus 로고
    • The therapeutic uses of chromatin-modifying agents
    • doi:10.1517/14728222.11.6.835
    • Mai A (2007) The therapeutic uses of chromatin-modifying agents. Expert Opin Ther Targets 11(6):835-851. doi:10.1517/14728222.11.6.835
    • (2007) Expert Opin Ther Targets , vol.11 , Issue.6 , pp. 835-851
    • Mai, A.1
  • 115
    • 56149090684 scopus 로고    scopus 로고
    • Epi-drugs to fight cancer: From chemistry to cancer treatment, the road ahead
    • doi:10.1016/j.biocel.2008.08.020
    • Mai A, Altucci L (2009) Epi-drugs to fight cancer: from chemistry to cancer treatment, the road ahead. Int J Biochem Cell Biol 41(1):199-213. doi:10.1016/j.biocel.2008.08.020
    • (2009) Int J Biochem Cell Biol , vol.41 , Issue.1 , pp. 199-213
    • Mai, A.1    Altucci, L.2
  • 118
    • 84857879142 scopus 로고    scopus 로고
    • Decitabine for acute myeloid leukemia
    • doi:10.1586/era.11.207
    • Marks PW (2012) Decitabine for acute myeloid leukemia. Expert Rev Anticancer Ther 12(3):299-305. doi:10.1586/era.11.207
    • (2012) Expert Rev Anticancer Ther , vol.12 , Issue.3 , pp. 299-305
    • Marks, P.W.1
  • 119
  • 120
    • 84870573126 scopus 로고    scopus 로고
    • EZH2 inhibition as a therapeutic strategy for lymphoma with EZH2-activating mutations
    • doi:10.1038/nature11606
    • McCabe MT, Ott HM, Ganji G et al (2012) EZH2 inhibition as a therapeutic strategy for lymphoma with EZH2-activating mutations. Nature 492(7427):108-112. doi:10.1038/nature11606
    • (2012) Nature , vol.492 , Issue.7427 , pp. 108-112
    • McCabe, M.T.1    Ott, H.M.2    Ganji, G.3
  • 121
  • 123
    • 84875217338 scopus 로고    scopus 로고
    • Anticancer efficacy of cisplatin and trichostatin A or 5-aza-2′-deoxycytidine on ovarian cancer
    • doi:10.1038/bjc.2013.10
    • Meng F, Sun G, Zhong M, Yu Y, Brewer MA (2013) Anticancer efficacy of cisplatin and trichostatin A or 5-aza-2′-deoxycytidine on ovarian cancer. Br J Cancer 108(3):579-586. doi:10.1038/bjc.2013.10
    • (2013) Br J Cancer , vol.108 , Issue.3 , pp. 579-586
    • Meng, F.1    Sun, G.2    Zhong, M.3    Yu, Y.4    Brewer, M.A.5
  • 124
    • 80053651202 scopus 로고    scopus 로고
    • Targeting MYC dependence in cancer by inhibiting BET bromodomains
    • doi:10.1073/pnas.1108190108
    • Mertz JA, Conery AR, Bryant BM et al (2011) Targeting MYC dependence in cancer by inhibiting BET bromodomains. Proc Natl Acad Sci USA 108(40):16669-16674. doi:10.1073/pnas.1108190108
    • (2011) Proc Natl Acad Sci USA , vol.108 , Issue.40 , pp. 16669-16674
    • Mertz, J.A.1    Conery, A.R.2    Bryant, B.M.3
  • 126
    • 84885675970 scopus 로고    scopus 로고
    • Discovery of epigenetic regulator I-BET762: Lead optimization to afford a clinical candidate inhibitor of the BET bromodomains
    • doi:10.1021/jm401088k
    • Mirguet O, Gosmini R, Toum J et al (2013) Discovery of epigenetic regulator I-BET762: lead optimization to afford a clinical candidate inhibitor of the BET bromodomains. J Med Chem 56(19):7501-7515. doi:10.1021/jm401088k
    • (2013) J Med Chem , vol.56 , Issue.19 , pp. 7501-7515
    • Mirguet, O.1    Gosmini, R.2    Toum, J.3
  • 127
    • 0034072130 scopus 로고    scopus 로고
    • DNA methylation and cancer
    • doi:10.1002/ (SICI)1097-4652(200005)183:2
    • Momparler RL, Bovenzi V (2000) DNA methylation and cancer. J Cell Physiol 183(2):145-154. doi:10.1002/ (SICI)1097-4652(200005)183:2〈145::AID- JCP1〉3.0.CO;2-V
    • (2000) J Cell Physiol , vol.183 , Issue.2 , pp. 145-154
    • Momparler, R.L.1    Bovenzi, V.2
  • 128
    • 84899117261 scopus 로고    scopus 로고
