메뉴 건너뛰기




Volumn 17, Issue 3, 2017, Pages 160-183

A chemical probe toolbox for dissecting the cancer epigenome

Author keywords

[No Author keywords available]

Indexed keywords

ACETYLLYSINE RECOGNITION DOMAIN INHIBITOR; DNA; DNA METHYLTRANSFERASE INHIBITOR; FLAVINE ADENINE NUCLEOTIDE; HISTONE ACETYLTRANSFERASE INHIBITOR; HISTONE DEMETHYLASE; HISTONE DEMETHYLASE INHIBITOR; HISTONE METHYLTRANSFERASE INHIBITOR; METHYLLYSINE RECOGNITION DOMAIN INHIBITOR; METHYLTRANSFERASE INHIBITOR; UNCLASSIFIED DRUG; EZH2 PROTEIN, HUMAN; HISTONE ACETYLTRANSFERASE; HISTONE LYSINE METHYLTRANSFERASE; HISTONE METHYLTRANSFERASE; SIRTUIN; TRANSCRIPTION FACTOR EZH2;

EID: 85013638949     PISSN: 1474175X     EISSN: 14741768     Source Type: Journal    
DOI: 10.1038/nrc.2016.148     Document Type: Review
Times cited : (76)

References (302)
  • 1
    • 33847076849 scopus 로고    scopus 로고
    • Chromatin modifications and their function
    • Kouzarides, T. Chromatin modifications and their function. Cell 128, 693-705 (2007).
    • (2007) Cell , vol.128 , pp. 693-705
    • Kouzarides, T.1
  • 2
    • 75749101495 scopus 로고    scopus 로고
    • Chromatin remodelling during development
    • Ho, L., & Crabtree, G. R. Chromatin remodelling during development. Nature 463, 474-484 (2010).
    • (2010) Nature , vol.463 , pp. 474-484
    • Ho, L.1    Crabtree, G.R.2
  • 4
    • 1542515338 scopus 로고    scopus 로고
    • A census of human cancer genes
    • Futreal, P. A., et al. A census of human cancer genes. Nat. Rev. Cancer 4, 177-183 (2004).
    • (2004) Nat. Rev. Cancer , vol.4 , pp. 177-183
    • Futreal, P.A.1
  • 5
    • 84863393263 scopus 로고    scopus 로고
    • Prognostic relevance of integrated genetic profiling in acute myeloid leukemia
    • Patel, J. P., et al. Prognostic relevance of integrated genetic profiling in acute myeloid leukemia. N. Engl. J. Med. 366, 1079-1089 (2012).
    • (2012) N. Engl. J. Med. , vol.366 , pp. 1079-1089
    • Patel, J.P.1
  • 6
    • 84866378702 scopus 로고    scopus 로고
    • The role of mutations in epigenetic regulators in myeloid malignancies
    • Shih, A. H., Abdel-Wahab, O., Patel, J. P., & Levine, R. L. The role of mutations in epigenetic regulators in myeloid malignancies. Nat. Rev. Cancer 12, 599-612 (2012).
    • (2012) Nat. Rev. Cancer , vol.12 , pp. 599-612
    • Shih, A.H.1    Abdel-Wahab, O.2    Patel, J.P.3    Levine, R.L.4
  • 7
    • 84890717859 scopus 로고    scopus 로고
    • Role of SWI/SNF in acute leukemia maintenance and enhancer-mediated Myc regulation
    • Shi, J., et al. Role of SWI/SNF in acute leukemia maintenance and enhancer-mediated Myc regulation. Genes Dev. 27, 2648-2662 (2013).
    • (2013) Genes Dev. , vol.27 , pp. 2648-2662
    • Shi, J.1
  • 8
    • 60849115270 scopus 로고    scopus 로고
    • IDH1 and IDH2 mutations in gliomas
    • Yan, H., et al. IDH1 and IDH2 mutations in gliomas. N. Engl. J. Med. 360, 765-773 (2009).
    • (2009) N. Engl. J. Med. , vol.360 , pp. 765-773
    • Yan, H.1
  • 9
    • 55949136562 scopus 로고    scopus 로고
    • Roles of the EZH2 histone methyltransferase in cancer epigenetics
    • Simon, J. A., & Lange, C. A. Roles of the EZH2 histone methyltransferase in cancer epigenetics. Mutat. Res. 647, 21-29 (2008).
    • (2008) Mutat. Res. , vol.647 , pp. 21-29
    • Simon, J.A.1    Lange, C.A.2
  • 10
    • 75749124332 scopus 로고    scopus 로고
    • Somatic mutations altering EZH2 (Tyr641) in follicular and diffuse large B cell lymphomas of germinal-center origin
    • Morin, R. D., et al. Somatic mutations altering EZH2 (Tyr641) in follicular and diffuse large B cell lymphomas of germinal-center origin. Nat. Genet. 42, 181-185 (2010).
    • (2010) Nat. Genet. , vol.42 , pp. 181-185
    • Morin, R.D.1
  • 11
    • 67349203626 scopus 로고    scopus 로고
    • Somatic mutations of the histone H3K27 demethylase gene UTX in human cancer
    • van Haaften, G., et al. Somatic mutations of the histone H3K27 demethylase gene UTX in human cancer. Nat. Genet. 41, 521-523 (2009).
    • (2009) Nat. Genet. , vol.41 , pp. 521-523
    • Van Haaften, G.1
  • 12
    • 79959653996 scopus 로고    scopus 로고
    • SWI/SNF nucleosome remodellers and cancer
    • Wilson, B. G., & Roberts, C. W. SWI/SNF nucleosome remodellers and cancer. Nat. Rev. Cancer 11, 481-492 (2011).
    • (2011) Nat. Rev. Cancer , vol.11 , pp. 481-492
    • Wilson, B.G.1    Roberts, C.W.2
  • 13
    • 84878745222 scopus 로고    scopus 로고
    • Proteomic and bioinformatic analysis of mammalian SWI/SNF complexes identifies extensive roles in human malignancy
    • Kadoch, C., et al. Proteomic and bioinformatic analysis of mammalian SWI/SNF complexes identifies extensive roles in human malignancy. Nat. Genet. 45, 592-601 (2013).
    • (2013) Nat. Genet. , vol.45 , pp. 592-601
    • Kadoch, C.1
  • 14
    • 2642647094 scopus 로고    scopus 로고
    • Truncating mutations of hSNF5/INI1 in aggressive paediatric cancer
    • Versteege, I., et al. Truncating mutations of hSNF5/INI1 in aggressive paediatric cancer. Nature 394, 203-206 (1998).
    • (1998) Nature , vol.394 , pp. 203-206
    • Versteege, I.1
  • 15
    • 77957731999 scopus 로고    scopus 로고
    • Frequent mutations of chromatin remodeling gene ARID1A in ovarian clear cell carcinoma
    • Jones, S., et al. Frequent mutations of chromatin remodeling gene ARID1A in ovarian clear cell carcinoma. Science 330, 228-231 (2010).
    • (2010) Science , vol.330 , pp. 228-231
    • Jones, S.1
  • 16
    • 77957946398 scopus 로고    scopus 로고
    • ARID1A mutations in endometriosis-Associated ovarian carcinomas
    • Wiegand, K. C., et al. ARID1A mutations in endometriosis-Associated ovarian carcinomas. N. Engl. J. Med. 363, 1532-1543 (2010).
    • (2010) N. Engl. J. Med. , vol.363 , pp. 1532-1543
    • Wiegand, K.C.1
  • 17
    • 84863621527 scopus 로고    scopus 로고
    • Cancer epigenetics: From mechanism to therapy
    • Dawson, M. A., & Kouzarides, T. Cancer epigenetics: from mechanism to therapy. Cell 150, 12-27 (2012).
    • (2012) Cell , vol.150 , pp. 12-27
    • Dawson, M.A.1    Kouzarides, T.2
  • 18
    • 84875745673 scopus 로고    scopus 로고
    • Interplay between the cancer genome and epigenome
    • Shen, H., & Laird, P. W. Interplay between the cancer genome and epigenome. Cell 153, 38-55 (2013).
    • (2013) Cell , vol.153 , pp. 38-55
    • Shen, H.1    Laird, P.W.2
  • 19
    • 50249186849 scopus 로고    scopus 로고
    • Inhibition of transcription factors with small organic molecules
    • Berg, T. Inhibition of transcription factors with small organic molecules. Curr. Opin. Chem. Biol. 12, 464-471 (2008).
    • (2008) Curr. Opin. Chem. Biol. , vol.12 , pp. 464-471
    • Berg, T.1
  • 20
    • 0036782706 scopus 로고    scopus 로고
    • Transcription factors as targets for cancer therapy
    • Darnell, J. E. Jr. Transcription factors as targets for cancer therapy. Nat. Rev. Cancer 2, 740-749 (2002).
