메뉴 건너뛰기




Volumn 68, Issue 7, 2014, Pages 802-811

Epigenetic therapies - A new direction in clinical medicine

Author keywords

[No Author keywords available]

Indexed keywords

5 AZA 2' DEOXYCYTIDINE; ASVASIRAN; AZACITIDINE; BEVASIRANIB; EPZ 005687; FR 235222; GSK 1210151A; GSK 126; HISTONE DEACETYLASE INHIBITOR; MIRAVIRSEN; MOCETINOSTAT; MS 147; MS 417; PROTEIN INHIBITOR; ROMIDEPSIN; RVX 208; SORAFENIB; UNCLASSIFIED DRUG; VORINOSTAT;

EID: 84902683042     PISSN: 13685031     EISSN: 17421241     Source Type: Journal    
DOI: 10.1111/ijcp.12436     Document Type: Review
Times cited : (9)

References (169)
  • 1
    • 80054720150 scopus 로고    scopus 로고
    • The biology of HDAC in cancer: The nuclear and epigenetic components
    • Hagelkruys A, Sawicka A, Rennmayr M, Seiser C,. The biology of HDAC in cancer: the nuclear and epigenetic components. Handb Exp Pharmacol 2011; 206: 13-37.
    • (2011) Handb Exp Pharmacol , vol.206 , pp. 13-37
    • Hagelkruys, A.1    Sawicka, A.2    Rennmayr, M.3    Seiser, C.4
  • 2
    • 84863770814 scopus 로고    scopus 로고
    • Cancer genetics and epigenetics: Two sides of the same coin?
    • You JS, Jones PA,. Cancer genetics and epigenetics: two sides of the same coin? Cancer Cell 2012; 22 (1): 9-20.
    • (2012) Cancer Cell , vol.22 , Issue.1 , pp. 9-20
    • You, J.S.1    Jones, P.A.2
  • 3
    • 84877930649 scopus 로고    scopus 로고
    • Targeting histone modifications-epigenetics in cancer
    • Waldmann T, Schneider R,. Targeting histone modifications-epigenetics in cancer. Curr Opin Cell Biol 2013; 25 (2): 184-9.
    • (2013) Curr Opin Cell Biol , vol.25 , Issue.2 , pp. 184-189
    • Waldmann, T.1    Schneider, R.2
  • 4
    • 53049089942 scopus 로고    scopus 로고
    • Cancer is a preventable disease that requires major lifestyle changes
    • Anand P, Kunnumakkara AB, Sundaram C, et al. Cancer is a preventable disease that requires major lifestyle changes. Pharm Res 2008; 25 (9): 2097-116.
    • (2008) Pharm Res , vol.25 , Issue.9 , pp. 2097-2116
    • Anand, P.1    Kunnumakkara, A.B.2    Sundaram, C.3
  • 5
    • 34247540446 scopus 로고    scopus 로고
    • Epigenetics and microRNAs
    • Chuang JC, Jones PA,. Epigenetics and microRNAs. Pediatr Res 2007; 61 (5 Pt 2): 24R-9R.
    • (2007) Pediatr Res , vol.61 , Issue.5 PART 2
    • Chuang, J.C.1    Jones, P.A.2
  • 6
    • 48449090298 scopus 로고    scopus 로고
    • Epigenetic biomarkers for human cancer: The time is now
    • Mulero-Navarro S, Esteller M,. Epigenetic biomarkers for human cancer: the time is now. Crit Rev Oncol Hematol 2008; 68 (1): 1-11.
    • (2008) Crit Rev Oncol Hematol , vol.68 , Issue.1 , pp. 1-11
    • Mulero-Navarro, S.1    Esteller, M.2
  • 7
    • 79953108690 scopus 로고    scopus 로고
    • Epigenetics in cancer: What's the future?
    • Boumber Y, Issa JP,. Epigenetics in cancer: what's the future? Oncology (Williston Park) 2011; 25 (3): 220-6.
    • (2011) Oncology (Williston Park) , vol.25 , Issue.3 , pp. 220-226
    • Boumber, Y.1    Issa, J.P.2
  • 8
    • 77958150580 scopus 로고    scopus 로고
    • Targeting DNA methylation for epigenetic therapy
    • Yang X, Lay F, Han H, Jones PA,. Targeting DNA methylation for epigenetic therapy. Trends Pharmacol Sci 2010; 31 (11): 536-46.
    • (2010) Trends Pharmacol Sci , vol.31 , Issue.11 , pp. 536-546
    • Yang, X.1    Lay, F.2    Han, H.3    Jones, P.A.4
  • 9
    • 84862681781 scopus 로고    scopus 로고
    • Epigenetic drug discovery: Targeting DNA methyltransferases
    • Foulks JM, Parnell KM, Nix RN, et al. Epigenetic drug discovery: targeting DNA methyltransferases. J Biomol Screen 2012; 17 (1): 2.
    • (2012) J Biomol Screen , vol.17 , Issue.1 , pp. 2
    • Foulks, J.M.1    Parnell, K.M.2    Nix, R.N.3
  • 10
    • 0020699979 scopus 로고
    • Hypomethylation distinguishes genes of some human cancers from their normal counterparts
    • Feinberg AP, Vogelstein B,. Hypomethylation distinguishes genes of some human cancers from their normal counterparts. Nature 1983; 301 (5895): 89-92.
    • (1983) Nature , vol.301 , Issue.5895 , pp. 89-92
    • Feinberg, A.P.1    Vogelstein, B.2
  • 11
    • 0033960560 scopus 로고    scopus 로고
    • Single-site methylation within the p53 promoter region reduces gene expression in a reporter gene construct: Possible in vivo relevance during tumorigenesis
    • Pogribny IP, Pogribna M, Christman JK, James SJ,. Single-site methylation within the p53 promoter region reduces gene expression in a reporter gene construct: possible in vivo relevance during tumorigenesis. Cancer Res 2000; 60 (3): 588-94.
    • (2000) Cancer Res , vol.60 , Issue.3 , pp. 588-594
    • Pogribny, I.P.1    Pogribna, M.2    Christman, J.K.3    James, S.J.4
  • 12
    • 79551712007 scopus 로고    scopus 로고
    • The role of epigenetic transcription repression and DNA methyltransferases in cancer
    • Daniel FI, Cherubini K, Yurgel LS, et al. The role of epigenetic transcription repression and DNA methyltransferases in cancer. Cancer 2011; 117 (4): 677-87.
    • (2011) Cancer , vol.117 , Issue.4 , pp. 677-687
    • Daniel, F.I.1    Cherubini, K.2    Yurgel, L.S.3
  • 13
    • 0022540321 scopus 로고
    • CpG-rich islands and the function of DNA methylation
    • Bird AP,. CpG-rich islands and the function of DNA methylation. Nature 1986; 321: 209-13.
    • (1986) Nature , vol.321 , pp. 209-213
    • Bird, A.P.1
  • 14
    • 35948981875 scopus 로고    scopus 로고
    • Genome-wide profiling of DNA methylation reveals a class of normally methylated CpG island promoters
    • Shen L, Kondo Y, Guo Y, et al. Genome-wide profiling of DNA methylation reveals a class of normally methylated CpG island promoters. PLoS Genet 2007; 3 (10): 2023-36.
    • (2007) PLoS Genet , vol.3 , Issue.10 , pp. 2023-2036
    • Shen, L.1    Kondo, Y.2    Guo, Y.3
  • 15
    • 18944383889 scopus 로고    scopus 로고
    • 5-Aza-deoxycytidine induces selective degradation of DNA methyltransferase 1 by a proteasomal pathway that requires the KEN box, bromo-adjacent homology domain, and nuclear localization signal
    • Ghoshal K, Datta J, Majumder S, et al. 5-Aza-deoxycytidine induces selective degradation of DNA methyltransferase 1 by a proteasomal pathway that requires the KEN box, bromo-adjacent homology domain, and nuclear localization signal. Mol Cell Biol 2005; 25 (11): 4727-41.
    • (2005) Mol Cell Biol , vol.25 , Issue.11 , pp. 4727-4741
    • Ghoshal, K.1    Datta, J.2    Majumder, S.3
  • 16
    • 77956415182 scopus 로고    scopus 로고
    • HOXB13, a target of DNMT3B, is methylated at an upstream CpG island, and functions as a tumor suppressor in primary colorectal tumors
    • Ghoshal K, Motiwala T, Claus R, et al. HOXB13, a target of DNMT3B, is methylated at an upstream CpG island, and functions as a tumor suppressor in primary colorectal tumors. PLoS ONE 2010; 5 (4): e10338.
    • (2010) PLoS ONE , vol.5 , Issue.4
    • Ghoshal, K.1    Motiwala, T.2    Claus, R.3
  • 17
    • 79960927422 scopus 로고    scopus 로고
    • Increased methylation variation in epigenetic domains across cancer types
    • Hansen KD, Timp W, Bravo HC, et al. Increased methylation variation in epigenetic domains across cancer types. Nat Genet 2011; 43 (8): 768-75.
    • (2011) Nat Genet , vol.43 , Issue.8 , pp. 768-775
    • Hansen, K.D.1    Timp, W.2    Bravo, H.C.3
  • 18
    • 70649095120 scopus 로고    scopus 로고
    • Differential methylation of tissue- and cancer-specific CpG island shores distinguishes human induced pluripotent stem cells, embryonic stem cells and fibroblasts
    • Doi A, Park IH, Wen B, et al. Differential methylation of tissue- and cancer-specific CpG island shores distinguishes human induced pluripotent stem cells, embryonic stem cells and fibroblasts. Nat Genet 2009; 41: 1350-3.