    • Epigenetics and oncology
    • doi:10.1002/phar.1408
    • Mummaneni P, Shord SS (2014) Epigenetics and oncology. Pharmacotherapy 34(5):495-505. doi:10.1002/phar.1408
    • (2014) Pharmacotherapy , vol.34 , Issue.5 , pp. 495-505
    • Mummaneni, P.1    Shord, S.S.2
  • 130
    • 4544236944 scopus 로고    scopus 로고
    • Generation of hydrogen peroxide primarily contributes to the induction of Fe(II)-dependent apoptosis in Jurkat cells by (-)-epigallocatechin gallate
    • DOI 10.1093/carcin/bgh168
    • Nakagawa H, Hasumi K, Woo JT, Nagai K, Wachi M (2004) Generation of hydrogen peroxide primarily contributes to the induction of Fe(II)-dependent apoptosis in Jurkat cells by (-)-epigallocatechin gallate. Carcinogenesis 25(9):1567-1574. doi:10.1093/carcin/bgh168 (Pubitemid 39214298)
    • (2004) Carcinogenesis , vol.25 , Issue.9 , pp. 1567-1574
    • Nakagawa, H.1    Hasumi, K.2    Woo, J.-T.3    Nagai, K.4    Wachi, M.5
  • 131
    • 84872183961 scopus 로고    scopus 로고
    • DNA methyltransferase inhibitor zebularine inhibits human hepatic carcinoma cells proliferation and induces apoptosis
    • doi:10.1371/journal.pone.0054036
    • Nakamura K, Aizawa K, Nakabayashi K et al (2013) DNA methyltransferase inhibitor zebularine inhibits human hepatic carcinoma cells proliferation and induces apoptosis. PLoS ONE 8(1):e54036. doi:10.1371/journal.pone.0054036
    • (2013) PLoS ONE , vol.8 , Issue.1
    • Nakamura, K.1    Aizawa, K.2    Nakabayashi, K.3
  • 133
    • 83355166904 scopus 로고    scopus 로고
    • Death receptor pathway activation and increase of ROS production by the triple epigenetic inhibitor UVI5008
    • doi:10.1158/1535-7163.MCT-11-0525
    • Nebbioso A, Pereira R, Khanwalkar H et al (2011) Death receptor pathway activation and increase of ROS production by the triple epigenetic inhibitor UVI5008. Mol Cancer Ther 10(12):2394-2404. doi:10.1158/1535-7163.MCT-11-0525
    • (2011) Mol Cancer Ther , vol.10 , Issue.12 , pp. 2394-2404
    • Nebbioso, A.1    Pereira, R.2    Khanwalkar, H.3
  • 134
    • 84869874482 scopus 로고    scopus 로고
    • Trials with 'epigenetic' drugs: An update
    • doi:10.1016/j.molonc.2012.09.004
    • Nebbioso A, Carafa V, Benedetti R, Altucci L (2012) Trials with 'epigenetic' drugs: an update. Mol Oncol 6(6):657-682. doi:10.1016/j.molonc. 2012.09.004
    • (2012) Mol Oncol , vol.6 , Issue.6 , pp. 657-682
    • Nebbioso, A.1    Carafa, V.2    Benedetti, R.3    Altucci, L.4
  • 135
    • 84866737098 scopus 로고    scopus 로고
    • Garcinol, a histone acetyltransferase inhibitor, radiosensitizes cancer cells by inhibiting non-homologous end joining
    • doi:10.1016/j.ijrobp.2012.01.017
    • Oike T, Ogiwara H, Torikai K, Nakano T, Yokota J, Kohno T (2012) Garcinol, a histone acetyltransferase inhibitor, radiosensitizes cancer cells by inhibiting non-homologous end joining. Int J Radiat Oncol Biol Phys 84(3):815-821. doi:10.1016/j.ijrobp.2012.01.017
    • (2012) Int J Radiat Oncol Biol Phys , vol.84 , Issue.3 , pp. 815-821
    • Oike, T.1    Ogiwara, H.2    Torikai, K.3    Nakano, T.4    Yokota, J.5    Kohno, T.6
  • 136
    • 0033615717 scopus 로고    scopus 로고
    • DNA methyltransferases Dnmt3a and Dnmt3b are essential for de novo methylation and mammalian development
    • DOI 10.1016/S0092-8674(00)81656-6
    • Okano M, Bell DW, Haber DA, Li E (1999) DNA methyltransferases Dnmt3a and Dnmt3b are essential for de novo methylation and mammalian development. Cell 99(3):247-257 (Pubitemid 29519904)
    • (1999) Cell , vol.99 , Issue.3 , pp. 247-257