    • (2002) Nat. Rev. Cancer , vol.2 , pp. 740-749
    • Darnell, J.E.1
  • 21
    • 77952545553 scopus 로고    scopus 로고
    • A complex task? Direct modulation of transcription factors with small molecules
    • Koehler, A. N. A complex task? Direct modulation of transcription factors with small molecules. Curr. Opin. Chem. Biol. 14, 331-340 (2010).
    • (2010) Curr. Opin. Chem. Biol. , vol.14 , pp. 331-340
    • Koehler, A.N.1
  • 22
    • 84932634729 scopus 로고    scopus 로고
    • Phthalimide conjugation as a strategy for in vivo target protein degradation
    • Winter, G. E., et al. Phthalimide conjugation as a strategy for in vivo target protein degradation. Science 348, 1376-1381 (2015).
    • (2015) Science , vol.348 , pp. 1376-1381
    • Winter, G.E.1
  • 23
    • 77249142404 scopus 로고    scopus 로고
    • The art of the chemical probe
    • Frye, S. V. The art of the chemical probe. Nat. Chem. Biol. 6, 159-161 (2010).
    • (2010) Nat. Chem. Biol. , vol.6 , pp. 159-161
    • Frye, S.V.1
  • 24
    • 85050578395 scopus 로고    scopus 로고
    • Inhibitors of emerging epigenetic targets for cancer therapy: A patent review 2010-2014
    • Tanaka, M., Roberts, J. M., Qi, J., & Bradner, J. E. Inhibitors of emerging epigenetic targets for cancer therapy: a patent review (2010-2014). Pharm. Pat. Anal. 4, 261-284 (2015).
    • (2015) Pharm. Pat. Anal. , vol.4 , pp. 261-284
    • Tanaka, M.1    Roberts, J.M.2    Qi, J.3    Bradner, J.E.4
  • 25
    • 69949148388 scopus 로고    scopus 로고
    • Protein methyltransferases as a target class for drug discovery
    • Copeland, R. A., Solomon, M. E., & Richon, V. M. Protein methyltransferases as a target class for drug discovery. Nat. Rev. Drug Discov. 8, 724-732 (2009).
    • (2009) Nat. Rev. Drug Discov. , vol.8 , pp. 724-732
    • Copeland, R.A.1    Solomon, M.E.2    Richon, V.M.3
  • 26
    • 59349115177 scopus 로고    scopus 로고
    • Chemical mechanisms of histone lysine and arginine modifications
    • Smith, B. C., & Denu, J. M. Chemical mechanisms of histone lysine and arginine modifications. Biochim. Biophys. Acta 1789, 45-57 (2009).
    • (2009) Biochim. Biophys. Acta , vol.1789 , pp. 45-57
    • Smith, B.C.1    Denu, J.M.2
  • 27
    • 84858636152 scopus 로고    scopus 로고
    • Current chemical biology approaches to interrogate protein methyltransferases
    • Luo, M. Current chemical biology approaches to interrogate protein methyltransferases. ACS Chem. Biol. 7, 443-463 (2012).
    • (2012) ACS Chem. Biol. , vol.7 , pp. 443-463
    • Luo, M.1
  • 28
    • 17444375685 scopus 로고    scopus 로고
    • HDOT1L links histone methylation to leukemogenesis
    • Okada, Y., et al. hDOT1L links histone methylation to leukemogenesis. Cell 121, 167-178 (2005).
    • (2005) Cell , vol.121 , pp. 167-178
    • Okada, Y.1
  • 29
    • 0018122025 scopus 로고
    • The antifungal antibiotic sinefungin as a very active inhibitor of methyltransferases and of the transformation of chick embryo fibroblasts by Rous sarcoma virus
    • Vedel, M., Lawrence, F., Robert-Gero, M., & Lederer, E. The antifungal antibiotic sinefungin as a very active inhibitor of methyltransferases and of the transformation of chick embryo fibroblasts by Rous sarcoma virus. Biochem. Biophys. Res. Commun. 85, 371-376 (1978).
    • (1978) Biochem. Biophys. Res. Commun. , vol.85 , pp. 371-376
    • Vedel, M.1    Lawrence, F.2    Robert-Gero, M.3    Lederer, E.4
  • 30
    • 30644474460 scopus 로고    scopus 로고
    • Identification of a specific inhibitor of the histone methyltransferase SU(VAR)
    • Greiner, D., Bonaldi, T., Eskeland, R., Roemer, E., & Imhof, A. Identification of a specific inhibitor of the histone methyltransferase SU(VAR)3 9. Nat. Chem. Biol. 1, 143-145 (2005).
    • (2005) Nat. Chem. Biol. 1 , vol.3 , Issue.9 , pp. 143-145
    • Greiner, D.1    Bonaldi, T.2    Eskeland, R.3    Roemer, E.4    Imhof, A.5
  • 31
    • 84874041667 scopus 로고    scopus 로고
    • Chaetocin is a nonspecific inhibitor of histone lysine methyltransferases
    • Cherblanc, F. L., Chapman, K. L., Brown, R., & Fuchter, M. J. Chaetocin is a nonspecific inhibitor of histone lysine methyltransferases. Nat. Chem. Biol. 9, 136-137 (2013).
    • (2013) Nat. Chem. Biol. , vol.9 , pp. 136-137
    • Cherblanc, F.L.1    Chapman, K.L.2    Brown, R.3    Fuchter, M.J.4
  • 32
    • 0021194198 scopus 로고
    • Effects of adenosine dialdehyde on S adenosylhomocysteine hydrolase and S adenosylmethionine-dependent transmethylations in mouse L929 cells
    • Bartel, R. L., & Borchardt, R. T. Effects of adenosine dialdehyde on S adenosylhomocysteine hydrolase and S adenosylmethionine-dependent transmethylations in mouse L929 cells. Mol. Pharmacol. 25, 418-424 (1984).
    • (1984) Mol. Pharmacol. , vol.25 , pp. 418-424
    • Bartel, R.L.1    Borchardt, R.T.2
  • 33
    • 0025975888 scopus 로고
    • 4, 5. Unsaturated 5. Halogenated nucleosides. Mechanism-based and competitive inhibitors of S adenosyl-l homocysteine hydrolase
    • Jarvi, E. T., et al. 4, 5. unsaturated 5. halogenated nucleosides. Mechanism-based and competitive inhibitors of S adenosyl-l homocysteine hydrolase. J. Med. Chem. 34, 647-656 (1991).
    • (1991) J. Med. Chem. , vol.34 , pp. 647-656
    • Jarvi, E.T.1
  • 34
    • 34248645046 scopus 로고    scopus 로고
    • eds Clarke, S. G., & Tamanoi, F. Elsevier
    • Clarke, S. G. in The Enzymes: Protein Methyltransferases Vol. 24 (eds Clarke, S. G., & Tamanoi, F.) 467-493 (Elsevier, 2006).
    • (2006) The Enzymes: Protein Methyltransferases , vol.24 , pp. 467-493
    • Clarke, S.G.1
  • 35
    • 67649371461 scopus 로고    scopus 로고
    • DZNep is a global histone methylation inhibitor that reactivates developmental genes not silenced by DNA methylation
    • Miranda, T. B., et al. DZNep is a global histone methylation inhibitor that reactivates developmental genes not silenced by DNA methylation. Mol. Cancer Ther. 8, 1579-1588 (2009).
    • (2009) Mol. Cancer Ther. , vol.8 , pp. 1579-1588
    • Miranda, T.B.1
  • 36
    • 0027131996 scopus 로고
    • Z-4' 5' didehydro 5' Deoxy 5' fluoroadenosine (MDL 28 842), an irreversible inhibitor of S adenosylhomocysteine hydrolase, suppresses proliferation of cultured keratinocytes and squamous carcinoma cell lines
    • Paller, A. S., Arnsmeier, S. L., Clark, S. H., & Mirkin, B. L. Z-4?, 5? didehydro 5? deoxy 5? fluoroadenosine (MDL 28, 842), an irreversible inhibitor of S adenosylhomocysteine hydrolase, suppresses proliferation of cultured keratinocytes and squamous carcinoma cell lines. Cancer Res. 53, 6058-6060 (1993).
    • (1993) Cancer Res. , vol.53 , pp. 6058-6060
    • Paller, A.S.1    Arnsmeier, S.L.2    Clark, S.H.3    Mirkin, B.L.4
  • 37
    • 34247625135 scopus 로고    scopus 로고
    • Pharmacologic disruption of Polycomb-repressive complex 2 mediated gene repression selectively induces apoptosis in cancer cells
    • Tan, J., et al. Pharmacologic disruption of Polycomb-repressive complex 2 mediated gene repression selectively induces apoptosis in cancer cells. Genes Dev. 21, 1050-1063 (2007).
    • (2007) Genes Dev. , vol.21 , pp. 1050-1063
    • Tan, J.1
  • 38
    • 0037099413 scopus 로고    scopus 로고
    • G9a histone methyltransferase plays a dominant role in euchromatic histone H3 lysine 9 methylation and is essential for early embryogenesis
    • Tachibana, M., et al. G9a histone methyltransferase plays a dominant role in euchromatic histone H3 lysine 9 methylation and is essential for early embryogenesis. Genes Dev. 16, 1779-1791 (2002).