    • (2009) Nat Genet , vol.41 , pp. 1350-1353
    • Doi, A.1    Park, I.H.2    Wen, B.3
  • 19
    • 59149084538 scopus 로고    scopus 로고
    • The human colon cancer methylome shows similar hypo- and hypermethylation at conserved tissue-specific CpG island shores
    • Irizarry RA, Ladd-Acosta C, Wen B, et al. The human colon cancer methylome shows similar hypo- and hypermethylation at conserved tissue-specific CpG island shores. Nat Genet 2009; 41 (2): 178-86.
    • (2009) Nat Genet , vol.41 , Issue.2 , pp. 178-186
    • Irizarry, R.A.1    Ladd-Acosta, C.2    Wen, B.3
  • 20
    • 79960556965 scopus 로고    scopus 로고
    • Epigenome-wide association studies for common human diseases
    • Rakyan VK, Down TA, Balding DJ, Beck S,. Epigenome-wide association studies for common human diseases. Nat Rev Genet 2011; 12 (8): 529-41.
    • (2011) Nat Rev Genet , vol.12 , Issue.8 , pp. 529-541
    • Rakyan, V.K.1    Down, T.A.2    Balding, D.J.3    Beck, S.4
  • 21
    • 84881086323 scopus 로고    scopus 로고
    • Cancer as a dysregulated epigenome allowing cellular growth advantage at the expense of the host
    • Timp W, Feinberg AP,. Cancer as a dysregulated epigenome allowing cellular growth advantage at the expense of the host. Nat Rev Cancer 2013; 13 (7): 497-510.
    • (2013) Nat Rev Cancer , vol.13 , Issue.7 , pp. 497-510
    • Timp, W.1    Feinberg, A.P.2
  • 22
    • 34547799171 scopus 로고    scopus 로고
    • DNA methyltransferases as targets for cancer therapy
    • Ghoshal K, Bai S,. DNA methyltransferases as targets for cancer therapy. Drugs Today (Barc) 2007; 43 (6): 395-422.
    • (2007) Drugs Today (Barc) , vol.43 , Issue.6 , pp. 395-422
    • Ghoshal, K.1    Bai, S.2
  • 23
    • 33646071894 scopus 로고    scopus 로고
    • Decitabine improves patient outcomes in myelodysplastic syndromes: Results of a Phase III randomized study
    • Kantarjian H, Issa JP, Rosenfeld CS, Bennett JM, Albitar M,. Decitabine improves patient outcomes in myelodysplastic syndromes: results of a Phase III randomized study. Cancer 2006; 106: 1794-803.
    • (2006) Cancer , vol.106 , pp. 1794-1803
    • Kantarjian, H.1    Issa, J.P.2    Rosenfeld, C.S.3    Bennett, J.M.4    Albitar, M.5
  • 26
    • 16244400457 scopus 로고    scopus 로고
    • FDA drug approval summary: Azacitidine (5-azacytidine, Vidaza) for injectable suspension
    • Kaminskas E, Farrell AT, Wang YC, Sridhara R, Pazdur R,. FDA drug approval summary: azacitidine (5-azacytidine, Vidaza) for injectable suspension. Oncologist 2005; 10 (3): 176-82.
    • (2005) Oncologist , vol.10 , Issue.3 , pp. 176-182
    • Kaminskas, E.1    Farrell, A.T.2    Wang, Y.C.3    Sridhara, R.4    Pazdur, R.5
  • 27
    • 74849127193 scopus 로고    scopus 로고
    • Epigenetic therapy: Histone acetylation, DNA methylation and anti-cancer drug discovery
    • Ganesan A, Nolan L, Crabb SJ, Packham G,. Epigenetic therapy: histone acetylation, DNA methylation and anti-cancer drug discovery. Curr Cancer Drug Targets 2009; 9: 963-81.
    • (2009) Curr Cancer Drug Targets , vol.9 , pp. 963-981
    • Ganesan, A.1    Nolan, L.2    Crabb, S.J.3    Packham, G.4
  • 28
    • 0027154222 scopus 로고
    • Effects of treatment with azacitidine on the in vivo and in vitro hematopoiesis in patients with myelodysplastic syndromes
    • Silverman LR, Holland JF, Weinberg RS, et al. Effects of treatment with azacitidine on the in vivo and in vitro hematopoiesis in patients with myelodysplastic syndromes. Leukemia 1993; 7 (Suppl. 1): 21-9.
    • (1993) Leukemia , vol.7 , Issue.SUPPL. 1 , pp. 21-29
    • Silverman, L.R.1    Holland, J.F.2    Weinberg, R.S.3
  • 29
    • 0037093195 scopus 로고    scopus 로고
    • Randomized controlled trial of azacitidine in patients with the myelodysplastic syndrome: A study of the cancer and leukemia group B
    • 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 2002; 20: 2429-40.
    • (2002) J Clin Oncol , vol.20 , pp. 2429-2440
    • Silverman, L.R.1    Demakos, E.P.2    Peterson, B.L.3
  • 30
    • 76549092572 scopus 로고    scopus 로고
    • 5-azacitidine prolongs overall survival in patients with myelodysplastic syndrome-a systematic review and meta-analysis
    • Gurion R, Vidal L, Gafter-Gvili A, et al. 5-azacitidine prolongs overall survival in patients with myelodysplastic syndrome-a systematic review and meta-analysis. Haematologica 2010; 95 (2): 303-10.
    • (2010) Haematologica , vol.95 , Issue.2 , pp. 303-310
    • Gurion, R.1    Vidal, L.2    Gafter-Gvili, A.3
  • 31
    • 84877574817 scopus 로고    scopus 로고
    • The future of epigenetic therapy in solid tumours-lessons from the past
    • Azad N, Zahnow CA, Rudin CM, Baylin SB,. The future of epigenetic therapy in solid tumours-lessons from the past. Nat Rev Clin Oncol 2013; 10 (5): 256-66.
    • (2013) Nat Rev Clin Oncol , vol.10 , Issue.5 , pp. 256-266
    • Azad, N.1    Zahnow, C.A.2    Rudin, C.M.3    Baylin, S.B.4
  • 32
    • 62849104641 scopus 로고    scopus 로고
    • Efficacy of azacitidine compared with that of conventional care regimens in the treatment of higher-risk myelodysplastic syndromes: A randomised, open-label, phase III study
    • Fenaux P, Mufti GJ, Hellstrom-Lindberg E, et al. Efficacy of azacitidine compared with that of conventional care regimens in the treatment of higher-risk myelodysplastic syndromes: a randomised, open-label, phase III study. Lancet Oncol 2009; 10 (3): 223-32.
    • (2009) Lancet Oncol , vol.10 , Issue.3 , pp. 223-232
    • Fenaux, P.1    Mufti, G.J.2    Hellstrom-Lindberg, E.3
  • 33
    • 0030985018 scopus 로고    scopus 로고
    • Continuous infusion of low-dose 5-Aza-2′-deoxycytidine in elderly patients with high-risk myelodysplastic syndrome
    • Wijermans PW, Krulder JW, Huijgens PC, Neve P,. Continuous infusion of low-dose 5-Aza-2′-deoxycytidine in elderly patients with high-risk myelodysplastic syndrome. Leukemia 1997; 11: 1-5.
    • (1997) Leukemia , vol.11 , pp. 1-5
    • Wijermans, P.W.1    Krulder, J.W.2    Huijgens, P.C.3    Neve, P.4
  • 34
    • 17444452612 scopus 로고    scopus 로고
    • Low-dose 5-aza-2′-deoxycytidine, a DNA hypomethylating agent, for the treatment of high-risk myelodysplastic syndrome: A multicenter phase II study in elderly patients
    • Wijermans P, Lubbert M, Verhoef G, et al. Low-dose 5-aza-2′- deoxycytidine, a DNA hypomethylating agent, for the treatment of high-risk myelodysplastic syndrome: a multicenter phase II study in elderly patients. J Clin Oncol 2000; 18: 956-62.
    • (2000) J Clin Oncol , vol.18 , pp. 956-962
    • Wijermans, P.1    Lubbert, M.2    Verhoef, G.3
  • 37
    • 84873171161 scopus 로고    scopus 로고
    • Epigenetics and epilepsy
    • Roopra A, Dingledine R, Hsieh J,. Epigenetics and epilepsy. Epilepsia 2012; 53 (Suppl. 9): 2-10.
    • (2012) Epilepsia , vol.53 , Issue.SUPPL. 9 , pp. 2-10
    • Roopra, A.1    Dingledine, R.2    Hsieh, J.3
  • 38
    • 1842411320 scopus 로고    scopus 로고
    • Crystal structure of the nucleosome core particle at 2.8 Å resolution
    • Luger K, Mader AW, Richmond RK, et al. Crystal structure of the nucleosome core particle at 2.8 Å resolution. Nature 1997; 389: 251-60.
    • (1997) Nature , vol.389 , pp. 251-260
    • Luger, K.1    Mader, A.W.2    Richmond, R.K.3
  • 39
    • 79955811415 scopus 로고    scopus 로고
    • Nucleosome structure(s) and stability: Variations on a theme
    • Andrews AJ, Luger K,. Nucleosome structure(s) and stability: variations on a theme. Annu Rev Biophys 2011; 40: 99-117.
    • (2011) Annu Rev Biophys , vol.40 , pp. 99-117
    • Andrews, A.J.1    Luger, K.2
  • 40
    • 84993729302 scopus 로고    scopus 로고
    • Epigenetic therapy of hematological malignancies: Where are we now?
    • Popovic R, Shah MY, Licht JD,. Epigenetic therapy of hematological malignancies: where are we now? Ther Adv Hematol 2013; 4: 81-91.