    • Okano, M.1    Bell, D.W.2    Haber, D.A.3    Li, E.4
  • 137
    • 84890606231 scopus 로고    scopus 로고
    • Phase I study of panobinostat plus everolimus in patients with relapsed or refractory lymphoma
    • doi:10.1158/1078-0432.CR-13-1906
    • Oki Y, Buglio D, Fanale M et al (2013) Phase I study of panobinostat plus everolimus in patients with relapsed or refractory lymphoma. Clin Cancer Res 19(24):6882-6890. doi:10.1158/1078-0432.CR-13-1906
    • (2013) Clin Cancer Res , vol.19 , Issue.24 , pp. 6882-6890
    • Oki, Y.1    Buglio, D.2    Fanale, M.3
  • 140
    • 77950246109 scopus 로고    scopus 로고
    • SRT1720, SRT2183, SRT1460, and resveratrol are not direct activators of SIRT1
    • doi:10.1074/jbc.M109.088682
    • Pacholec M, Bleasdale JE, Chrunyk B et al (2010) SRT1720, SRT2183, SRT1460, and resveratrol are not direct activators of SIRT1. J Biol Chem 285(11):8340-8351. doi:10.1074/jbc.M109.088682
    • (2010) J Biol Chem , vol.285 , Issue.11 , pp. 8340-8351
    • Pacholec, M.1    Bleasdale, J.E.2    Chrunyk, B.3
  • 141
    • 84869889829 scopus 로고    scopus 로고
    • New approaches for cancer treatment: Antitumor drugs based on gene-targeted nucleic acids
    • Patutina OA, Mironova NL, Vlassov V, Zenkova MA (2009) New approaches for cancer treatment: antitumor drugs based on gene-targeted nucleic acids. Acta Nat 1(2):44-60
    • (2009) Acta Nat , vol.1 , Issue.2 , pp. 44-60
    • Patutina, O.A.1    Mironova, N.L.2    Vlassov, V.3    Zenkova, M.A.4
  • 142
    • 84858006066 scopus 로고    scopus 로고
    • Sensitization of tumor cells by targeting histone deacetylases
    • doi:10.1016/j.bcp.2011.11.010
    • Perego P, Zuco V, Gatti L, Zunino F (2012) Sensitization of tumor cells by targeting histone deacetylases. Biochem Pharmacol 83(8):987-994. doi:10.1016/j.bcp.2011.11.010
    • (2012) Biochem Pharmacol , vol.83 , Issue.8 , pp. 987-994
    • Perego, P.1    Zuco, V.2    Gatti, L.3    Zunino, F.4
  • 143
    • 84876794613 scopus 로고    scopus 로고
    • 5-hydroxymethylcytosine and its potential roles in development and cancer
    • doi:10.1186/1756-8935-6-10
    • Pfeifer GP, Kadam S, Jin SG (2013) 5-hydroxymethylcytosine and its potential roles in development and cancer. Epigenetics Chromatin 6(1):10. doi:10.1186/1756-8935-6-10
    • (2013) Epigenetics Chromatin , vol.6 , Issue.1 , pp. 10
    • Pfeifer, G.P.1    Kadam, S.2    Jin, S.G.3
  • 144
    • 84855340600 scopus 로고    scopus 로고
    • Phase I study of the histone deacetylase inhibitor entinostat in combination with 13-cis retinoic acid in patients with solid tumours
    • doi:10.1038/bjc.2011.527
    • Pili R, Salumbides B, Zhao M et al (2012) Phase I study of the histone deacetylase inhibitor entinostat in combination with 13-cis retinoic acid in patients with solid tumours. Br J Cancer 106(1):77-84. doi:10.1038/bjc.2011.527
    • (2012) Br J Cancer , vol.106 , Issue.1 , pp. 77-84
    • Pili, R.1    Salumbides, B.2    Zhao, M.3
  • 146
    • 33749006252 scopus 로고    scopus 로고
    • Class II histone deacetylases are associated with VHL-independent regulation of hypoxia-inducible factor 1alpha
    • DOI 10.1158/0008-5472.CAN-05-4598
    • Qian DZ, Kachhap SK, Collis SJ et al (2006) Class II histone deacetylases are associated with VHL-independent regulation of hypoxia-inducible factor 1 alpha. Cancer Res 66(17):8814-8821. doi:10.1158/0008-5472.CAN-05-4598 (Pubitemid 44449199)
    • (2006) Cancer Research , vol.66 , Issue.17 , pp. 8814-8821
    • Qian, D.Z.1    Kachhap, S.K.2    Collis, S.J.3    Verheul, H.M.W.4    Carducci, M.A.5    Atadja, P.6    Pili, R.7