    • (2002) Genes Dev. , vol.16 , pp. 1779-1791
    • Tachibana, M.1
  • 39
    • 33645751058 scopus 로고    scopus 로고
    • Silenced tumor suppressor genes reactivated by DNA demethylation do not return to a fully euchromatic chromatin state
    • McGarvey, K. M., et al. Silenced tumor suppressor genes reactivated by DNA demethylation do not return to a fully euchromatic chromatin state. Cancer Res. 66, 3541-3549 (2006).
    • (2006) Cancer Res. , vol.66 , pp. 3541-3549
    • McGarvey, K.M.1
  • 40
    • 44249128891 scopus 로고    scopus 로고
    • Downregulation of histone H3 lysine 9 methyltransferase G9a induces centrosome disruption and chromosome instability in cancer cells
    • Kondo, Y., et al. Downregulation of histone H3 lysine 9 methyltransferase G9a induces centrosome disruption and chromosome instability in cancer cells. PLoS ONE 3, e2037 (2008).
    • (2008) PLoS ONE , vol.3 , pp. e2037
    • Kondo, Y.1
  • 41
    • 84889684498 scopus 로고    scopus 로고
    • The histone H3 methyltransferase G9A epigenetically activates the serine-glycine synthesis pathway to sustain cancer cell survival and proliferation
    • Ding, J., et al. The histone H3 methyltransferase G9A epigenetically activates the serine-glycine synthesis pathway to sustain cancer cell survival and proliferation. Cell Metab. 18, 896-907 (2013).
    • (2013) Cell Metab. , vol.18 , pp. 896-907
    • Ding, J.1
  • 42
    • 33846783261 scopus 로고    scopus 로고
    • Reversal of H3K9me2 by a small-molecule inhibitor for the G9a histone methyltransferase
    • Kubicek, S., et al. Reversal of H3K9me2 by a small-molecule inhibitor for the G9a histone methyltransferase. Mol. Cell 25, 473-481 (2007).
    • (2007) Mol. Cell , vol.25 , pp. 473-481
    • Kubicek, S.1
  • 43
    • 62049083789 scopus 로고    scopus 로고
    • Structural basis for G9a like protein lysine methyltransferase inhibition by BIX 01294
    • Chang, Y., et al. Structural basis for G9a like protein lysine methyltransferase inhibition by BIX 01294. Nat. Struct. Mol. Biol. 16, 312-317 (2009).
    • (2009) Nat. Struct. Mol. Biol. , vol.16 , pp. 312-317
    • Chang, Y.1
  • 44
    • 73249124141 scopus 로고    scopus 로고
    • Discovery of a 2, 4 diamino 7 aminoalkoxyquinazoline as a potent and selective inhibitor of histone lysine methyltransferase G9a
    • Liu, F., et al. Discovery of a 2, 4 diamino 7 aminoalkoxyquinazoline as a potent and selective inhibitor of histone lysine methyltransferase G9a. J. Med. Chem. 52, 7950-7953 (2009).
    • (2009) J. Med. Chem. , vol.52 , pp. 7950-7953
    • Liu, F.1
  • 45
    • 77955363182 scopus 로고    scopus 로고
    • Protein lysine methyltransferase G9a inhibitors: Design, syn thesis, and structure activity relationships of 2, 4 diamino 7 aminoalkoxy-quinazolines
    • Liu, F., et al. Protein lysine methyltransferase G9a inhibitors: design, synthesis, and structure activity relationships of 2, 4 diamino 7 aminoalkoxy-quinazolines. J. Med. Chem. 53, 5844-5857 (2010).
    • (2010) J. Med. Chem. , vol.53 , pp. 5844-5857
    • Liu, F.1
  • 46
    • 80052375547 scopus 로고    scopus 로고
    • Optimization of cellular activity of G9a inhibitors 7 aminoalkoxy-quinazolines
    • Liu, F., et al. Optimization of cellular activity of G9a inhibitors 7 aminoalkoxy-quinazolines. J. Med. Chem. 54, 6139-6150 (2011).
    • (2011) J. Med. Chem. , vol.54 , pp. 6139-6150
    • Liu, F.1
  • 47
    • 79960493567 scopus 로고    scopus 로고
    • A chemical probe selectively inhibits G9a and GLP methyltransferase activity in cells
    • Vedadi, M., et al. A chemical probe selectively inhibits G9a and GLP methyltransferase activity in cells. Nat. Chem. Biol. 7, 566-574 (2011).
    • (2011) Nat. Chem. Biol. , vol.7 , pp. 566-574
    • Vedadi, M.1
  • 48
    • 84887923349 scopus 로고    scopus 로고
    • Discovery of an in vivo chemical probe of the lysine methyltransferases G9a and GLP
    • Liu, F., et al. Discovery of an in vivo chemical probe of the lysine methyltransferases G9a and GLP. J. Med. Chem. 56, 8931-8942 (2013).
    • (2013) J. Med. Chem. , vol.56 , pp. 8931-8942
    • Liu, F.1
  • 49
    • 84893972052 scopus 로고    scopus 로고
    • The methyltransferase G9a regulates HoxA9 dependent transcription in AML
    • Lehnertz, B., et al. The methyltransferase G9a regulates HoxA9 dependent transcription in AML. Genes Dev. 28, 317-327 (2014).
    • (2014) Genes Dev. , vol.28 , pp. 317-327
    • Lehnertz, B.1
  • 50
    • 84869162797 scopus 로고    scopus 로고
    • G9a/GLP-dependent histone H3K9me2 patterning during human hematopoietic stem cell lineage commitment
    • Chen, X., et al. G9a/GLP-dependent histone H3K9me2 patterning during human hematopoietic stem cell lineage commitment. Genes Dev. 26, 2499-2511 (2012).
    • (2012) Genes Dev. , vol.26 , pp. 2499-2511
    • Chen, X.1
  • 51
    • 84894065529 scopus 로고    scopus 로고
    • Discovery and development of potent and selective inhibitors of histone methyltransferase G9a
    • Sweis, R. F., et al. Discovery and development of potent and selective inhibitors of histone methyltransferase G9a. ACS Med. Chem. Lett. 5, 205-209 (2014).
    • (2014) ACS Med. Chem. Lett. , vol.5 , pp. 205-209
    • Sweis, R.F.1
  • 52
    • 84940093455 scopus 로고    scopus 로고
    • The histone methyltransferase inhibitor A-366 uncovers a role for G9a/GLP in the epigenetics of leukemia
    • Pappano, W. N., et al. The histone methyltransferase inhibitor A-366 uncovers a role for G9a/GLP in the epigenetics of leukemia. PLoS ONE 10, e0131716 (2015).
    • (2015) PLoS ONE , vol.10 , pp. e0131716
    • Pappano, W.N.1
  • 53
    • 84864140929 scopus 로고    scopus 로고
    • A small-molecule probe of the histone methyltransferase G9a induces cellular senescence in pancreatic adenocarcinoma
    • Yuan, Y., et al. A small-molecule probe of the histone methyltransferase G9a induces cellular senescence in pancreatic adenocarcinoma. ACS Chem. Biol. 7, 1152-1157 (2012).
    • (2012) ACS Chem. Biol. , vol.7 , pp. 1152-1157
    • Yuan, Y.1
  • 54
    • 79960044951 scopus 로고    scopus 로고
    • MLL-rearranged leukemia is dependent on aberrant H3K79 methylation by DOT1L
    • Bernt, K. M., et al. MLL-rearranged leukemia is dependent on aberrant H3K79 methylation by DOT1L. Cancer Cell 20, 66-78 (2011).
    • (2011) Cancer Cell , vol.20 , pp. 66-78
    • Bernt, K.M.1
  • 55
    • 84993940217 scopus 로고    scopus 로고
    • DOT1L as a therapeutic target for the treatment of DNMT3A mutant acute myeloid leukemia
    • Rau, R. E., et al. DOT1L as a therapeutic target for the treatment of DNMT3A mutant acute myeloid leukemia. Blood 128, 971-981 (2016).
    • (2016) Blood , vol.128 , pp. 971-981
    • Rau, R.E.1
  • 56
    • 0344837759 scopus 로고    scopus 로고
    • Structure of the catalytic domain of human DOT1L, a non-SET domain nucleosomal histone methyltransferase
    • Min, J., Feng, Q., Li, Z., Zhang, Y., & Xu, R. M. Structure of the catalytic domain of human DOT1L, a non-SET domain nucleosomal histone methyltransferase. Cell 112, 711-723 (2003).
    • (2003) Cell , vol.112 , pp. 711-723
    • Min, J.1    Feng, Q.2    Li, Z.3    Zhang, Y.4    Xu, R.M.5
  • 57
    • 79960367903 scopus 로고    scopus 로고
    • Chemogenetic analysis of human protein methyltransferases
    • Richon, V. M., et al. Chemogenetic analysis of human protein methyltransferases. Chem. Biol. Drug Des. 78, 199-210 (2011).