    • (2013) Ther Adv Hematol , vol.4 , pp. 81-91
    • Popovic, R.1    Shah, M.Y.2    Licht, J.D.3
  • 41
    • 33646070846 scopus 로고    scopus 로고
    • A bivalent chromatin structure marks key developmental genes in embryonic stem cells
    • Bernstein BE, Mikkelsen TS, Xie X, et al. A bivalent chromatin structure marks key developmental genes in embryonic stem cells. Cell 2006; 125: 315-26.
    • (2006) Cell , vol.125 , pp. 315-326
    • Bernstein, B.E.1    Mikkelsen, T.S.2    Xie, X.3
  • 42
    • 0034610814 scopus 로고    scopus 로고
    • The language of covalent histone modifications
    • Strahl BD, Allis CD,. The language of covalent histone modifications. Nature 2000; 403: 41-5.
    • (2000) Nature , vol.403 , pp. 41-45
    • Strahl, B.D.1    Allis, C.D.2
  • 43
    • 0035839136 scopus 로고    scopus 로고
    • Translating the histone code
    • Jenuwein T, Allis CD,. Translating the histone code. Science 2001; 293: 1074-80.
    • (2001) Science , vol.293 , pp. 1074-1080
    • Jenuwein, T.1    Allis, C.D.2
  • 44
    • 36448949026 scopus 로고    scopus 로고
    • Multivalent engagement of chromatin modifications by linked binding modules
    • Ruthenburg AJ, Li H, Patel DJ, Allis CD,. Multivalent engagement of chromatin modifications by linked binding modules. Nat Rev Mol Cell Biol 2007; 8: 983-94.
    • (2007) Nat Rev Mol Cell Biol , vol.8 , pp. 983-994
    • Ruthenburg, A.J.1    Li, H.2    Patel, D.J.3    Allis, C.D.4
  • 45
    • 77952853306 scopus 로고    scopus 로고
    • Histone deacetylases and epigenetic therapies of hematological malignancies
    • Mercurio C, Minucci S, Pelicci PG,. Histone deacetylases and epigenetic therapies of hematological malignancies. Pharmacol Res 2010; 62: 18-34.
    • (2010) Pharmacol Res , vol.62 , pp. 18-34
    • Mercurio, C.1    Minucci, S.2    Pelicci, P.G.3
  • 46
    • 80052653226 scopus 로고    scopus 로고
    • Therapeutic strategies to enhance the anticancer efficacy of histone deacetylase inhibitors
    • Miller CP, Singh MM, Rivera-Del Valle N, et al. Therapeutic strategies to enhance the anticancer efficacy of histone deacetylase inhibitors. J Biomed Biotechnol 2011; 2011: 514261.
    • (2011) J Biomed Biotechnol , vol.2011 , pp. 514261
    • Miller, C.P.1    Singh, M.M.2    Rivera-Del Valle, N.3
  • 47
    • 84859181036 scopus 로고    scopus 로고
    • Histone recognition and large-scale structural analysis of the human bromodomain family
    • Filippakopoulos P, Picaud S, Mangos M, et al. Histone recognition and large-scale structural analysis of the human bromodomain family. Cell 2012; 149: 214-31.
    • (2012) Cell , vol.149 , pp. 214-231
    • Filippakopoulos, P.1    Picaud, S.2    Mangos, M.3
  • 48
    • 0030797585 scopus 로고    scopus 로고
    • Activation of p53 sequence-specific DNA binding by acetylation of the p53 C-terminal domain
    • Gu W, Roeder RG,. Activation of p53 sequence-specific DNA binding by acetylation of the p53 C-terminal domain. Cell 1997; 90: 595-606.
    • (1997) Cell , vol.90 , pp. 595-606
    • Gu, W.1    Roeder, R.G.2
  • 49
    • 0037377060 scopus 로고    scopus 로고
    • Ubiquitination, phosphorylation and acetylation: The molecular basis for p53 regulation
    • Brooks CL, Gu W,. Ubiquitination, phosphorylation and acetylation: the molecular basis for p53 regulation. Curr Opin Cell Biol 2003; 15: 164-71.
    • (2003) Curr Opin Cell Biol , vol.15 , pp. 164-171
    • Brooks, C.L.1    Gu, W.2
  • 50
    • 0034676439 scopus 로고    scopus 로고
    • Deacetylation of p53 modulates its effect on cell growth and apoptosis
    • Luo J, Su F, Chen D, Shiloh A, Gu W,. Deacetylation of p53 modulates its effect on cell growth and apoptosis. Nature 2000; 408: 377-81.
    • (2000) Nature , vol.408 , pp. 377-381
    • Luo, J.1    Su, F.2    Chen, D.3    Shiloh, A.4    Gu, W.5
  • 52
    • 0036898253 scopus 로고    scopus 로고
    • Acetylation inactivates the transcriptional repressor BCL6
    • Bereshchenko OR, Gu W, Dalla-Favera R,. Acetylation inactivates the transcriptional repressor BCL6. Nat Genet 2002; 32: 606-13.
    • (2002) Nat Genet , vol.32 , pp. 606-613
    • Bereshchenko, O.R.1    Gu, W.2    Dalla-Favera, R.3
  • 53
    • 80054736157 scopus 로고    scopus 로고
    • Potential non-oncological applications of histone deacetylase inhibitors
    • Ververis K, Karagiannis TC,. Potential non-oncological applications of histone deacetylase inhibitors. Am J Transl Res 2011; 3: 454-67.
    • (2011) Am J Transl Res , vol.3 , pp. 454-467
    • Ververis, K.1    Karagiannis, T.C.2
  • 54
    • 78650848615 scopus 로고    scopus 로고
    • Histone deacetylase inhibitors in the treatment of lymphoma
    • Lemoine M, Younes A,. Histone deacetylase inhibitors in the treatment of lymphoma. Discov Med 2010; 10: 462-70.
    • (2010) Discov Med , vol.10 , pp. 462-470
    • Lemoine, M.1    Younes, A.2
  • 55
    • 33845996135 scopus 로고    scopus 로고
    • Phase 2 trial of oral vorinostat (suberoylanilide hydroxamic acid, SAHA) for refractory cutaneous T-cell lymphoma (CTCL)
    • 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). Blood 2007; 109: 31-9.
    • (2007) Blood , vol.109 , pp. 31-39
    • Duvic, M.1    Talpur, R.2    Ni, X.3
  • 56
    • 34547683194 scopus 로고    scopus 로고
    • Phase IIB multicenter trial of vorinostat in patients with persistent, progressive, or treatment refractory cutaneous T-cell lymphoma
    • Olsen EA, Kim YH, Kuzel TM, et al. Phase IIB multicenter trial of vorinostat in patients with persistent, progressive, or treatment refractory cutaneous T-cell lymphoma. J Clin Oncol 2007; 25: 3109-15.
    • (2007) J Clin Oncol , vol.25 , pp. 3109-3115
    • Olsen, E.A.1    Kim, Y.H.2    Kuzel, T.M.3
  • 57
    • 77952140455 scopus 로고    scopus 로고
    • Update on the treatment of cutaneous T-cell lymphoma (CTCL): Focus on vorinostat
    • Duvic M, Vu J,. Update on the treatment of cutaneous T-cell lymphoma (CTCL): focus on vorinostat. Biologics 2007; 1: 377-92.
    • (2007) Biologics , vol.1 , pp. 377-392
    • Duvic, M.1    Vu, J.2
  • 58
    • 36148950997 scopus 로고    scopus 로고
    • FDA approval summary: Vorinostat for treatment of advanced primary cutaneous T-cell lymphoma
    • Mann BS, Johnson JR, Cohen MH, Justice R, Pazdur R,. FDA approval summary: Vorinostat for treatment of advanced primary cutaneous T-cell lymphoma. Oncologist 2007; 12: 1247-52.
    • (2007) Oncologist , vol.12 , pp. 1247-1252
    • Mann, B.S.1    Johnson, J.R.2    Cohen, M.H.3    Justice, R.4    Pazdur, R.5
  • 59
    • 84855471990 scopus 로고    scopus 로고
    • HDAC inhibitors in cancer biology: Emerging mechanisms and clinical applications
    • Khan O, La Thangue NB,. HDAC inhibitors in cancer biology: emerging mechanisms and clinical applications. Immunol Cell Biol 2012; 90: 85-94.
    • (2012) Immunol Cell Biol , vol.90 , pp. 85-94
    • Khan, O.1    La Thangue, N.B.2
  • 60
    • 79960185557 scopus 로고    scopus 로고
    • Romidepsin (Istodax, NSC 630176, FR901228, FK228, depsipeptide): A natural product recently approved for cutaneous T-cell lymphoma
    • VanderMolen KM, McCulloch W, Pearce CJ, Oberlies NH,. Romidepsin (Istodax, NSC 630176, FR901228, FK228, depsipeptide): a natural product recently approved for cutaneous T-cell lymphoma. J Antibiot (Tokyo) 2011; 64: 525-31.
    • (2011) J Antibiot (Tokyo) , vol.64 , pp. 525-531
    • Vandermolen, K.M.1    McCulloch, W.2    Pearce, C.J.3    Oberlies, N.H.4
  • 61
    • 67449127082 scopus 로고    scopus 로고
    • Clinical studies of histone deacetylase inhibitors
    • Prince HM, Bishton MJ, Harrison SJ,. Clinical studies of histone deacetylase inhibitors. Clin Cancer Res 2009; 15: 3958-69.
    • (2009) Clin Cancer Res , vol.15 , pp. 3958-3969
    • Prince, H.M.1    Bishton, M.J.2    Harrison, S.J.3
  • 62
    • 73949149251 scopus 로고    scopus 로고
    • Phase II multi-institutional trial of the histone deacetylase inhibitor romidepsin as monotherapy for patients with cutaneous T-cell lymphoma
    • Piekarz RL, Frye R, Turner M, et al. Phase II multi-institutional trial of the histone deacetylase inhibitor romidepsin as monotherapy for patients with cutaneous T-cell lymphoma. J Clin Oncol 2009; 27: 5410-7.