  • 148
    • 84855283939 scopus 로고    scopus 로고
    • The histone deacetylase inhibitor trichostatin A alters microRNA expression profiles in apoptosis-resistant breast cancer cells
    • doi:10.3892/or.2011.1488
    • Rhodes LV, Nitschke AM, Segar HC et al (2012) The histone deacetylase inhibitor trichostatin A alters microRNA expression profiles in apoptosis-resistant breast cancer cells. Oncol Rep 27(1):10-16. doi:10.3892/or.2011.1488
    • (2012) Oncol Rep , vol.27 , Issue.1 , pp. 10-16
    • Rhodes, L.V.1    Nitschke, A.M.2    Segar, H.C.3
  • 149
    • 44949163006 scopus 로고    scopus 로고
    • A new path to the cancer epigenome
    • doi:10.1038/nbt0608-655
    • Richon VM (2008) A new path to the cancer epigenome. Nat Biotechnol 26(6):655-656. doi:10.1038/nbt0608-655
    • (2008) Nat Biotechnol , vol.26 , Issue.6 , pp. 655-656
    • Richon, V.M.1
  • 151
    • 0035962631 scopus 로고    scopus 로고
    • DNA methylation, methyltransferases, and cancer
    • doi:10.1038/sj.onc.1204341
    • Robertson KD (2001) DNA methylation, methyltransferases, and cancer. Oncogene 20(24):3139-3155. doi:10.1038/sj.onc.1204341
    • (2001) Oncogene , vol.20 , Issue.24 , pp. 3139-3155
    • Robertson, K.D.1
  • 152
    • 0033153303 scopus 로고    scopus 로고
    • The human DNA methyltransferases (DNMTs) 1, 3a and 3b: Coordinate mRNA expression in normal tissues and overexpression in tumors
    • DOI 10.1093/nar/27.11.2291
    • Robertson KD, Uzvolgyi E, Liang G et al (1999) The human DNA methyltransferases (DNMTs) 1, 3a and 3b: coordinate mRNA expression in normal tissues and overexpression in tumors. Nucleic Acids Res 27(11):2291-2298 (Pubitemid 29245646)
    • (1999) Nucleic Acids Research , vol.27 , Issue.11 , pp. 2291-2298
    • Robertson, K.D.1    Uzvolgyi, E.2    Liang, G.3    Talmadge, C.4    Sumegi, J.5    Gonzales, F.A.6    Jones, P.A.7
  • 153
    • 49749083488 scopus 로고    scopus 로고
    • Resveratrol: A natural compound with pharmacological potential in neurodegenerative diseases
    • doi:10.1111/j.1755-5949.2008.00045.x
    • Rocha-Gonzalez HI, Ambriz-Tututi M, Granados-Soto V (2008) Resveratrol: a natural compound with pharmacological potential in neurodegenerative diseases. CNS Neurosci Ther 14(3):234-247. doi:10.1111/j.1755-5949.2008.00045.x
    • (2008) CNS Neurosci Ther , vol.14 , Issue.3 , pp. 234-247
    • Rocha-Gonzalez, H.I.1    Ambriz-Tututi, M.2    Granados-Soto, V.3
  • 154
    • 79952384705 scopus 로고    scopus 로고
    • Cancer epigenetics reaches mainstream oncology
    • doi:10.1038/nm.2305
    • Rodriguez-Paredes M, Esteller M (2011) Cancer epigenetics reaches mainstream oncology. Nat Med 17(3):330-339. doi:10.1038/nm.2305
    • (2011) Nat Med , vol.17 , Issue.3 , pp. 330-339
    • Rodriguez-Paredes, M.1    Esteller, M.2
  • 155
    • 84857232181 scopus 로고    scopus 로고
    • LBH-589 (panobinostat) potentiates fludarabine anti-leukemic activity through a JNK- and XIAP-dependent mechanism
    • doi:10.1016/j.leukres.2011.10.020
    • Rosato R, Hock S, Dent P, Dai Y, Grant S (2012) LBH-589 (panobinostat) potentiates fludarabine anti-leukemic activity through a JNK- and XIAP-dependent mechanism. Leuk Res 36(4):491-498. doi:10.1016/j.leukres.2011.10.020
    • (2012) Leuk Res , vol.36 , Issue.4 , pp. 491-498
    • Rosato, R.1    Hock, S.2    Dent, P.3    Dai, Y.4    Grant, S.5
  • 156
    • 36448949026 scopus 로고    scopus 로고
    • Multivalent engagement of chromatin modifications by linked binding modules
    • DOI 10.1038/nrm2298, PII NRM2298