    • (2011) Chem. Biol. Drug Des. , vol.78 , pp. 199-210
    • Richon, V.M.1
  • 58
    • 79960058024 scopus 로고    scopus 로고
    • Selective killing of mixed lineage leukemia cells by a potent small-molecule DOT1L inhibitor
    • Daigle, S. R., et al. Selective killing of mixed lineage leukemia cells by a potent small-molecule DOT1L inhibitor. Cancer Cell 20, 53-65 (2011).
    • (2011) Cancer Cell , vol.20 , pp. 53-65
    • Daigle, S.R.1
  • 59
    • 84871737742 scopus 로고    scopus 로고
    • Catalytic site remodelling of the DOT1L methyltransferase by selective inhibitors
    • Yu, W., et al. Catalytic site remodelling of the DOT1L methyltransferase by selective inhibitors. Nat. Commun. 3, 1288 (2012).
    • (2012) Nat. Commun. , vol.3 , pp. 1288
    • Yu, W.1
  • 60
    • 84868101932 scopus 로고    scopus 로고
    • Conformational adaptation drives potent, selective and durable inhibition of the human protein methyltransferase DOT1L
    • Basavapathruni, A., et al. Conformational adaptation drives potent, selective and durable inhibition of the human protein methyltransferase DOT1L. Chem. Biol. Drug Des. 80, 971-980 (2012).
    • (2012) Chem. Biol. Drug Des. , vol.80 , pp. 971-980
    • Basavapathruni, A.1
  • 61
    • 84886864184 scopus 로고    scopus 로고
    • Potent inhibition of DOT1L as treatment of MLL-fusion leukemia
    • Daigle, S. R., et al. Potent inhibition of DOT1L as treatment of MLL-fusion leukemia. Blood 122, 1017-1025 (2013).
    • (2013) Blood , vol.122 , pp. 1017-1025
    • Daigle, S.R.1
  • 62
    • 80055019988 scopus 로고    scopus 로고
    • Selective inhibitors of histone methyltransferase DOT1L: Design, syn
    • thesis, and crystallographic studies
    • Yao, Y., et al. Selective inhibitors of histone methyltransferase DOT1L: design, synthesis, and crystallographic studies. J. Am. Chem. Soc. 133, 16746-16749 (2011).
    • (2011) J. Am. Chem. Soc. , vol.133 , pp. 16746-16749
    • Yao, Y.1
  • 63
    • 84875217644 scopus 로고    scopus 로고
    • Bromo-deaza-SAH: A potent and selective DOT1L inhibitor
    • Yu, W., et al. Bromo-deaza-SAH: a potent and selective DOT1L inhibitor. Bioorg. Med. Chem. 21, 1787-1794 (2013).
    • (2013) Bioorg. Med. Chem. , vol.21 , pp. 1787-1794
    • Yu, W.1
  • 64
    • 84877312453 scopus 로고    scopus 로고
    • Activity and metabolic stability of non-ribose containing inhibitors of histone methyltransferase DOT1L
    • thesis
    • Deng, L., et al. Synthesis, activity and metabolic stability of non-ribose containing inhibitors of histone methyltransferase DOT1L. MedChemComm. 4, 822-826 (2013).
    • (2013) MedChemComm. , vol.4 , pp. 822-826
    • Deng, L.1    Syn, E.2
  • 65
    • 84925620054 scopus 로고    scopus 로고
    • Structure-guided DOT1L probe optimization by label-free ligand displacement
    • Yi, J. S., et al. Structure-guided DOT1L probe optimization by label-free ligand displacement. ACS Chem. Biol. 10, 667-674 (2015).
    • (2015) ACS Chem. Biol. , vol.10 , pp. 667-674
    • Yi, J.S.1
  • 66
    • 84872531591 scopus 로고    scopus 로고
    • High-Affinity small-molecule peptidomimetic inhibitors of MLL1/WDR5 protein-protein interaction
    • Karatas, H., et al. High-Affinity, small-molecule peptidomimetic inhibitors of MLL1/WDR5 protein-protein interaction. J. Am. Chem. Soc.135, 669-682 (2013).
    • (2013) J. Am. Chem. Soc.135 , pp. 669-682
    • Karatas, H.1
  • 67
    • 84892841527 scopus 로고    scopus 로고
    • Targeting MLL1 H3K4 methyltransferase activity in mixed-lineage leukemia
    • Cao, F., et al. Targeting MLL1 H3K4 methyltransferase activity in mixed-lineage leukemia. Mol. Cell 53, 247-261 (2014).
    • (2014) Mol. Cell , vol.53 , pp. 247-261
    • Cao, F.1
  • 68
    • 84857195696 scopus 로고    scopus 로고
    • Menin-MLL inhibitors reverse oncogenic activity of MLL fusion proteins in leukemia
    • Grembecka, J., et al. Menin-MLL inhibitors reverse oncogenic activity of MLL fusion proteins in leukemia. Nat. Chem. Biol. 8, 277-284 (2012).
    • (2012) Nat. Chem. Biol. , vol.8 , pp. 277-284
    • Grembecka, J.1
  • 69
    • 84896871890 scopus 로고    scopus 로고
    • High-Affinity small-molecule inhibitors of the menin-mixed lineage leukemia (MLL) interaction closely mimic a natural protein-protein interaction
    • He, S., et al. High-Affinity small-molecule inhibitors of the menin-mixed lineage leukemia (MLL) interaction closely mimic a natural protein-protein interaction. J. Med. Chem. 57, 1543-1556 (2014).
    • (2014) J. Med. Chem. , vol.57 , pp. 1543-1556
    • He, S.1
  • 70
    • 84870573126 scopus 로고    scopus 로고
    • EZH2 inhibition as a therapeutic strategy for lymphoma with EZH2 activating mutations
    • McCabe, M. T., et al. EZH2 inhibition as a therapeutic strategy for lymphoma with EZH2 activating mutations. Nature 492, 108-112 (2012).
    • (2012) Nature , vol.492 , pp. 108-112
    • McCabe, M.T.1
  • 71
    • 84867632489 scopus 로고    scopus 로고
    • A selective inhibitor of EZH2 blocks H3K27 methylation and kills mutant lymphoma cells
    • Knutson, S. K., et al. A selective inhibitor of EZH2 blocks H3K27 methylation and kills mutant lymphoma cells. Nat. Chem. Biol. 8, 890-896 (2012).
    • (2012) Nat. Chem. Biol. , vol.8 , pp. 890-896
    • Knutson, S.K.1
  • 72
    • 84871841675 scopus 로고    scopus 로고
    • Selective inhibition of Ezh2 by a small molecule inhibitor blocks tumor cells proliferation
    • Qi, W., et al. Selective inhibition of Ezh2 by a small molecule inhibitor blocks tumor cells proliferation. Proc. Natl Acad. Sci. USA 109, 21360-21365 (2012).
    • (2012) Proc. Natl Acad. Sci. USA , vol.109 , pp. 21360-21365
    • Qi, W.1
  • 73
    • 84905572005 scopus 로고    scopus 로고
    • Gene silencing triggers polycomb repressive complex 2 recruitment to CpG islands genome wide
    • Riising, E. M., et al. Gene silencing triggers polycomb repressive complex 2 recruitment to CpG islands genome wide. Mol. Cell 55, 347-360 (2014).
    • (2014) Mol. Cell , vol.55 , pp. 347-360
    • Riising, E.M.1
  • 74
    • 55949132133 scopus 로고    scopus 로고
    • Ezh1 and Ezh2 maintain repressive chromatin through different mechanisms
    • Margueron, R., et al. Ezh1 and Ezh2 maintain repressive chromatin through different mechanisms. Mol. Cell 32, 503-518 (2008).
    • (2008) Mol. Cell , vol.32 , pp. 503-518
    • Margueron, R.1
  • 75
    • 18644382388 scopus 로고    scopus 로고
    • The polycomb group protein EZH2 is involved in progression of prostate cancer
    • Varambally, S., et al. The polycomb group protein EZH2 is involved in progression of prostate cancer. Nature 419, 624-629 (2002).
    • (2002) Nature , vol.419 , pp. 624-629
    • Varambally, S.1
  • 76
    • 0141816752 scopus 로고    scopus 로고
    • EZH2 is a marker of aggressive breast cancer and promotes neoplastic transformation of breast epithelial cells
    • Kleer, C. G., et al. EZH2 is a marker of aggressive breast cancer and promotes neoplastic transformation of breast epithelial cells. Proc. Natl Acad. Sci. USA 100, 11606-11611 (2003).
    • (2003) Proc. Natl Acad. Sci. USA , vol.100 , pp. 11606-11611
    • Kleer, C.G.1
  • 77
    • 80052029516 scopus 로고    scopus 로고
    • Frequent mutation of histone-modifying genes in non-Hodgkin lymphoma
    • Morin, R. D., et al. Frequent mutation of histone-modifying genes in non-Hodgkin lymphoma. Nature 476, 298-303 (2011).