    • (2009) J Clin Oncol , vol.27 , pp. 5410-5417
    • Piekarz, R.L.1    Frye, R.2    Turner, M.3
  • 63
    • 77954879663 scopus 로고    scopus 로고
    • Final results from a multicenter, international, pivotal study of romidepsin in refractory cutaneous T-cell lymphoma
    • Whittaker SJ, Demierre MF, Kim EJ, et al. Final results from a multicenter, international, pivotal study of romidepsin in refractory cutaneous T-cell lymphoma. J Clin Oncol 2010; 28: 4485-91.
    • (2010) J Clin Oncol , vol.28 , pp. 4485-4491
    • Whittaker, S.J.1    Demierre, M.F.2    Kim, E.J.3
  • 64
    • 79952977561 scopus 로고    scopus 로고
    • Phase 2 trial of romidepsin in patients with peripheral T-cell lymphoma
    • Piekarz RL, Frye R, Prince HM, et al. Phase 2 trial of romidepsin in patients with peripheral T-cell lymphoma. Blood 2011; 117: 5827-34.
    • (2011) Blood , vol.117 , pp. 5827-5834
    • Piekarz, R.L.1    Frye, R.2    Prince, H.M.3
  • 65
    • 55949136562 scopus 로고    scopus 로고
    • Roles of the EZH2 histone methyltransferase in cancer epigenetics
    • Simon JA, Lange CA,. Roles of the EZH2 histone methyltransferase in cancer epigenetics. Mutat Res 2008; 647: 21-9.
    • (2008) Mutat Res , vol.647 , pp. 21-29
    • Simon, J.A.1    Lange, C.A.2
  • 66
    • 77954758157 scopus 로고    scopus 로고
    • Polycomb group protein-mediated repression of transcription
    • Morey L, Helin K,. Polycomb group protein-mediated repression of transcription. Trends Biochem Sci 2010; 35: 323-32.
    • (2010) Trends Biochem Sci , vol.35 , pp. 323-332
    • Morey, L.1    Helin, K.2
  • 67
    • 79955513100 scopus 로고    scopus 로고
    • Aberrations of EZH2 in cancer
    • Chase A, Cross NC,. Aberrations of EZH2 in cancer. Clin Cancer Res 2011; 17: 2613-8.
    • (2011) Clin Cancer Res , vol.17 , pp. 2613-2618
    • Chase, A.1    Cross, N.C.2
  • 68
    • 79952105934 scopus 로고    scopus 로고
    • Enhancer of zeste homolog 2: A potential target for tumor therapy
    • Xiao Y,. Enhancer of zeste homolog 2: a potential target for tumor therapy. Int J Biochem Cell Biol 2011; 43: 474-7.
    • (2011) Int J Biochem Cell Biol , vol.43 , pp. 474-477
    • Xiao, Y.1
  • 69
    • 84867632489 scopus 로고    scopus 로고
    • A selective inhibitor of EZH2 blocks H3K27 methylation and kills mutant lymphoma cells
    • Knutson SK, Wigle TJ, Warholic NM, et al. A selective inhibitor of EZH2 blocks H3K27 methylation and kills mutant lymphoma cells. Nat Chem Biol 2012; 8: 890-6.
    • (2012) Nat Chem Biol , vol.8 , pp. 890-896
    • Knutson, S.K.1    Wigle, T.J.2    Warholic, N.M.3
  • 70
    • 84870573126 scopus 로고    scopus 로고
    • EZH2 inhibition as a therapeutic strategy for lymphoma with EZH2-activating mutations
    • McCabe MT, Ott HM, Ganji G, et al. EZH2 inhibition as a therapeutic strategy for lymphoma with EZH2-activating mutations. Nature 2012; 492: 108-12.
    • (2012) Nature , vol.492 , pp. 108-112
    • McCabe, M.T.1    Ott, H.M.2    Ganji, G.3
  • 71
    • 77953995002 scopus 로고    scopus 로고
    • Covalent histone modifications-miswritten, misinterpreted and mis-erased in human cancers
    • Chi P, Allis CD, Wang GG,. Covalent histone modifications-miswritten, misinterpreted and mis-erased in human cancers. Nat Rev Cancer 2010; 10: 457-69.
    • (2010) Nat Rev Cancer , vol.10 , pp. 457-469
    • Chi, P.1    Allis, C.D.2    Wang, G.G.3
  • 72
    • 79952112943 scopus 로고    scopus 로고
    • Dynamics of histone lysine methylation: Structures of methyl writers and erasers
    • Upadhyay AK, Cheng X,. Dynamics of histone lysine methylation: structures of methyl writers and erasers. Prog Drug Res 2011; 67: 107-24.
    • (2011) Prog Drug Res , vol.67 , pp. 107-124
    • Upadhyay, A.K.1    Cheng, X.2
  • 74
    • 84862738480 scopus 로고    scopus 로고
    • BET domain co-regulators in obesity, inflammation and cancer
    • Belkina AC, Denis GV,. BET domain co-regulators in obesity, inflammation and cancer. Nat Rev Cancer 2012; 12: 465-77.
    • (2012) Nat Rev Cancer , vol.12 , pp. 465-477
    • Belkina, A.C.1    Denis, G.V.2
  • 75
    • 0026608175 scopus 로고
    • The bromodomain: A conserved sequence found in human, Drosophila and yeast proteins
    • Haynes SR, et al. The bromodomain: a conserved sequence found in human, Drosophila and yeast proteins. Nucl Acids Res 1992; 20: 2603.
    • (1992) Nucl Acids Res , vol.20 , pp. 2603
    • Haynes, S.R.1
  • 76
    • 34250357662 scopus 로고    scopus 로고
    • The double bromodomain-containing chromatin adaptor Brd4 and transcriptional regulation
    • Wu SY, Chiang CM,. The double bromodomain-containing chromatin adaptor Brd4 and transcriptional regulation. J Biol Chem 2007; 282: 13141-5.
    • (2007) J Biol Chem , vol.282 , pp. 13141-13145
    • Wu, S.Y.1    Chiang, C.M.2
  • 77
    • 84866338076 scopus 로고    scopus 로고
    • Druggability analysis and structural classification of bromodomain acetyl-lysine binding sites
    • Vidler LR, Brown N, Knapp S, Hoelder S,. Druggability analysis and structural classification of bromodomain acetyl-lysine binding sites. J Med Chem 2012; 55: 7346-59.
    • (2012) J Med Chem , vol.55 , pp. 7346-7359
    • Vidler, L.R.1    Brown, N.2    Knapp, S.3    Hoelder, S.4
  • 78
    • 84870693696 scopus 로고    scopus 로고
    • Modulation of Epigenetic Targets for Anticancer Therapy: Clinicopathological Relevance, Structural Data and Drug Discovery Perspectives
    • Andreol F, Barbosa AJ, Parenti MD, Del Rio A,. Modulation of Epigenetic Targets for Anticancer Therapy: clinicopathological Relevance, Structural Data and Drug Discovery Perspectives. Curr Pharm Des 2013; 19: 578-613.
    • (2013) Curr Pharm des , vol.19 , pp. 578-613
    • Andreol, F.1    Barbosa, A.J.2    Parenti, M.D.3    Del Rio, A.4
  • 79
    • 70149105669 scopus 로고    scopus 로고
    • The role of human bromodomains in chromatin biology and gene transcription
    • Sanchez R, Zhou MM,. The role of human bromodomains in chromatin biology and gene transcription. Curr Opin Drug Discov Devel 2009; 12: 659-65.
    • (2009) Curr Opin Drug Discov Devel , vol.12 , pp. 659-665
    • Sanchez, R.1    Zhou, M.M.2
  • 80
    • 84862809749 scopus 로고    scopus 로고
    • Identification of a novel series of BET family bromodomain inhibitors: Binding mode and profile of I-BET151 (GSK1210151A)
    • Seal J, Lamotte Y, Donche F, et al. Identification of a novel series of BET family bromodomain inhibitors: binding mode and profile of I-BET151 (GSK1210151A). Bioorg Med Chem Lett 2012; 22: 2968-72.
    • (2012) Bioorg Med Chem Lett , vol.22 , pp. 2968-2972
    • Seal, J.1    Lamotte, Y.2    Donche, F.3
  • 81
    • 78650806593 scopus 로고    scopus 로고
    • Suppression of inflammation by a synthetic histone mimic
    • Nicodeme E, Jeffrey KL, Schaefer U, et al. Suppression of inflammation by a synthetic histone mimic. Nature 2010; 468: 1119-23.
    • (2010) Nature , vol.468 , pp. 1119-1123
    • Nicodeme, E.1    Jeffrey, K.L.2    Schaefer, U.3
  • 82
    • 80052955256 scopus 로고    scopus 로고
    • BET bromodomain inhibition as a therapeutic strategy to target c-Myc
    • Delmore JE, Issa GC, Lemieux ME, et al. BET bromodomain inhibition as a therapeutic strategy to target c-Myc. Cell 2011; 146: 904-17.
    • (2011) Cell , vol.146 , pp. 904-917
    • Delmore, J.E.1    Issa, G.C.2    Lemieux, M.E.3
  • 83
    • 80053651202 scopus 로고    scopus 로고
    • Targeting MYC dependence in cancer by inhibiting BET bromodomains
    • Mertz JA, Conery AR, Bryant BM, et al. Targeting MYC dependence in cancer by inhibiting BET bromodomains. Proc Natl Acad Sci USA 2011; 108: 16669-74.