    • Ruthenburg AJ, Li H, Patel DJ, Allis CD (2007) Multivalent engagement of chromatin modifications by linked binding modules. Nat Rev Mol Cell Biol 8(12):983-994. doi:10.1038/nrm2298 (Pubitemid 350174643)
    • (2007) Nature Reviews Molecular Cell Biology , vol.8 , Issue.12 , pp. 983-994
    • Ruthenburg, A.J.1    Li, H.2    Patel, D.J.3    David, A.C.4
  • 158
    • 2542421889 scopus 로고    scopus 로고
    • Bisubstrate analogue structure-activity relationships for p300 histone acetyltransferase inhibitors
    • DOI 10.1016/j.bmc.2004.03.070, PII S0968089604002627
    • Sagar V, Zheng W, Thompson PR, Cole PA (2004) Bisubstrate analogue structure-activity relationships for p300 histone acetyltransferase inhibitors. Bioorg Med Chem 12(12):3383-3390. doi:10.1016/j.bmc.2004.03.070 (Pubitemid 38680727)
    • (2004) Bioorganic and Medicinal Chemistry , vol.12 , Issue.12 , pp. 3383-3390
    • Sagar, V.1    Zheng, W.2    Thompson, P.R.3    Cole, P.A.4
  • 159
    • 84893834544 scopus 로고    scopus 로고
    • Inhibition of lysine-specific demethylase 1 by the acyclic diterpenoid geranylgeranoic acid and its derivatives
    • doi:10.1016/j.bbrc.2013.12.144
    • Sakane C, Okitsu T, Wada A, Sagami H, Shidoji Y (2014) Inhibition of lysine-specific demethylase 1 by the acyclic diterpenoid geranylgeranoic acid and its derivatives. Biochem Biophys Res Commun 444(1):24-29. doi:10.1016/j.bbrc.2013.12.144
    • (2014) Biochem Biophys Res Commun , vol.444 , Issue.1 , pp. 24-29
    • Sakane, C.1    Okitsu, T.2    Wada, A.3    Sagami, H.4    Shidoji, Y.5
  • 160
    • 84875271390 scopus 로고    scopus 로고
    • DNA methyltransferase inhibitor RG108 and histone deacetylase inhibitors cooperate to enhance NB4 cell differentiation and E-cadherin re-expression by chromatin remodelling
    • doi:10.1042/CBI20110649
    • Savickiene J, Treigyte G, Jazdauskaite A, Borutinskaite V, Navakauskiene R (2012) DNA methyltransferase inhibitor RG108 and histone deacetylase inhibitors cooperate to enhance NB4 cell differentiation and E-cadherin re-expression by chromatin remodelling. Cell Biol Int 36(11):1067-1078. doi:10.1042/CBI20110649
    • (2012) Cell Biol Int , vol.36 , Issue.11 , pp. 1067-1078
    • Savickiene, J.1    Treigyte, G.2    Jazdauskaite, A.3    Borutinskaite, V.4    Navakauskiene, R.5
  • 161
    • 74049130225 scopus 로고    scopus 로고
    • Solving the Dnmt2 enigma
    • doi:10.1007/s00412-009-0240-6
    • Schaefer M, Lyko F (2010) Solving the Dnmt2 enigma. Chromosoma 119(1):35-40. doi:10.1007/s00412-009-0240-6
    • (2010) Chromosoma , vol.119 , Issue.1 , pp. 35-40
    • Schaefer, M.1    Lyko, F.2
  • 162
    • 77955884641 scopus 로고    scopus 로고
    • RNA methylation by Dnmt2 protects transfer RNAs against stress-induced cleavage
    • doi: 10.1101/gad.586710
    • Schaefer M, Pollex T, Hanna K, Tuorto F, Meusburger M, Helm M, Lyko F (2010) RNA methylation by Dnmt2 protects transfer RNAs against stress-induced cleavage. Genes Dev 24(15):1590-1595. doi: 10.1101/gad.586710
    • (2010) Genes Dev , vol.24 , Issue.15 , pp. 1590-1595
    • Schaefer, M.1    Pollex, T.2    Hanna, K.3    Tuorto, F.4    Meusburger, M.5    Helm, M.6    Lyko, F.7
  • 165
    • 78751706756 scopus 로고    scopus 로고
    • Resveratrol and cellular mechanisms of cancer prevention
    • doi:10.1111/j.1749-6632.2010.05870.x
    • Shukla Y, Singh R (2011) Resveratrol and cellular mechanisms of cancer prevention. Ann N Y Acad Sci 1215:1-8. doi:10.1111/j.1749-6632.2010.05870.x
    • (2011) Ann N Y Acad Sci , vol.1215 , pp. 1-8
    • Shukla, Y.1    Singh, R.2
  • 166