    • (2011) Nature , vol.476 , pp. 298-303
    • Morin, R.D.1
  • 78
    • 78650062951 scopus 로고    scopus 로고
    • EZH2 mediated epigenetic silencing in germinal center B cells contributes to proliferation and lymphomagenesis
    • Velichutina, I., et al. EZH2 mediated epigenetic silencing in germinal center B cells contributes to proliferation and lymphomagenesis. Blood 116, 5247-5255 (2010).
    • (2010) Blood , vol.116 , pp. 5247-5255
    • Velichutina, I.1
  • 79
    • 84877815031 scopus 로고    scopus 로고
    • EZH2 is required for germinal center formation and somatic EZH2 mutations promote lymphoid transformation
    • Béguelin, W., et al. EZH2 is required for germinal center formation and somatic EZH2 mutations promote lymphoid transformation. Cancer Cell 23, 677-692 (2013).
    • (2013) Cancer Cell , vol.23 , pp. 677-692
    • Béguelin, W.1
  • 80
    • 84946227498 scopus 로고    scopus 로고
    • Loss of BAP1 function leads to EZH2 dependent transformation
    • LaFave, L. M., et al. Loss of BAP1 function leads to EZH2 dependent transformation. Nat. Med. 21, 1344-1349 (2015).
    • (2015) Nat. Med. , vol.21 , pp. 1344-1349
    • LaFave, L.M.1
  • 81
    • 84888303259 scopus 로고    scopus 로고
    • Identification of EZH2 and EZH1 small molecule inhibitors with selective impact on diffuse large B cell lymphoma cell growth
    • Garapaty-Rao, S., et al. Identification of EZH2 and EZH1 small molecule inhibitors with selective impact on diffuse large B cell lymphoma cell growth. Chem. Biol. 20, 1329-1339 (2013).
    • (2013) Chem. Biol. , vol.20 , pp. 1329-1339
    • Garapaty-Rao, S.1
  • 82
    • 84898034702 scopus 로고    scopus 로고
    • Discovery and optimization of tetramethylpiperidinyl benzamides as inhibitors of EZH2
    • Nasveschuk, C. G., et al. Discovery and optimization of tetramethylpiperidinyl benzamides as inhibitors of EZH2. ACS Med. Chem. Lett. 5, 378-383 (2014).
    • (2014) ACS Med. Chem. Lett. , vol.5 , pp. 378-383
    • Nasveschuk, C.G.1
  • 83
    • 84879750981 scopus 로고    scopus 로고
    • An orally bioavailable chemical probe of the lysine methyltransferases EZH2 and EZH1
    • Konze, K. D., et al. An orally bioavailable chemical probe of the lysine methyltransferases EZH2 and EZH1. ACS Chem. Biol. 8, 1324-1334 (2013).
    • (2013) ACS Chem. Biol. , vol.8 , pp. 1324-1334
    • Konze, K.D.1
  • 84
    • 84877324084 scopus 로고    scopus 로고
    • Durable tumor regression in genetically altered malignant rhabdoid tumors by inhibition of methyltransferase EZH2
    • Knutson, S. K., et al. Durable tumor regression in genetically altered malignant rhabdoid tumors by inhibition of methyltransferase EZH2. Proc. Natl Acad. Sci. USA 110, 7922-7927 (2013).
    • (2013) Proc. Natl Acad. Sci. USA , vol.110 , pp. 7922-7927
    • Knutson, S.K.1
  • 85
    • 84871248432 scopus 로고    scopus 로고
    • Identification of potent, selective, cell-Active inhibitors of the histone lysine methyltransferase EZH2
    • Verma, S. K., et al. Identification of potent, selective, cell-Active inhibitors of the histone lysine methyltransferase EZH2. ACS Med. Chem. Lett. 3, 1091-1096 (2012).
    • (2012) ACS Med. Chem. Lett. , vol.3 , pp. 1091-1096
    • Verma, S.K.1
  • 86
    • 84921318899 scopus 로고    scopus 로고
    • Selective inhibition of EZH2 by EPZ 6438 leads to potent antitumor activity in EZH2 mutant non-Hodgkin lymphoma
    • Knutson, S. K., et al. Selective inhibition of EZH2 by EPZ 6438 leads to potent antitumor activity in EZH2 mutant non-Hodgkin lymphoma. Mol. Cancer Ther. 13, 842-854 (2014).
    • (2014) Mol. Cancer Ther. , vol.13 , pp. 842-854
    • Knutson, S.K.1
  • 87
    • 84875715709 scopus 로고    scopus 로고
    • Inhibition of EZH2 suppresses self-renewal and induces radiation sensitivity in atypical rhabdoid teratoid tumor cells
    • Alimova, I., et al. Inhibition of EZH2 suppresses self-renewal and induces radiation sensitivity in atypical rhabdoid teratoid tumor cells. Neuro Oncol. 15, 149-160 (2013).
    • (2013) Neuro Oncol. , vol.15 , pp. 149-160
    • Alimova, I.1
  • 88
    • 84874106911 scopus 로고    scopus 로고
    • The Polycomb complex PRC2 supports aberrant self-renewal in a mouse model of MLL AF9;Nras(G12D) acute myeloid leukemia
    • Shi, J., et al. The Polycomb complex PRC2 supports aberrant self-renewal in a mouse model of MLL AF9;Nras(G12D) acute myeloid leukemia. Oncogene 32, 930-938 (2013).
    • (2013) Oncogene , vol.32 , pp. 930-938
    • Shi, J.1
  • 89
    • 84859475727 scopus 로고    scopus 로고
    • Polycomb repressive complex 2 is required for MLL AF9 leukemia
    • Neff, T., et al. Polycomb repressive complex 2 is required for MLL AF9 leukemia. Proc. Natl Acad. Sci. USA 109, 5028-5033 (2012).
    • (2012) Proc. Natl Acad. Sci. USA , vol.109 , pp. 5028-5033
    • Neff, T.1
  • 90
    • 84939270225 scopus 로고    scopus 로고
    • Targeting EZH2 and PRC2 dependence as novel anticancer therapy
    • Xu, B., Konze, K. D., Jin, J., & Wang, G. G. Targeting EZH2 and PRC2 dependence as novel anticancer therapy. Exp. Hematol. 43, 698-712 (2015).
    • (2015) Exp. Hematol. , vol.43 , pp. 698-712
    • Xu, B.1    Konze, K.D.2    Jin, J.3    Wang, G.G.4
  • 91
    • 84945186017 scopus 로고    scopus 로고
    • Structural basis of histone H3K27 trimethylation by an active polycomb repressive complex 2
    • aac4383
    • Jiao, L., & Liu, X. Structural basis of histone H3K27 trimethylation by an active polycomb repressive complex 2. Science 350, aac4383 (2015).
    • (2015) Science , pp. 350
    • Jiao, L.1    Liu, X.2
  • 92
    • 0035854372 scopus 로고    scopus 로고
    • State of the arg: Protein methylation at arginine comes of age
    • McBride, A. E., & Silver, P. A. State of the arg: protein methylation at arginine comes of age. Cell 106, 5-8 (2001).
    • (2001) Cell , vol.106 , pp. 5-8
    • McBride, A.E.1    Silver, P.A.2
  • 93
    • 84871712509 scopus 로고    scopus 로고
    • Protein arginine methyltransferases and cancer
    • Yang, Y., & Bedford, M. T. Protein arginine methyltransferases and cancer. Nat. Rev. Cancer 13, 37-50 (2013).
    • (2013) Nat. Rev. Cancer , vol.13 , pp. 37-50
    • Yang, Y.1    Bedford, M.T.2
  • 94
    • 84948410742 scopus 로고    scopus 로고
    • The dual epigenetic role of PRMT5 in acute myeloid leukemia: Gene activation and repression via histone arginine methylation
    • Tarighat, S. S., et al. The dual epigenetic role of PRMT5 in acute myeloid leukemia: gene activation and repression via histone arginine methylation. Leukemia 30, 789-799 (2016).
    • (2016) Leukemia , vol.30 , pp. 789-799
    • Tarighat, S.S.1
  • 95
    • 84890288945 scopus 로고    scopus 로고
    • Protein arginine methyltransferase 5 (PRMT5) inhibition induces lymphoma cell death through reactivation of the retinoblastoma tumor suppressor pathway and polycomb repressor complex 2 (PRC2) silencing
    • Chung, J., et al. Protein arginine methyltransferase 5 (PRMT5) inhibition induces lymphoma cell death through reactivation of the retinoblastoma tumor suppressor pathway and polycomb repressor complex 2 (PRC2) silencing. J. Biol. Chem. 288, 35534-35547 (2013).
    • (2013) J. Biol. Chem. , vol.288 , pp. 35534-35547
    • Chung, J.1
  • 96
    • 84960916279 scopus 로고    scopus 로고
    • MTAP deletion confers enhanced dependency on the PRMT5 arginine methyltransferase in cancer cells
    • Kryukov, G. V., et al. MTAP deletion confers enhanced dependency on the PRMT5 arginine methyltransferase in cancer cells. Science 351, 1214-1218 (2016).