    • (2011) Proc Natl Acad Sci USA , vol.108 , pp. 16669-16674
    • Mertz, J.A.1    Conery, A.R.2    Bryant, B.M.3
  • 84
    • 84881192460 scopus 로고    scopus 로고
    • BET Bromodomains Mediate Transcriptional Pause Release in Heart Failure
    • Anand P, Brown JD, Lin CY, et al. BET Bromodomains Mediate Transcriptional Pause Release in Heart Failure. Cell 2013; 154: 569-82.
    • (2013) Cell , vol.154 , pp. 569-582
    • Anand, P.1    Brown, J.D.2    Lin, C.Y.3
  • 85
    • 84865222098 scopus 로고    scopus 로고
    • Down-regulation of NF-kappaB transcriptional activity in HIV-associated kidney disease by BRD4 inhibition
    • Zhang G, Liu R, Zhong Y, et al. Down-regulation of NF-kappaB transcriptional activity in HIV-associated kidney disease by BRD4 inhibition. J Biol Chem 2012; 287: 28840-51.
    • (2012) J Biol Chem , vol.287 , pp. 28840-28851
    • Zhang, G.1    Liu, R.2    Zhong, Y.3
  • 86
    • 84894302630 scopus 로고    scopus 로고
    • New York: Nova Science Pub Inc, ISBN-13: 978-1600215742. 2007
    • Barbieri M,. Biosemiotic Research Trends. New York: Nova Science Pub Inc, 2007. ISBN-13: 978-1600215742. 2007.
    • (2007) Biosemiotic Research Trends
    • Barbieri, M.1
  • 87
    • 2042437650 scopus 로고    scopus 로고
    • Initial sequencing and analysis of the human genome
    • International Human Genome Sequencing Consortium
    • International Human Genome Sequencing Consortium. Initial sequencing and analysis of the human genome. Nature 2001; 409: 860-921.
    • (2001) Nature , vol.409 , pp. 860-921
  • 88
    • 23944504276 scopus 로고    scopus 로고
    • Synergy between sequence and size in large-scale genomics
    • Gregory TR,. Synergy between sequence and size in large-scale genomics. Nat Rev Genet 2005; 6: 699-708.
    • (2005) Nat Rev Genet , vol.6 , pp. 699-708
    • Gregory, T.R.1
  • 89
    • 72649102150 scopus 로고    scopus 로고
    • Distributions of selectively constrained sites and deleterious mutation rates in the hominid and murid genomes
    • Eory L, Halligan DL, Keightley PD,. Distributions of selectively constrained sites and deleterious mutation rates in the hominid and murid genomes. Mol Biol Evol 2010; 27: 177-92.
    • (2010) Mol Biol Evol , vol.27 , pp. 177-192
    • Eory, L.1    Halligan, D.L.2    Keightley, P.D.3
  • 90
  • 91
    • 84883499858 scopus 로고    scopus 로고
    • Epigenomics: The science of no-longer-junk DNA. Why study it in chronic kidney disease?
    • Ko YA, Susztak K,. Epigenomics: the science of no-longer-junk DNA. Why study it in chronic kidney disease? Semin Nephrol 2013; 33 (4): 354-62.
    • (2013) Semin Nephrol , vol.33 , Issue.4 , pp. 354-362
    • Ko, Y.A.1    Susztak, K.2
  • 93
    • 80055013170 scopus 로고    scopus 로고
    • From 'JUNK' to just unexplored noncoding knowledge: The case of transcribed Alus
    • Pandey R, Mukerji M,. From 'JUNK' to just unexplored noncoding knowledge: the case of transcribed Alus. Brief Funct Genomics 2011; 10: 294-311.
    • (2011) Brief Funct Genomics , vol.10 , pp. 294-311
    • Pandey, R.1    Mukerji, M.2
  • 94
    • 38349169664 scopus 로고    scopus 로고
    • Mechanisms of post-transcriptional regulation by microRNAs: Are the answers in sight?
    • Filipowicz W, Bhattacharyya SN, Sonenberg N. Mechanisms of post-transcriptional regulation by microRNAs: are the answers in sight? Nat Rev Genet 2008; 9: 102-14.
    • (2008) Nat Rev Genet , vol.9 , pp. 102-114
    • Filipowicz, W.1    Bhattacharyya, S.N.2    Sonenberg, N.3
  • 97
    • 33845755467 scopus 로고    scopus 로고
    • Illuminating the silence: Understanding the structure and function of small RNAs
    • Rana TM,. Illuminating the silence: understanding the structure and function of small RNAs. Nat Rev Mol Cell Biol 2007; 8: 23-36.
    • (2007) Nat Rev Mol Cell Biol , vol.8 , pp. 23-36
    • Rana, T.M.1
  • 98
    • 84861866572 scopus 로고    scopus 로고
    • The mechanics of miRNA-mediated gene silencing: A look under the hood of miRISC
    • Fabian MR, Sonenberg N,. The mechanics of miRNA-mediated gene silencing: a look under the hood of miRISC. Nat Struct Mol Biol 2012; 19: 586-93.
    • (2012) Nat Struct Mol Biol , vol.19 , pp. 586-593
    • Fabian, M.R.1    Sonenberg, N.2
  • 99
    • 0035261255 scopus 로고    scopus 로고
    • Post-transcriptional gene silencing by double-stranded RNA
    • Hammond SM, Caudy AA, Hannon GJ,. Post-transcriptional gene silencing by double-stranded RNA. Nat Rev Genet 2001; 2: 110-9.
    • (2001) Nat Rev Genet , vol.2 , pp. 110-119
    • Hammond, S.M.1    Caudy, A.A.2    Hannon, G.J.3
  • 100
    • 78650513539 scopus 로고    scopus 로고
    • A brief introduction to RNAi and microRNAs in stem cells
    • Murashov AK,. A brief introduction to RNAi and microRNAs in stem cells. Methods Mol Biol 2010; 650: 15-25.
    • (2010) Methods Mol Biol , vol.650 , pp. 15-25
    • Murashov, A.K.1
  • 102
    • 34248634004 scopus 로고    scopus 로고
    • Therapeutic potential for microRNAs
    • Esau CC, Monia BP,. Therapeutic potential for microRNAs. Adv Drug Deliv Rev 2007; 59: 101-14.
    • (2007) Adv Drug Deliv Rev , vol.59 , pp. 101-114
    • Esau, C.C.1    Monia, B.P.2
  • 103
    • 77956339881 scopus 로고    scopus 로고
    • OncomiR addiction in an in vivo model of microRNA-21-induced pre-B-cell lymphoma
    • Medina PP, Nolde M, Slack FJ,. OncomiR addiction in an in vivo model of microRNA-21-induced pre-B-cell lymphoma. Nature 2010; 467: 86-90.
    • (2010) Nature , vol.467 , pp. 86-90
    • Medina, P.P.1    Nolde, M.2    Slack, F.J.3
  • 104
    • 44849093562 scopus 로고    scopus 로고
    • Oncogene addiction
    • Weinstein IB, Joe A,. Oncogene addiction. Cancer Res 2008; 68: 3077-80.
    • (2008) Cancer Res , vol.68 , pp. 3077-3080
    • Weinstein, I.B.1    Joe, A.2
  • 106
    • 0028307739 scopus 로고
    • Hypoxia and drug resistance
    • Teicher BA,. Hypoxia and drug resistance. Cancer Metastasis Rev 1994; 13: 139-68.
    • (1994) Cancer Metastasis Rev , vol.13 , pp. 139-168
    • Teicher, B.A.1
  • 108
    • 77957243894 scopus 로고    scopus 로고
    • MicroRNA-210 as a novel therapy for treatment of ischemic heart disease
    • Hu S, Huang M, Li Z, et al. MicroRNA-210 as a novel therapy for treatment of ischemic heart disease. Circulation 2010; 122 (11 Suppl): S124-31.
    • (2010) Circulation , vol.122 , Issue.11 SUPPL.
    • Hu, S.1    Huang, M.2    Li, Z.3
  • 109
    • 79957900919 scopus 로고    scopus 로고
    • Systemic delivery of tumor suppressor microRNA mimics using a neutral lipid emulsion inhibits lung tumors in mice
    • Trang P, Wiggins JF, Daige CL, et al. Systemic delivery of tumor suppressor microRNA mimics using a neutral lipid emulsion inhibits lung tumors in mice. Mol Ther 2011; 19 (6): 1116-22.
    • (2011) Mol Ther , vol.19 , Issue.6 , pp. 1116-1122
    • Trang, P.1    Wiggins, J.F.2    Daige, C.L.3
  • 110
    • 79952197420 scopus 로고    scopus 로고
    • The concept of multiple-target anti-miRNA antisense oligonucleotide technology
    • Wang Z,. The concept of multiple-target anti-miRNA antisense oligonucleotide technology. Methods Mol Biol 2011; 676: 51-7.
    • (2011) Methods Mol Biol , vol.676 , pp. 51-57
    • Wang, Z.1
  • 111
    • 84855350458 scopus 로고    scopus 로고
    • Inhibition of miR-15 Protects Against Cardiac Ischemic Injury
    • Hullinger TG, Montgomery RL, Seto AG, et al. Inhibition of miR-15 Protects Against Cardiac Ischemic Injury. Circ Res 2012; 110 (1): 71-81.
    • (2012) Circ Res , vol.110 , Issue.1 , pp. 71-81
    • Hullinger, T.G.1    Montgomery, R.L.2    Seto, A.G.3
  • 112
    • 80054971110 scopus 로고    scopus 로고
    • Inhibition of miR-33a/b in non-human primates raises plasma HDL and lowers VLDL triglycerides
    • Rayner KJ, Esau CC, Hussain FN, et al. Inhibition of miR-33a/b in non-human primates raises plasma HDL and lowers VLDL triglycerides. Nature 2011; 478 (7369): 404-7.