    • 79957993618 scopus 로고    scopus 로고
    • The class I HDAC inhibitor MGCD0103 induces cell cycle arrest and apoptosis in colon cancer initiating cells by upregulating Dickkopf-1 and non-canonical Wnt signaling
    • Sikandar S, Dizon D, Shen X, Li Z, Besterman J, Lipkin SM (2010) The class I HDAC inhibitor MGCD0103 induces cell cycle arrest and apoptosis in colon cancer initiating cells by upregulating Dickkopf-1 and non-canonical Wnt signaling. Oncotarget 1(7):596-605
    • (2010) Oncotarget , vol.1 , Issue.7 , pp. 596-605
    • Sikandar, S.1    Dizon, D.2    Shen, X.3    Li, Z.4    Besterman, J.5    Lipkin, S.M.6
  • 167
    • 84865466261 scopus 로고    scopus 로고
    • Histone deacetylase inhibitors: Structure-based modeling and isoformselectivity prediction
    • doi:10.1021/ci300160y
    • Silvestri L, Ballante F, Mai A, Marshall GR, Ragno R (2012) Histone deacetylase inhibitors: structure-based modeling and isoformselectivity prediction. J Chem Inf Model 52(8):2215-2235. doi:10.1021/ci300160y
    • (2012) J Chem Inf Model , vol.52 , Issue.8 , pp. 2215-2235
    • Silvestri, L.1    Ballante, F.2    Mai, A.3    Marshall, G.R.4    Ragno, R.5
  • 169
    • 59349115177 scopus 로고    scopus 로고
    • Chemical mechanisms of histone lysine and arginine modifications
    • doi:10.1016/j.bbagrm.2008.06.005
    • Smith BC, Denu JM (2009) Chemical mechanisms of histone lysine and arginine modifications. Biochim Biophys Acta 1789(1):45- 57. doi:10.1016/j.bbagrm.2008.06.005
    • (2009) Biochim Biophys Acta , vol.1789 , Issue.1 , pp. 45-57
    • Smith, B.C.1    Denu, J.M.2
  • 170
    • 4644265735 scopus 로고    scopus 로고
    • Low-dose suramin enhanced paclitaxel activity in chemotherapy-naive and paclitaxel-pretreated human breast xenograft tumors
    • DOI 10.1158/1078-0432.CCR-04-0595
    • Song S, Yu B, Wei Y, Wientjes MG, Au JL (2004) Low-dose suramin enhanced paclitaxel activity in chemotherapy-naive and paclitaxel- pretreated human breast xenograft tumors. Clin Cancer Res 10(18 Pt 1):6058-6065. doi:10.1158/1078-0432.CR-04-0595 (Pubitemid 39287510)
    • (2004) Clinical Cancer Research , vol.10 , Issue.18 I , pp. 6058-6065
    • Song, S.1    Yu, B.2    Wei, Y.3    Wientjes, M.G.4    Au, J.L.-S.5
  • 171
    • 70349780606 scopus 로고    scopus 로고
    • The emerging therapeutic potential of histone methyltransferase and demethylase inhibitors
    • doi:10.1002/cmdc.200900301
    • Spannhoff A, Hauser AT, Heinke R, Sippl W, Jung M (2009) The emerging therapeutic potential of histone methyltransferase and demethylase inhibitors. ChemMedChem 4(10):1568-1582. doi:10.1002/cmdc.200900301
    • (2009) ChemMedChem , vol.4 , Issue.10 , pp. 1568-1582
    • Spannhoff, A.1    Hauser, A.T.2    Heinke, R.3    Sippl, W.4    Jung, M.5
  • 172
    • 84897550318 scopus 로고    scopus 로고
    • shRNA screening identifies JMJD1C as being required for leukemia maintenance
    • doi:10.1182/ blood-2013-08-522094
    • Sroczynska P, Cruickshank VA, Bukowski JP et al (2014) shRNA screening identifies JMJD1C as being required for leukemia maintenance. Blood 123(12):1870-1882. doi:10.1182/ blood-2013-08-522094
    • (2014) Blood , vol.123 , Issue.12 , pp. 1870-1882
    • Sroczynska, P.1    Cruickshank, V.A.2    Bukowski, J.P.3
  • 175
    • 84862878080 scopus 로고    scopus 로고
    • Design, synthesis, and biological evaluation of 2-aminobenzanilide derivatives as potent and selective HDAC inhibitors
    • doi:10.1002/cmdc.201200193