    • (2016) Science , vol.351 , pp. 1214-1218
    • Kryukov, G.V.1
  • 97
    • 84927747737 scopus 로고    scopus 로고
    • Selective inhibition of protein arginine methyltransferase 5 blocks initiation and maintenance of B cell transformation
    • Alinari, L., et al. Selective inhibition of protein arginine methyltransferase 5 blocks initiation and maintenance of B cell transformation. Blood 125, 2530-2543 (2015).
    • (2015) Blood , vol.125 , pp. 2530-2543
    • Alinari, L.1
  • 98
    • 84937397052 scopus 로고    scopus 로고
    • A selective inhibitor of PRMT5 with in vivo and in vitro potency in MCL models
    • Chan-Penebre, E., et al. A selective inhibitor of PRMT5 with in vivo and in vitro potency in MCL models. Nat. Chem. Biol. 11, 432-437 (2015).
    • (2015) Nat. Chem. Biol. , vol.11 , pp. 432-437
    • Chan-Penebre, E.1
  • 99
    • 84935871823 scopus 로고    scopus 로고
    • Aryl pyrazoles as potent inhibitors of arginine methyltransferases: Identification of the first PRMT6 tool compound
    • Mitchell, L. H., et al. Aryl pyrazoles as potent inhibitors of arginine methyltransferases: identification of the first PRMT6 tool compound. ACS Med. Chem. Lett. 6, 655-659 (2015).
    • (2015) ACS Med. Chem. Lett. , vol.6 , pp. 655-659
    • Mitchell, L.H.1
  • 100
    • 84927720612 scopus 로고    scopus 로고
    • A potent, selective and cell-Active allosteric inhibitor of protein arginine methyltransferase 3 (PRMT3)
    • Kaniskan, H. U., et al. A potent, selective and cell-Active allosteric inhibitor of protein arginine methyltransferase 3 (PRMT3). Angew. Chem. Int. Ed. 54, 5166-5170 (2015).
    • (2015) Angew. Chem. Int. Ed. , vol.54 , pp. 5166-5170
    • Kaniskan, H.U.1
  • 101
    • 84961820776 scopus 로고    scopus 로고
    • A potent, selective, and cell-Active inhibitor of human type i protein arginine methyltransferases
    • Eram, M. S., et al. A potent, selective, and cell-Active inhibitor of human type I protein arginine methyltransferases. ACS Chem. Biol. 11, 772-781 (2016).
    • (2016) ACS Chem. Biol. , vol.11 , pp. 772-781
    • Eram, M.S.1
  • 102
    • 11144332565 scopus 로고    scopus 로고
    • Histone demethylation mediated by the nuclear amine oxidase homolog LSD1
    • Shi, Y., et al. Histone demethylation mediated by the nuclear amine oxidase homolog LSD1. Cell 119, 941-953 (2004).
    • (2004) Cell , vol.119 , pp. 941-953
    • Shi, Y.1
  • 103
    • 24144462170 scopus 로고    scopus 로고
    • LSD1 demethylates repressive histone marks to promote androgen-receptor-dependent transcription
    • Metzger, E., et al. LSD1 demethylates repressive histone marks to promote androgen-receptor-dependent transcription. Nature 437, 436-439 (2005).
    • (2005) Nature , vol.437 , pp. 436-439
    • Metzger, E.1
  • 104
    • 84880719085 scopus 로고    scopus 로고
    • The histone LSD1 demethylase in stemness and cancer transcription programs
    • Amente, S., Lania, L., & Majello, B. The histone LSD1 demethylase in stemness and cancer transcription programs. Biochim. Biophys. Acta 1829, 981-986 (2013).
    • (2013) Biochim. Biophys. Acta , vol.1829 , pp. 981-986
    • Amente, S.1    Lania, L.2    Majello, B.3
  • 105
    • 0034282790 scopus 로고    scopus 로고
    • Identification of a novel gene, GASC1, within an amplicon at 9p23 24 frequently detected in esophageal cancer cell lines
    • Yang, Z. Q., et al. Identification of a novel gene, GASC1, within an amplicon at 9p23 24 frequently detected in esophageal cancer cell lines. Cancer Res. 60, 4735-4739 (2000).
    • (2000) Cancer Res. , vol.60 , pp. 4735-4739
    • Yang, Z.Q.1
  • 106
    • 0035909531 scopus 로고    scopus 로고
    • A novel nuclear protein, 5qNCA (LOC51780) is a candidate for the myeloid leukemia tumor suppressor gene on chromosome 5 band q31
    • Hu, Z., et al. A novel nuclear protein, 5qNCA (LOC51780) is a candidate for the myeloid leukemia tumor suppressor gene on chromosome 5 band q31. Oncogene 20, 6946-6954 (2001).
    • (2001) Oncogene , vol.20 , pp. 6946-6954
    • Hu, Z.1
  • 107
    • 33646138230 scopus 로고    scopus 로고
    • JHDM2A, a JmjC-containing H3K9 demethylase, facilitates transcription activation by androgen receptor
    • Yamane, K., et al. JHDM2A, a JmjC-containing H3K9 demethylase, facilitates transcription activation by androgen receptor. Cell 125, 483-495 (2006).
    • (2006) Cell , vol.125 , pp. 483-495
    • Yamane, K.1
  • 108
    • 33847392500 scopus 로고    scopus 로고
    • Cooperative demethylation by JMJD2C and LSD1 promotes androgen receptor-dependent gene expression
    • Wissmann, M., et al. Cooperative demethylation by JMJD2C and LSD1 promotes androgen receptor-dependent gene expression. Nat. Cell Biol. 9, 347-353 (2007).
    • (2007) Nat. Cell Biol. , vol.9 , pp. 347-353
    • Wissmann, M.1
  • 109
    • 33745187327 scopus 로고    scopus 로고
    • Histone H3 lysine 4 demethylation is a target of nonselective antidepressive medications
    • Lee, M. G., Wynder, C., Schmidt, D. M., McCafferty, D. G., & Shiekhattar, R. Histone H3 lysine 4 demethylation is a target of nonselective antidepressive medications. Chem. Biol. 13, 563-567 (2006).
    • (2006) Chem. Biol. , vol.13 , pp. 563-567
    • Lee, M.G.1    Wynder, C.2    Schmidt, D.M.3    McCafferty, D.G.4    Shiekhattar, R.5
  • 110
    • 34147173308 scopus 로고    scopus 로고
    • Trans 2 phenylcyclopropylamine is a mechanism-based inactivator of the histone demethylase LSD1
    • Schmidt, D. M., & McCafferty, D. G. trans 2 phenylcyclopropylamine is a mechanism-based inactivator of the histone demethylase LSD1. Biochemistry 46, 4408-4416 (2007).
    • (2007) Biochemistry , vol.46 , pp. 4408-4416
    • Schmidt, D.M.1    McCafferty, D.G.2
  • 111
    • 34447338875 scopus 로고    scopus 로고
    • Structural basis for the inhibition of the LSD1 histone demethylase by the antidepressant trans 2 phenylcyclopropylamine
    • Yang, M., et al. Structural basis for the inhibition of the LSD1 histone demethylase by the antidepressant trans 2 phenylcyclopropylamine. Biochemistry 46, 8058-8065 (2007).
    • (2007) Biochemistry , vol.46 , pp. 8058-8065
    • Yang, M.1
  • 112
    • 72249117352 scopus 로고    scopus 로고
    • Identification of cell-Active lysine specific demethylase 1 selective inhibitors
    • Ueda, R., et al. Identification of cell-Active lysine specific demethylase 1 selective inhibitors. J. Am. Chem. Soc.131, 17536-17537 (2009).
    • (2009) J. Am. Chem. Soc. , vol.131 , pp. 17536-17537
    • Ueda, R.1
  • 113
    • 77952355653 scopus 로고    scopus 로고
    • Biochemical, structural, and biological evaluation of tranylcypromine derivatives as inhibitors of histone demethylases LSD1 and LSD2
    • Binda, C., et al. Biochemical, structural, and biological evaluation of tranylcypromine derivatives as inhibitors of histone demethylases LSD1 and LSD2. J. Am. Chem. Soc. 132, 6827-6833 (2010).
    • (2010) J. Am. Chem. Soc. , vol.132 , pp. 6827-6833
    • Binda, C.1
  • 114
    • 77955025931 scopus 로고    scopus 로고
    • Structurally designed trans 2 phenylcyclopropylamine derivatives potently inhibit histone demethylase LSD1/KDM1
    • Mimasu, S., et al. Structurally designed trans 2 phenylcyclopropylamine derivatives potently inhibit histone demethylase LSD1/KDM1. Biochemistry 49, 6494-6503 (2010).
    • (2010) Biochemistry , vol.49 , pp. 6494-6503
    • Mimasu, S.1
  • 115
    • 84859837026 scopus 로고    scopus 로고
    • The histone demethylase KDM1A sustains the oncogenic potential of MLL AF9 leukemia stem cells
    • Harris, W. J., et al. The histone demethylase KDM1A sustains the oncogenic potential of MLL AF9 leukemia stem cells. Cancer Cell 21, 473-487 (2012).