    • (2011) Nature , vol.478 , Issue.7369 , pp. 404-407
    • Rayner, K.J.1    Esau, C.C.2    Hussain, F.N.3
  • 113
    • 57149086661 scopus 로고    scopus 로고
    • Atu027, a liposomal small interfering RNA formulation targeting protein kinase N3, inhibits cancer progression
    • Aleku M, Schulz P, Keil O, et al. Atu027, a liposomal small interfering RNA formulation targeting protein kinase N3, inhibits cancer progression. Cancer Res 2008; 68 (23): 9788-98.
    • (2008) Cancer Res , vol.68 , Issue.23 , pp. 9788-9798
    • Aleku, M.1    Schulz, P.2    Keil, O.3
  • 114
    • 78349299473 scopus 로고    scopus 로고
    • Atu027 prevents pulmonary metastasis in experimental and spontaneous mouse metastasis models
    • Santel A, Aleku M, Röder N, et al. Atu027 prevents pulmonary metastasis in experimental and spontaneous mouse metastasis models. Clin Cancer Res 2010; 16 (22): 5469-80.
    • (2010) Clin Cancer Res , vol.16 , Issue.22 , pp. 5469-5480
    • Santel, A.1    Aleku, M.2    Röder, N.3
  • 115
    • 84855168044 scopus 로고    scopus 로고
    • Phase i clinical development of Atu027, a siRNA formulation targeting PKN3 in patients with advanced solid tumors
    • Strumberg D, Schultheis B, Traugott U, et al. Phase I clinical development of Atu027, a siRNA formulation targeting PKN3 in patients with advanced solid tumors. Int J Clin Pharmacol Ther 2012; 50 (1): 76-8.
    • (2012) Int J Clin Pharmacol Ther , vol.50 , Issue.1 , pp. 76-78
    • Strumberg, D.1    Schultheis, B.2    Traugott, U.3
  • 116
    • 84877120847 scopus 로고    scopus 로고
    • First-in-humans trial of an RNA interference therapeutic targeting VEGF and KSP in cancer patients with liver involvement
    • Tabernero J, Shapiro GI, LoRusso PM, et al. First-in-humans trial of an RNA interference therapeutic targeting VEGF and KSP in cancer patients with liver involvement. Cancer Discov 2013; 3 (4): 406-17.
    • (2013) Cancer Discov , vol.3 , Issue.4 , pp. 406-417
    • Tabernero, J.1    Shapiro, G.I.2    Lorusso, P.M.3
  • 117
    • 84872175978 scopus 로고    scopus 로고
    • Dose-ranging evaluation of intravitreal siRNA PF-04523655 for diabetic macular edema (the DEGAS study)
    • Nguyen QD, Schachar RA, Nduaka CI, et al. Dose-ranging evaluation of intravitreal siRNA PF-04523655 for diabetic macular edema (the DEGAS study). Invest Ophthalmol Vis Sci 2012; 53 (12): 7666-74.
    • (2012) Invest Ophthalmol Vis Sci , vol.53 , Issue.12 , pp. 7666-7674
    • Nguyen, Q.D.1    Schachar, R.A.2    Nduaka, C.I.3
  • 118
    • 84875432959 scopus 로고    scopus 로고
    • Epigenetic therapy: Use of agents targeting deacetylation and methylation in cancer management
    • Ho AS, Turcan S, Chan TA,. Epigenetic therapy: use of agents targeting deacetylation and methylation in cancer management. Onco Targets Ther 2013; 6: 223-32.
    • (2013) Onco Targets Ther , vol.6 , pp. 223-232
    • Ho, A.S.1    Turcan, S.2    Chan, T.A.3
  • 119
    • 84879124659 scopus 로고    scopus 로고
    • Epigenetic dysregulation in schizophrenia: Molecular and clinical aspects of histone deacetylase inhibitors
    • Hasan A, Mitchell A, Schneider A, Halene T, Akbarian S,. Epigenetic dysregulation in schizophrenia: molecular and clinical aspects of histone deacetylase inhibitors. Eur Arch Psychiatry Clin Neurosci 2013; 263 (4): 273-84.
    • (2013) Eur Arch Psychiatry Clin Neurosci , vol.263 , Issue.4 , pp. 273-284
    • Hasan, A.1    Mitchell, A.2    Schneider, A.3    Halene, T.4    Akbarian, S.5
  • 120
    • 84895871392 scopus 로고    scopus 로고
    • Epigenetic mechanisms in mood disorders: Targeting neuroplasticity
    • Fass DM, Schroeder FA, Perlis RH, Haggarty SJ,. Epigenetic mechanisms in mood disorders: targeting neuroplasticity. Neuroscience 2014; 264C: 112-30.
    • (2014) Neuroscience , vol.264 C , pp. 112-130
    • Fass, D.M.1    Schroeder, F.A.2    Perlis, R.H.3    Haggarty, S.J.4
  • 121
    • 33745918328 scopus 로고    scopus 로고
    • Association between enhanced type i collagen expression and epigenetic repression of the FLI1 gene in scleroderma fibroblasts
    • Wang Y, Fan PS, Kahaleh B,. Association between enhanced type I collagen expression and epigenetic repression of the FLI1 gene in scleroderma fibroblasts. Arthritis Rheum 2006; 54 (7): 2271-9.
    • (2006) Arthritis Rheum , vol.54 , Issue.7 , pp. 2271-2279
    • Wang, Y.1    Fan, P.S.2    Kahaleh, B.3
  • 122
    • 78649292318 scopus 로고    scopus 로고
    • Targeting histone deacetylases for the treatment of Huntington's disease
    • Gray SG,. Targeting histone deacetylases for the treatment of Huntington's disease. CNS Neurosci Ther 2010; 16 (6): 348-61.
    • (2010) CNS Neurosci Ther , vol.16 , Issue.6 , pp. 348-361
    • Gray, S.G.1
  • 123
    • 83755224359 scopus 로고    scopus 로고
    • Suberoylanilide hydroxamic acid, a histone deacetylase inhibitor, protects dopaminergic neurons from neurotoxin-induced damage
    • Chen S, Wu H, Ossola B, et al. Suberoylanilide hydroxamic acid, a histone deacetylase inhibitor, protects dopaminergic neurons from neurotoxin-induced damage. Br J Pharmacol 2012; 165 (2): 494-505.
    • (2012) Br J Pharmacol , vol.165 , Issue.2 , pp. 494-505
    • Chen, S.1    Wu, H.2    Ossola, B.3
  • 124
    • 84859492923 scopus 로고    scopus 로고
    • HDAC inhibitor therapy in autoimmunity and transplantation
    • Hancock WW, Akimova T, Beier UH, Liu Y, Wang L,. HDAC inhibitor therapy in autoimmunity and transplantation. Ann Rheum Dis 2012; 71 (Suppl. 2): i46-54.
    • (2012) Ann Rheum Dis , vol.71 , Issue.SUPPL. 2
    • Hancock, W.W.1    Akimova, T.2    Beier, U.H.3    Liu, Y.4    Wang, L.5
  • 125
    • 84880616633 scopus 로고    scopus 로고
    • The class III histone deacetylase sirtuin 1 in immune suppression and its therapeutic potential in rheumatoid arthritis
    • Kong S, Yeung P, Fang D,. The class III histone deacetylase sirtuin 1 in immune suppression and its therapeutic potential in rheumatoid arthritis. J Genet Genomics 2013; 40 (7): 347-54.
    • (2013) J Genet Genomics , vol.40 , Issue.7 , pp. 347-354
    • Kong, S.1    Yeung, P.2    Fang, D.3
  • 126
    • 58149307916 scopus 로고    scopus 로고
    • Histone deacetylase inhibitors: A new mode for inhibition of cholesterol metabolism
    • Chittur SV, Sangster-Guity N, McCormick PJ,. Histone deacetylase inhibitors: a new mode for inhibition of cholesterol metabolism. BMC Genomics 2008; 9: 507.
    • (2008) BMC Genomics , vol.9 , pp. 507
    • Chittur, S.V.1    Sangster-Guity, N.2    McCormick, P.J.3
  • 127
    • 84872249052 scopus 로고    scopus 로고
    • Histone deacetylase inhibition decreases cholesterol levels in neuronal cells by modulating key genes in cholesterol synthesis, uptake and efflux
    • Nunes MJ, Moutinho M, Gama MJ, Rodrigues CM, Rodrigues E,. Histone deacetylase inhibition decreases cholesterol levels in neuronal cells by modulating key genes in cholesterol synthesis, uptake and efflux. PLoS ONE 2013; 8 (1): e53394.
    • (2013) PLoS ONE , vol.8 , Issue.1
    • Nunes, M.J.1    Moutinho, M.2    Gama, M.J.3    Rodrigues, C.M.4    Rodrigues, E.5
  • 128
    • 68049118572 scopus 로고    scopus 로고
    • Combination therapy using the small interfering RNA bevasiranib
    • Singerman L,. Combination therapy using the small interfering RNA bevasiranib. Retina 2009; 29 (6 Suppl): S49-50.
    • (2009) Retina , vol.29 , Issue.6 SUPPL.
    • Singerman, L.1
  • 129
    • 77952500113 scopus 로고    scopus 로고
    • Current status of vascular endothelial growth factor inhibition in age-related macular degeneration
    • Mousa SA, Mousa SS,. Current status of vascular endothelial growth factor inhibition in age-related macular degeneration. BioDrugs 2010; 24 (3): 183-94.