    • Stolfa DA, Stefanachi A, Gajer JM et al (2012) Design, synthesis, and biological evaluation of 2-aminobenzanilide derivatives as potent and selective HDAC inhibitors. ChemMedChem 7(7):1256-1266. doi:10.1002/cmdc.201200193
    • (2012) ChemMedChem , vol.7 , Issue.7 , pp. 1256-1266
    • Stolfa, D.A.1    Stefanachi, A.2    Gajer, J.M.3
  • 176
    • 0036613250 scopus 로고    scopus 로고
    • A genomic screen for genes upregulated by demethylation and histone deacetylase inhibition in human colorectal cancer
    • doi:10.1038/ng892
    • Suzuki H, Gabrielson E, Chen W et al (2002) A genomic screen for genes upregulated by demethylation and histone deacetylase inhibition in human colorectal cancer. Nat Genet 31(2):141-149. doi:10.1038/ng892
    • (2002) Nat Genet , vol.31 , Issue.2 , pp. 141-149
    • Suzuki, H.1    Gabrielson, E.2    Chen, W.3
  • 177
    • 84894065529 scopus 로고    scopus 로고
    • Discovery and development of potent and selective inhibitors of histone methyltransferase g9a
    • doi:10.1021/ml400496h
    • Sweis RF, Pliushchev M, Brown PJ et al (2014) Discovery and development of potent and selective inhibitors of histone methyltransferase g9a. ACS Med Chem Lett 5(2):205-209. doi:10.1021/ml400496h
    • (2014) ACS Med Chem Lett , vol.5 , Issue.2 , pp. 205-209
    • Sweis, R.F.1    Pliushchev, M.2    Brown, P.J.3
  • 179
    • 35848961668 scopus 로고    scopus 로고
    • How chromatin-binding modules interpret histone modifications: Lessons from professional pocket pickers
    • DOI 10.1038/nsmb1338, PII NSMB1338
    • Taverna SD, Li H, Ruthenburg AJ, Allis CD, Patel DJ (2007) How chromatin-binding modules interpret histone modifications: lessons from professional pocket pickers. Nat Struct Mol Biol 14(11):1025-1040. doi:10.1038/nsmb1338 (Pubitemid 350060344)
    • (2007) Nature Structural and Molecular Biology , vol.14 , Issue.11 , pp. 1025-1040
    • Taverna, S.D.1    Li, H.2    Ruthenburg, A.J.3    Allis, C.D.4    Patel, D.J.5
  • 181
    • 84863328591 scopus 로고    scopus 로고
    • The journey of resveratrol from yeast to human
    • Timmers S, Auwerx J, Schrauwen P (2012) The journey of resveratrol from yeast to human. Aging 4(3):146-158
    • (2012) Aging , vol.4 , Issue.3 , pp. 146-158
    • Timmers, S.1    Auwerx, J.2    Schrauwen, P.3
  • 183
    • 72249117352 scopus 로고    scopus 로고
    • Identification of cell-active lysine specific demethylase 1-selective inhibitors
    • doi:10.1021/ja907055q
    • Ueda R, Suzuki T, Mino K et al (2009) Identification of cell-active lysine specific demethylase 1-selective inhibitors. J Am Chem Soc 131(48):17536-17537. doi:10.1021/ja907055q
    • (2009) J Am Chem Soc , vol.131 , Issue.48 , pp. 17536-17537
    • Ueda, R.1    Suzuki, T.2    Mino, K.3
  • 184
    • 67349203626 scopus 로고    scopus 로고
    • Somatic mutations of the histone H3K27 demethylase gene UTX in human cancer
    • doi:10.1038/ng.349
    • van Haaften G, Dalgliesh GL, Davies H et al (2009) Somatic mutations of the histone H3K27 demethylase gene UTX in human cancer. Nat Genet 41(5):521-523. doi:10.1038/ng.349
    • (2009) Nat Genet , vol.41 , Issue.5 , pp. 521-523
    • Van Haaften, G.1    Dalgliesh, G.L.2    Davies, H.3
  • 186
    • 84867702707 scopus 로고    scopus 로고
    • Sirtuin activators and inhibitors
    • doi:10.1002/biof.1032
    • Villalba JM, Alcain FJ (2012) Sirtuin activators and inhibitors. Bio- Factors 38(5):349-359. doi:10.1002/biof.1032
    • (2012) Bio- Factors , vol.38 , Issue.5 , pp. 349-359
    • Villalba, J.M.1    Alcain, F.J.2
  • 187
    • 0037303184 scopus 로고    scopus 로고
    • DNA demethylating agents and chromatin-remodelling drugs: Which, how and why?