    • (2012) Cancer Cell , vol.21 , pp. 473-487
    • Harris, W.J.1
  • 116
    • 84874598620 scopus 로고    scopus 로고
    • A novel selective LSD1/KDM1A inhibitor epigenetically blocks herpes simplex virus lytic replication and reactivation from latency
    • Liang, Y., et al. A novel selective LSD1/KDM1A inhibitor epigenetically blocks herpes simplex virus lytic replication and reactivation from latency. mBio 4, e00558 12 (2013).
    • (2013) MBio , vol.4 , pp. e0055812
    • Liang, Y.1
  • 117
    • 84937429405 scopus 로고    scopus 로고
    • A DNA hypomethylation signature predicts antitumor activity of LSD1 inhibitors in SCLC
    • Mohammad, H. P., et al. A DNA hypomethylation signature predicts antitumor activity of LSD1 inhibitors in SCLC. Cancer Cell 28, 57-69 (2015).
    • (2015) Cancer Cell , vol.28 , pp. 57-69
    • Mohammad, H.P.1
  • 118
    • 84862777766 scopus 로고    scopus 로고
    • Inhibition of the LSD1 (KDM1A) demethylase reactivates the all-Trans-retinoic acid differentiation pathway in acute myeloid leukemia
    • Schenk, T., et al. Inhibition of the LSD1 (KDM1A) demethylase reactivates the all-Trans-retinoic acid differentiation pathway in acute myeloid leukemia. Nat. Med. 18, 605-611 (2012).
    • (2012) Nat. Med. , vol.18 , pp. 605-611
    • Schenk, T.1
  • 119
    • 84926475466 scopus 로고    scopus 로고
    • Reversible inhibitors of LSD1 as therapeutic agents in acute myeloid leukemia: Clinical significance and progress to date
    • Mould, D. P., McGonagle, A. E., Wiseman, D. H., Williams, E. L., & Jordan, A. M. Reversible inhibitors of LSD1 as therapeutic agents in acute myeloid leukemia: clinical significance and progress to date. Med. Res. Rev. 35, 586-618 (2015).
    • (2015) Med. Res. Rev. , vol.35 , pp. 586-618
    • Mould, D.P.1    McGonagle, A.E.2    Wiseman, D.H.3    Williams, E.L.4    Jordan, A.M.5
  • 120
    • 84924423842 scopus 로고    scopus 로고
    • Novel anti-Tumor activity of targeted LSD1 inhibition by GSK2879552
    • Mohammad, H., et al. Novel anti-Tumor activity of targeted LSD1 inhibition by GSK2879552. Eur. J. Cancer 50, 72 (2014).
    • (2014) Eur. J. Cancer , vol.50 , pp. 72
    • Mohammad, H.1
  • 121
    • 36249005491 scopus 로고    scopus 로고
    • Succinate inhibition of alpha-ketoglutarate-dependent enzymes in a yeast model of paraganglioma
    • Smith, E. H., Janknecht, R., & Maher, L. J. III. Succinate inhibition of alpha-ketoglutarate-dependent enzymes in a yeast model of paraganglioma. Hum. Mol. Genet. 16, 3136-3148 (2007).
    • (2007) Hum. Mol. Genet. , vol.16 , pp. 3136-3148
    • Smith, E.H.1    Janknecht, R.2    Maher, L.J.3
  • 122
    • 56749151276 scopus 로고    scopus 로고
    • Inhibitor scaffolds for 2 oxoglutarate-dependent histone lysine demethylases
    • Rose, N. R., et al. Inhibitor scaffolds for 2 oxoglutarate-dependent histone lysine demethylases. J. Med. Chem. 51, 7053-7056 (2008).
    • (2008) J. Med. Chem. , vol.51 , pp. 7053-7056
    • Rose, N.R.1
  • 123
    • 77649224314 scopus 로고    scopus 로고
    • Selective inhibitors of the JMJD2 histone demethylases: Combined nondenaturing mass spectrometric screening and crystallographic approaches
    • Rose, N. R., et al. Selective inhibitors of the JMJD2 histone demethylases: combined nondenaturing mass spectrometric screening and crystallographic approaches. J. Med. Chem. 53, 1810-1818 (2010).
    • (2010) J. Med. Chem. , vol.53 , pp. 1810-1818
    • Rose, N.R.1
  • 124
    • 70350315800 scopus 로고    scopus 로고
    • Inhibition of the histone lysine demethylase JMJD2A by ejection of structural Zn(II)
    • Sekirnik, R., et al. Inhibition of the histone lysine demethylase JMJD2A by ejection of structural Zn(II). Chem. Commun. (Camb.) 42, 6376-6378 (2009).
    • (2009) Chem. Commun. (Camb.) , vol.42 , pp. 6376-6378
    • Sekirnik, R.1
  • 125
    • 33646061354 scopus 로고    scopus 로고
    • A mechanism-based inactivator for histone demethylase LSD1
    • Culhane, J. C., et al. A mechanism-based inactivator for histone demethylase LSD1. J. Am. Chem. Soc. 128, 4536-4537 (2006).
    • (2006) J. Am. Chem. Soc. , vol.128 , pp. 4536-4537
    • Culhane, J.C.1
  • 126
    • 65349129552 scopus 로고    scopus 로고
    • Synthesis and activity of N oxalylglycine and its derivatives as Jumonji C domain-containing histone lysine demethylase inhibitors
    • Hamada, S., et al. Synthesis and activity of N oxalylglycine and its derivatives as Jumonji C domain-containing histone lysine demethylase inhibitors. Bioorg. Med. Chem. Lett. 19, 2852-2855 (2009).
    • (2009) Bioorg. Med. Chem. Lett. , vol.19 , pp. 2852-2855
    • Hamada, S.1
  • 127
    • 78649528262 scopus 로고    scopus 로고
    • Quantitative high-Throughput screening identifies 8 hydroxyquinolines as cell-Active histone demethylase inhibitors
    • King, O. N., et al. Quantitative high-Throughput screening identifies 8 hydroxyquinolines as cell-Active histone demethylase inhibitors. PLoS ONE 5, e15535 (2010).
    • (2010) PLoS ONE , vol.5 , pp. e15535
    • King, O.N.1
  • 128
    • 78650107087 scopus 로고    scopus 로고
    • Inhibition of the histone demethylase JMJD2E by 3 substituted pyridine 2, 4 dicarboxylates
    • Thalhammer, A., et al. Inhibition of the histone demethylase JMJD2E by 3 substituted pyridine 2, 4 dicarboxylates. Org. Biomol. Chem. 9, 127-135 (2011).
    • (2011) Org. Biomol. Chem. , vol.9 , pp. 127-135
    • Thalhammer, A.1
  • 129
    • 79959204866 scopus 로고    scopus 로고
    • A selective inhibitor and probe of the cellular functions of Jumonji C domain-containing histone demethylases
    • Luo, X., et al. A selective inhibitor and probe of the cellular functions of Jumonji C domain-containing histone demethylases. J. Am. Chem. Soc.133, 9451-9456 (2011).
    • (2011) J. Am. Chem. Soc. , vol.133 , pp. 9451-9456
    • Luo, X.1
  • 130
    • 84865120905 scopus 로고    scopus 로고
    • A selective jumonji H3K27 demethylase inhibitor modulates the proinflammatory macrophage response
    • Kruidenier, L., et al. A selective jumonji H3K27 demethylase inhibitor modulates the proinflammatory macrophage response. Nature 488, 404-408 (2012).
    • (2012) Nature , vol.488 , pp. 404-408
    • Kruidenier, L.1
  • 131
    • 84907526739 scopus 로고    scopus 로고
    • Contrasting roles of histone 3 lysine 27 demethylases in acute lymphoblastic leukaemia
    • Ntziachristos, P., et al. Contrasting roles of histone 3 lysine 27 demethylases in acute lymphoblastic leukaemia. Nature 514, 513-517 (2014).
    • (2014) Nature , vol.514 , pp. 513-517
    • Ntziachristos, P.1
  • 132
    • 84961291627 scopus 로고    scopus 로고
    • Pharmacologic inhibition of histone demethylation as a therapy for pediatric brainstem glioma
    • Hashizume, R., et al. Pharmacologic inhibition of histone demethylation as a therapy for pediatric brainstem glioma. Nat. Med. 20, 1394-1396 (2014).
    • (2014) Nat. Med. , vol.20 , pp. 1394-1396
    • Hashizume, R.1
  • 133
    • 84908029332 scopus 로고    scopus 로고
    • Inhibition of demethylases by GSK J1/J4
    • Heinemann, B., et al. Inhibition of demethylases by GSK J1/J4. Nature 514, E1-E2 (2014).
    • (2014) Nature , vol.514 , pp. E1-E2
    • Heinemann, B.1
  • 134
    • 35848961668 scopus 로고    scopus 로고
    • How chromatin-binding modules interpret histone modifications: Lessons from professional pocket pickers
    • Taverna, S. D., Li, H., Ruthenburg, A. J., Allis, C. D., & Patel, D. J. How chromatin-binding modules interpret histone modifications: lessons from professional pocket pickers. Nat. Struct. Mol. Biol. 14, 1025-1040 (2007).