    • (2010) BioDrugs , vol.24 , Issue.3 , pp. 183-194
    • Mousa, S.A.1    Mousa, S.S.2
  • 130
    • 65549088044 scopus 로고    scopus 로고
    • Drug inhibition of HDAC3 and epigenetic control of differentiation in Apicomplexa parasites
    • Bougdour A, Maubon D, Baldacci P, et al. Drug inhibition of HDAC3 and epigenetic control of differentiation in Apicomplexa parasites. J Exp Med 2009; 206 (4): 953-66.
    • (2009) J Exp Med , vol.206 , Issue.4 , pp. 953-966
    • Bougdour, A.1    Maubon, D.2    Baldacci, P.3
  • 131
    • 79951849229 scopus 로고    scopus 로고
    • RNA interference therapy in lung transplant patients infected with respiratory syncytial virus
    • Zamora MR, Budev M, Rolfe M, et al. RNA interference therapy in lung transplant patients infected with respiratory syncytial virus. Am J Respir Crit Care Med 2011; 183 (4): 531-8.
    • (2011) Am J Respir Crit Care Med , vol.183 , Issue.4 , pp. 531-538
    • Zamora, M.R.1    Budev, M.2    Rolfe, M.3
  • 133
    • 84877258007 scopus 로고    scopus 로고
    • Treatment of HCV infection by targeting microRNA
    • Janssen HL, Reesink HW, Lawitz EJ, et al. Treatment of HCV infection by targeting microRNA. N Engl J Med 2013; 368 (18): 1685-94.
    • (2013) N Engl J Med , vol.368 , Issue.18 , pp. 1685-1694
    • Janssen, H.L.1    Reesink, H.W.2    Lawitz, E.J.3
  • 134
    • 84867709301 scopus 로고    scopus 로고
    • Epigenetics, depression and antidepressant treatment
    • Menke A, Klengel T, Binder EB,. Epigenetics, depression and antidepressant treatment. Curr Pharm Des 2012; 18 (36): 5879-89.
    • (2012) Curr Pharm des , vol.18 , Issue.36 , pp. 5879-5889
    • Menke, A.1    Klengel, T.2    Binder, E.B.3
  • 135
    • 84880598197 scopus 로고    scopus 로고
    • DNA methylation in schizophrenia: Progress and challenges of epigenetic studies
    • Nishioka M, Bundo M, Kasai K, Iwamoto K,. DNA methylation in schizophrenia: progress and challenges of epigenetic studies. Genome Med 2012; 4 (12): 96.
    • (2012) Genome Med , vol.4 , Issue.12 , pp. 96
    • Nishioka, M.1    Bundo, M.2    Kasai, K.3    Iwamoto, K.4
  • 136
    • 84881507348 scopus 로고    scopus 로고
    • A selective HDAC 1/2 inhibitor modulates chromatin and gene expression in brain and alters mouse behavior in two mood-related tests
    • Schroeder FA, Lewis MC, Fass DM, et al. A selective HDAC 1/2 inhibitor modulates chromatin and gene expression in brain and alters mouse behavior in two mood-related tests. PLoS ONE 2013; 8 (8): e71323.
    • (2013) PLoS ONE , vol.8 , Issue.8
    • Schroeder, F.A.1    Lewis, M.C.2    Fass, D.M.3
  • 137
    • 84875422528 scopus 로고    scopus 로고
    • Comprehensive DNA methylation analysis of peripheral blood cells derived from patients with first-episode schizophrenia
    • Nishioka M, Bundo M, Koike S, et al. Comprehensive DNA methylation analysis of peripheral blood cells derived from patients with first-episode schizophrenia. J Hum Genet 2013; 58 (2): 91-7.
    • (2013) J Hum Genet , vol.58 , Issue.2 , pp. 91-97
    • Nishioka, M.1    Bundo, M.2    Koike, S.3
  • 138
    • 48149096013 scopus 로고    scopus 로고
    • Epigenetic mechanisms in drug addiction
    • Renthal W, Nestler EJ,. Epigenetic mechanisms in drug addiction. Trends Mol Med 2008; 14 (8): 341-50.
    • (2008) Trends Mol Med , vol.14 , Issue.8 , pp. 341-350
    • Renthal, W.1    Nestler, E.J.2
  • 139
    • 51649083501 scopus 로고    scopus 로고
    • A microRNA DNA methylation signature for human cancer metastasis
    • Lujambio A, Calin GA, Villanueva A, et al. A microRNA DNA methylation signature for human cancer metastasis. Proc Natl Acad Sci USA 2008; 105 (36): 13556-61.
    • (2008) Proc Natl Acad Sci USA , vol.105 , Issue.36 , pp. 13556-13561
    • Lujambio, A.1    Calin, G.A.2    Villanueva, A.3
  • 140
    • 65649092616 scopus 로고    scopus 로고
    • Regulation of RUNX3 tumor suppressor gene expression in cutaneous melanoma
    • Kitago M, Martinez SR, Nakamura T, Sim MS, Hoon DS,. Regulation of RUNX3 tumor suppressor gene expression in cutaneous melanoma. Clin Cancer Res 2009; 15 (9): 2988-94.
    • (2009) Clin Cancer Res , vol.15 , Issue.9 , pp. 2988-2994
    • Kitago, M.1    Martinez, S.R.2    Nakamura, T.3    Sim, M.S.4    Hoon, D.S.5
  • 141
    • 0037238431 scopus 로고    scopus 로고
    • Differential targets of CpG island hypermethylation in primary and metastatic head and neck squamous cell carcinoma (HNSCC)
    • Smiraglia DJ, Smith LT, Lang JC, et al. Differential targets of CpG island hypermethylation in primary and metastatic head and neck squamous cell carcinoma (HNSCC). J Med Genet 2003; 40 (1): 25-33.
    • (2003) J Med Genet , vol.40 , Issue.1 , pp. 25-33
    • Smiraglia, D.J.1    Smith, L.T.2    Lang, J.C.3
  • 142
    • 0141565167 scopus 로고    scopus 로고
    • Genetic and epigenetic alterations in sentinel lymph nodes metastatic lesions compared to their corresponding primary breast tumors
    • Cavalli LR, Urban CA, Dai D, et al. Genetic and epigenetic alterations in sentinel lymph nodes metastatic lesions compared to their corresponding primary breast tumors. Cancer Genet Cytogenet 2003; 146 (1): 33-40.
    • (2003) Cancer Genet Cytogenet , vol.146 , Issue.1 , pp. 33-40
    • Cavalli, L.R.1    Urban, C.A.2    Dai, D.3
  • 143
    • 84862201480 scopus 로고    scopus 로고
    • Methylation signature of lymph node metastases in breast cancer patients
    • Barekati Z, Radpour R, Lu Q, et al. Methylation signature of lymph node metastases in breast cancer patients. BMC Cancer 2012; 12: 244.
    • (2012) BMC Cancer , vol.12 , pp. 244
    • Barekati, Z.1    Radpour, R.2    Lu, Q.3
  • 144
    • 77149177050 scopus 로고    scopus 로고
    • Stage III colorectal cancer: Molecular disparity between primary cancers and lymph node metastases
    • Messick CA, Church JM, Liu X, Ting AH, Kalady MF,. Stage III colorectal cancer: molecular disparity between primary cancers and lymph node metastases. Ann Surg Oncol 2010; 17 (2): 425-31.
    • (2010) Ann Surg Oncol , vol.17 , Issue.2 , pp. 425-431
    • Messick, C.A.1    Church, J.M.2    Liu, X.3    Ting, A.H.4    Kalady, M.F.5
  • 145
    • 84863638528 scopus 로고    scopus 로고
    • Epigenetic alterations in bladder cancer and their potential clinical implications
    • Han H, Wolff EM, Liang G,. Epigenetic alterations in bladder cancer and their potential clinical implications. Adv Urol 2012; 2012: 546917.
    • (2012) Adv Urol , vol.2012 , pp. 546917
    • Han, H.1    Wolff, E.M.2    Liang, G.3
  • 146
    • 29244469466 scopus 로고    scopus 로고
    • The epigenetic progenitor origin of human cancer
    • Feinberg AP, Ohlsson R, Henikoff S,. The epigenetic progenitor origin of human cancer. Nat Rev Genet 2006; 7 (1): 21-33.
    • (2006) Nat Rev Genet , vol.7 , Issue.1 , pp. 21-33
    • Feinberg, A.P.1    Ohlsson, R.2    Henikoff, S.3
  • 147
    • 25844530426 scopus 로고    scopus 로고
    • Epigenetic changes in solid and hematopoietic tumors
    • Toyota M, Issa JP,. Epigenetic changes in solid and hematopoietic tumors. Semin Oncol 2005; 32 (5): 521-30.
    • (2005) Semin Oncol , vol.32 , Issue.5 , pp. 521-530
    • Toyota, M.1    Issa, J.P.2
  • 148
    • 40949105143 scopus 로고    scopus 로고
    • Further upregulation of beta-catenin/Tcf transcription is involved in the development of macroscopic tumors in the colon of ApcMin/+ mice
    • Oyama T, Yamada Y, Hata K, et al. Further upregulation of beta-catenin/Tcf transcription is involved in the development of macroscopic tumors in the colon of ApcMin/+ mice. Carcinogenesis 2008; 29 (3): 666-72.
    • (2008) Carcinogenesis , vol.29 , Issue.3 , pp. 666-672
    • Oyama, T.1    Yamada, Y.2    Hata, K.3
  • 149
    • 0034665332 scopus 로고    scopus 로고
    • Fields of aberrant CpG island hypermethylation in Barrett's esophagus and associated adenocarcinoma
    • Eads CA, Lord RV, Kurumboor SK, et al. Fields of aberrant CpG island hypermethylation in Barrett's esophagus and associated adenocarcinoma. Cancer Res 2000; 60 (18): 5021-6.