    • Villar-Garea A, Esteller M (2003) DNA demethylating agents and chromatin-remodelling drugs: which, how and why? Curr Drug Metab 4(1):11-31
    • (2003) Curr Drug Metab , vol.4 , Issue.1 , pp. 11-31
    • Villar-Garea, A.1    Esteller, M.2
  • 188
    • 79960255863 scopus 로고    scopus 로고
    • Recurrent DNMT3A mutations in patients with myelodysplastic syndromes
    • doi:10.1038/leu.2011.44
    • Walter MJ, Ding L, Shen D et al (2011) Recurrent DNMT3A mutations in patients with myelodysplastic syndromes. Leukemia 25(7):1153-1158. doi:10.1038/leu.2011.44
    • (2011) Leukemia , vol.25 , Issue.7 , pp. 1153-1158
    • Walter, M.J.1    Ding, L.2    Shen, D.3
  • 189
    • 84862509994 scopus 로고    scopus 로고
    • Resveratrol in cardiovascular disease: What is known from current research?
    • doi:10.1007/s10741-011-9260-4
    • Wang H, Yang YJ, Qian HY, Zhang Q, Xu H, Li JJ (2012) Resveratrol in cardiovascular disease: what is known from current research? Heart Fail Rev 17(3):437-448. doi:10.1007/s10741-011-9260-4
    • (2012) Heart Fail Rev , vol.17 , Issue.3 , pp. 437-448
    • Wang, H.1    Yang, Y.J.2    Qian, H.Y.3    Zhang, Q.4    Xu, H.5    Li, J.J.6
  • 190
    • 33646124469 scopus 로고    scopus 로고
    • Reversal of histone lysine trimethylation by the JMJD2 family of histone demethylases
    • doi:10.1016/j.cell.2006.03.028
    • Whetstine JR, Nottke A, Lan F et al (2006) Reversal of histone lysine trimethylation by the JMJD2 family of histone demethylases. Cell 125(3):467-481. doi:10.1016/j.cell.2006.03.028
    • (2006) Cell , vol.125 , Issue.3 , pp. 467-481
    • Whetstine, J.R.1    Nottke, A.2    Lan, F.3
  • 191
    • 84903748276 scopus 로고    scopus 로고
    • Cytostatic and apoptotic effects of DNMT and HDAC inhibitors in endometrial cancer cells
    • Xu S, Ren J, Chen HB et al (2014) Cytostatic and apoptotic effects of DNMT and HDAC inhibitors in endometrial cancer cells. Curr Pharm Des 20(11):1881-1887
    • (2014) Curr Pharm des , vol.20 , Issue.11 , pp. 1881-1887
    • Xu, S.1    Ren, J.2    Chen, H.B.3
  • 192
    • 84896716141 scopus 로고    scopus 로고
    • Resveratrol inhibits the growth of gastric cancer by inducing G1 phase arrest and senescence in a Sirt1-dependent manner
    • doi:10.1371/journal.pone.0070627
    • Yang Q, Wang B, Zang W et al (2013) Resveratrol inhibits the growth of gastric cancer by inducing G1 phase arrest and senescence in a Sirt1-dependent manner. PLoS ONE 8(11):e70627. doi:10.1371/journal.pone.0070627
    • (2013) PLoS ONE , vol.8 , Issue.11
    • Yang, Q.1    Wang, B.2    Zang, W.3
  • 193
    • 80055000824 scopus 로고    scopus 로고
    • RNAi screen identifies Brd4 as a therapeutic target in acute myeloid leukaemia
    • doi:10.1038/nature10334
    • Zuber J, Shi J, Wang E et al (2011) RNAi screen identifies Brd4 as a therapeutic target in acute myeloid leukaemia. Nature 478(7370):524-528. doi:10.1038/nature10334
    • (2011) Nature , vol.478 , Issue.7370 , pp. 524-528
    • Zuber, J.1    Shi, J.2    Wang, E.3


* 이 정보는 Elsevier사의 SCOPUS DB에서 KISTI가 분석하여 추출한 것입니다.