    • (2000) Cancer Res , vol.60 , Issue.18 , pp. 5021-5026
    • Eads, C.A.1    Lord, R.V.2    Kurumboor, S.K.3
  • 150
    • 0034326238 scopus 로고    scopus 로고
    • Predicting lung cancer by detecting aberrant promoter methylation in sputum
    • Palmisano WA, Divine KK, Saccomanno G, et al. Predicting lung cancer by detecting aberrant promoter methylation in sputum. Cancer Res 2000; 60: 5954-8.
    • (2000) Cancer Res , vol.60 , pp. 5954-5958
    • Palmisano, W.A.1    Divine, K.K.2    Saccomanno, G.3
  • 151
    • 3142672297 scopus 로고    scopus 로고
    • DNA methylation polymorphisms precede any histological sign of atherosclerosis in mice lacking apolipoprotein e
    • Lund G, Andersson L, Lauria M, et al. DNA methylation polymorphisms precede any histological sign of atherosclerosis in mice lacking apolipoprotein E. J Biol Chem 2004; 279 (28): 29147-54.
    • (2004) J Biol Chem , vol.279 , Issue.28 , pp. 29147-29154
    • Lund, G.1    Andersson, L.2    Lauria, M.3
  • 152
    • 79952805247 scopus 로고    scopus 로고
    • Cardiovascular epigenetics: Basic concepts and results from animal and human studies
    • Baccarelli A, Rienstra M, Benjamin EJ,. Cardiovascular epigenetics: basic concepts and results from animal and human studies. Circ Cardiovasc Genet 2010; 3 (6): 567-73.
    • (2010) Circ Cardiovasc Genet , vol.3 , Issue.6 , pp. 567-573
    • Baccarelli, A.1    Rienstra, M.2    Benjamin, E.J.3
  • 153
    • 33845333815 scopus 로고    scopus 로고
    • Mapping geographic zones of cancer risk with epigenetic biomarkers in normal breast tissue
    • Yan PS, Venkataramu C, Ibrahim A, et al. Mapping geographic zones of cancer risk with epigenetic biomarkers in normal breast tissue. Clin Cancer Res 2006; 12: 6626-36.
    • (2006) Clin Cancer Res , vol.12 , pp. 6626-6636
    • Yan, P.S.1    Venkataramu, C.2    Ibrahim, A.3
  • 154
    • 84872496432 scopus 로고    scopus 로고
    • MicroRNAs as potential biomarkers in human solid tumors
    • Shen J, Stass SA, Jiang F,. MicroRNAs as potential biomarkers in human solid tumors. Cancer Lett 2013; 329 (2): 125-36.
    • (2013) Cancer Lett , vol.329 , Issue.2 , pp. 125-136
    • Shen, J.1    Stass, S.A.2    Jiang, F.3
  • 155
    • 84878242633 scopus 로고    scopus 로고
    • Methylated DNA and microRNA in Body Fluids as Biomarkers for Cancer Detection
    • Ma Y, Wang X, Jin H,. Methylated DNA and microRNA in Body Fluids as Biomarkers for Cancer Detection. Int J Mol Sci 2013; 14 (5): 10307-31.
    • (2013) Int J Mol Sci , vol.14 , Issue.5 , pp. 10307-10331
    • Ma, Y.1    Wang, X.2    Jin, H.3
  • 156
    • 84857159930 scopus 로고    scopus 로고
    • Performance evaluation of the DNA methylation biomarker SHOX2 for the aid in diagnosis of lung cancer based on the analysis of bronchial aspirates
    • Dietrich D, Kneip C, Raji O, et al. Performance evaluation of the DNA methylation biomarker SHOX2 for the aid in diagnosis of lung cancer based on the analysis of bronchial aspirates. Int J Oncol 2012; 40 (3): 825-32.
    • (2012) Int J Oncol , vol.40 , Issue.3 , pp. 825-832
    • Dietrich, D.1    Kneip, C.2    Raji, O.3
  • 157
    • 84891743167 scopus 로고    scopus 로고
    • Prospective evaluation of methylated SEPT9 in plasma for detection of asymptomatic colorectal cancer
    • Church TR, Wandell M, Lofton-Day C, et al. Prospective evaluation of methylated SEPT9 in plasma for detection of asymptomatic colorectal cancer. Gut 2014; 63 (2): 317-25.
    • (2014) Gut , vol.63 , Issue.2 , pp. 317-325
    • Church, T.R.1    Wandell, M.2    Lofton-Day, C.3
  • 158
    • 84866661786 scopus 로고    scopus 로고
    • Detection of methylated SEPT9 in plasma is a reliable screening method for both left- and right-sided colon cancers
    • Tóth K, Sipos F, Kalmár A, et al. Detection of methylated SEPT9 in plasma is a reliable screening method for both left- and right-sided colon cancers. PLoS ONE 2012; 7 (9): e46000.
    • (2012) PLoS ONE , vol.7 , Issue.9
    • Tóth, K.1    Sipos, F.2    Kalmár, A.3
  • 159
    • 84902667985 scopus 로고    scopus 로고
    • Investigation of the HDAC inhibitor resminostat in patients with sorafenib-resistant hepatocellular carcinoma (HCC): Clinical data from the Phase I/II SHELTER Study
    • Poster presented at the 19-21 January San Francisco, CA
    • Bitzer M, Horger M, Ganten T, et al. Investigation of the HDAC inhibitor resminostat in patients with sorafenib-resistant hepatocellular carcinoma (HCC): clinical data from the Phase I/II SHELTER Study. Poster presented at the 19-21 January, 2012 Gastrointestinal Cancer Symposium (ASCO, ASTRO, SSO), San Francisco, CA.
    • (2012) Gastrointestinal Cancer Symposium (ASCO, ASTRO, SSO)
    • Bitzer, M.1    Horger, M.2    Ganten, T.3
  • 160
    • 79952266135 scopus 로고    scopus 로고
    • Phase 1b-2a study to reverse platinum resistance through use of a hypomethylating agent, azacitidine, in patients with platinum-resistant or platinum-refractory epithelial ovarian cancer
    • Fu S, Hu W, Iyer R, et al. Phase 1b-2a study to reverse platinum resistance through use of a hypomethylating agent, azacitidine, in patients with platinum-resistant or platinum-refractory epithelial ovarian cancer. Cancer 2011; 117 (8): 1661-9.
    • (2011) Cancer , vol.117 , Issue.8 , pp. 1661-1669
    • Fu, S.1    Hu, W.2    Iyer, R.3
  • 161
    • 81355146483 scopus 로고    scopus 로고
    • N6-methyladenosine in nuclear RNA is a major substrate of the obesity-associated FTO
    • Jia G, Fu Y, Zhao X, et al. N6-methyladenosine in nuclear RNA is a major substrate of the obesity-associated FTO. Nat Chem Biol 2011; 7 (12): 885-7.
    • (2011) Nat Chem Biol , vol.7 , Issue.12 , pp. 885-887
    • Jia, G.1    Fu, Y.2    Zhao, X.3
  • 162
    • 84872274463 scopus 로고    scopus 로고
    • ALKBH5 is a mammalian RNA demethylase that impacts RNA metabolism and mouse fertility
    • Zheng G, Dahl JA, Niu Y, et al. ALKBH5 is a mammalian RNA demethylase that impacts RNA metabolism and mouse fertility. Mol Cell 2013; 49 (1): 18-29.
    • (2013) Mol Cell , vol.49 , Issue.1 , pp. 18-29
    • Zheng, G.1    Dahl, J.A.2    Niu, Y.3
  • 163
    • 84894599601 scopus 로고    scopus 로고
    • Sprouts of RNA epigenetics: The discovery of mammalian RNA demethylases
    • Zheng G, Dahl JA, Niu Y, et al. Sprouts of RNA epigenetics: the discovery of mammalian RNA demethylases. RNA Biol 2013; 10 (6): 915-8.
    • (2013) RNA Biol , vol.10 , Issue.6 , pp. 915-918
    • Zheng, G.1    Dahl, J.A.2    Niu, Y.3
  • 164
    • 73749088654 scopus 로고    scopus 로고
    • Epigenetic opportunities and challenges in cancer
    • Best JD, Carey N,. Epigenetic opportunities and challenges in cancer. Drug Discov Today 2010; 15 (1-2): 65-70.
    • (2010) Drug Discov Today , vol.15 , Issue.12 , pp. 65-70
    • Best, J.D.1    Carey, N.2
  • 165
    • 79952116389 scopus 로고    scopus 로고
    • Targeted epigenetic therapies: The next frontier?
    • Tuma RS,. Targeted epigenetic therapies: the next frontier? J Natl Cancer Inst 2010; 102 (24): 1824-5.
    • (2010) J Natl Cancer Inst , vol.102 , Issue.24 , pp. 1824-1825
    • Tuma, R.S.1
  • 166
    • 84867082035 scopus 로고    scopus 로고
    • Epigenetic cancer therapy: Rationales, targets and drugs
    • Rius M, Lyko F,. Epigenetic cancer therapy: rationales, targets and drugs. Oncogene 2012; 31 (39): 4257-65.
    • (2012) Oncogene , vol.31 , Issue.39 , pp. 4257-4265
    • Rius, M.1    Lyko, F.2
  • 167
    • 84891476446 scopus 로고    scopus 로고
    • Cancer epigenetics: New therapies and new challenges
    • Hatzimichael E, Crook T,. Cancer epigenetics: new therapies and new challenges. J Drug Deliv 2013; 2013: 529312.
    • (2013) J Drug Deliv , vol.2013 , pp. 529312
    • Hatzimichael, E.1    Crook, T.2


* 이 정보는 Elsevier사의 SCOPUS DB에서 KISTI가 분석하여 추출한 것입니다.