    • (2007) Nat. Struct. Mol. Biol. , vol.14 , pp. 1025-1040
    • Taverna, S.D.1    Li, H.2    Ruthenburg, A.J.3    Allis, C.D.4    Patel, D.J.5
  • 135
    • 67349266612 scopus 로고    scopus 로고
    • Haematopoietic malignancies caused by dysregulation of a chromatin-binding PHD finger
    • Wang, G. G., et al. Haematopoietic malignancies caused by dysregulation of a chromatin-binding PHD finger. Nature 459, 847-851 (2009).
    • (2009) Nature , vol.459 , pp. 847-851
    • Wang, G.G.1
  • 136
    • 84874116695 scopus 로고    scopus 로고
    • Discovery of a chemical probe for the L3MBTL3 methyllysine reader domain
    • James, L. I., et al. Discovery of a chemical probe for the L3MBTL3 methyllysine reader domain. Nat. Chem. Biol. 9, 184-191 (2013).
    • (2013) Nat. Chem. Biol. , vol.9 , pp. 184-191
    • James, L.I.1
  • 137
    • 84884993346 scopus 로고    scopus 로고
    • Small-molecule ligands of methyl-lysine binding proteins: Optimization of selectivity for L3MBTL3
    • James, L. I., et al. Small-molecule ligands of methyl-lysine binding proteins: optimization of selectivity for L3MBTL3. J. Med. Chem. 56, 7358-7371 (2013).
    • (2013) J. Med. Chem. , vol.56 , pp. 7358-7371
    • James, L.I.1
  • 138
    • 84958983631 scopus 로고    scopus 로고
    • Selective inhibition of CBX6: A methyllysine reader protein in the Polycomb family
    • Milosevich, N., et al. Selective inhibition of CBX6: a methyllysine reader protein in the Polycomb family. ACS Med. Chem. Lett. 7, 139-144 (2016).
    • (2016) ACS Med. Chem. Lett. , vol.7 , pp. 139-144
    • Milosevich, N.1
  • 139
    • 84955558290 scopus 로고    scopus 로고
    • A cellular chemical probe targeting the chromodomains of Polycomb repressive complex 1
    • Stuckey, J. I., et al. A cellular chemical probe targeting the chromodomains of Polycomb repressive complex 1. Nat. Chem. Biol. 12, 180-187 (2016).
    • (2016) Nat. Chem. Biol. , vol.12 , pp. 180-187
    • Stuckey, J.I.1
  • 140
    • 68949212379 scopus 로고    scopus 로고
    • Lysine acetylation targets protein complexes and co regulates major cellular functions
    • Choudhary, C., et al. Lysine acetylation targets protein complexes and co regulates major cellular functions. Science 325, 834-840 (2009).
    • (2009) Science , vol.325 , pp. 834-840
    • Choudhary, C.1
  • 142
    • 84934282991 scopus 로고    scopus 로고
    • The role of histone acetyltransferases in normal and malignant hematopoiesis
    • Sun, X. J., Man, N., Tan, Y., Nimer, S. D., & Wang, L. The role of histone acetyltransferases in normal and malignant hematopoiesis. Front. Oncol. 5, 108 (2015).
    • (2015) Front. Oncol. , vol.5 , pp. 108
    • Sun, X.J.1    Man, N.2    Tan, Y.3    Nimer, S.D.4    Wang, L.5
  • 143
    • 77954204335 scopus 로고    scopus 로고
    • Virtual ligand screening of the p300/CBP histone acetyltransferase: Identification of a selective small molecule inhibitor
    • Bowers, E. M., et al. Virtual ligand screening of the p300/CBP histone acetyltransferase: identification of a selective small molecule inhibitor. Chem. Biol. 17, 471-482 (2010).
    • (2010) Chem. Biol. , vol.17 , pp. 471-482
    • Bowers, E.M.1
  • 144
    • 84962381628 scopus 로고    scopus 로고
    • Targeting p300 addiction in CBP-deficient cancers causes synthetic lethality via apoptotic cell death due to abrogation of MYC expression
    • Ogiwara, H., et al. Targeting p300 addiction in CBP-deficient cancers causes synthetic lethality via apoptotic cell death due to abrogation of MYC expression. Cancer Discov. 6, 430-445 (2015).
    • (2015) Cancer Discov. , vol.6 , pp. 430-445
    • Ogiwara, H.1
  • 145
    • 80051470522 scopus 로고    scopus 로고
    • The leukemogenicity of AML1 ETO is dependent on site-specific lysine acetylation
    • Wang, L., et al. The leukemogenicity of AML1 ETO is dependent on site-specific lysine acetylation. Science 333, 765-769 (2011).
    • (2011) Science , vol.333 , pp. 765-769
    • Wang, L.1
  • 146
    • 77956047888 scopus 로고    scopus 로고
    • Nitric oxide-mediated histone hyperacetylation in oral cancer: Target for a water-soluble HAT inhibitor
    • Arif, M., et al. Nitric oxide-mediated histone hyperacetylation in oral cancer: target for a water-soluble HAT inhibitor, CTK7A. Chem. Biol. 17, 903-913 (2010).
    • (2010) CTK7A. Chem. Biol. , vol.17 , pp. 903-913
    • Arif, M.1
  • 147
    • 84877642396 scopus 로고    scopus 로고
    • Small-molecule inhibitors of acetyltransferase p300 identified by high-Throughput screening are potent anticancer agents
    • Yang, H., et al. Small-molecule inhibitors of acetyltransferase p300 identified by high-Throughput screening are potent anticancer agents. Mol. Cancer Ther. 12, 610-620 (2013).
    • (2013) Mol. Cancer Ther. , vol.12 , pp. 610-620
    • Yang, H.1
  • 148
    • 69949169110 scopus 로고    scopus 로고
    • Inhibition of lysine acetyltransferase KAT3B/p300 activity by a naturally occurring hydroxynaphthoquinone, plumbagin
    • Ravindra, K. C., et al. Inhibition of lysine acetyltransferase KAT3B/p300 activity by a naturally occurring hydroxynaphthoquinone, plumbagin. J. Biol. Chem. 284, 24453-24464 (2009).
    • (2009) J. Biol. Chem. , vol.284 , pp. 24453-24464
    • Ravindra, K.C.1
  • 149
    • 84879756597 scopus 로고    scopus 로고
    • Probing p300/CBP associated factor (PCAF)-dependent pathways with a small molecule inhibitor
    • Modak, R., et al. Probing p300/CBP associated factor (PCAF)-dependent pathways with a small molecule inhibitor. ACS Chem. Biol. 8, 1311-1323 (2013).
    • (2013) ACS Chem. Biol. , vol.8 , pp. 1311-1323
    • Modak, R.1
  • 150
    • 84979496867 scopus 로고    scopus 로고
    • Salicylate diflunisal and their metabolites inhibit CBP/p300 and exhibit anticancer activity
    • Shirakawa, K., et al. Salicylate, diflunisal and their metabolites inhibit CBP/p300 and exhibit anticancer activity. eLife 5, e11156 (2016).
    • (2016) ELife , vol.5 , pp. e11156
    • Shirakawa, K.1
  • 151
    • 39149109887 scopus 로고    scopus 로고
    • The structural basis of protein acetylation by the p300/CBP transcriptional coactivator
    • Liu, X., et al. The structural basis of protein acetylation by the p300/CBP transcriptional coactivator. Nature 451, 846-850 (2008).
    • (2008) Nature , vol.451 , pp. 846-850
    • Liu, X.1
  • 152
    • 84883743725 scopus 로고    scopus 로고
    • Structure of the p300 catalytic core and implications for chromatin targeting and HAT regulation
    • Delvecchio, M., Gaucher, J., Aguilar-Gurrieri, C., Ortega, E., & Panne, D. Structure of the p300 catalytic core and implications for chromatin targeting and HAT regulation. Nat. Struct. Mol. Biol. 20, 1040-1046 (2013).
    • (2013) Nat. Struct. Mol. Biol. , vol.20 , pp. 1040-1046
    • Delvecchio, M.1    Gaucher, J.2    Aguilar-Gurrieri, C.3    Ortega, E.4    Panne, D.5
  • 153
    • 33748451151 scopus 로고    scopus 로고
    • Anticancer activities of histone deacetylase inhibitors
    • Bolden, J. E., Peart, M. J., & Johnstone, R. W. Anticancer activities of histone deacetylase inhibitors. Nat. Rev. Drug Discov. 5, 769-784 (2006).
    • (2006) Nat. Rev. Drug Discov. , vol.5 , pp. 769-784
    • Bolden, J.E.1    Peart, M.J.2    Johnstone, R.W.3
  • 154
    • 0035961036 scopus 로고    scopus 로고
    • Synthesis of 7200 small molecules based on a substructural analysis of the histone deacetylase inhibitors trichostatin and trapoxin
    • Sternson, S. M., Wong, J. C., Grozinger, C. M., & Schreiber, S. L. Synthesis of 7200 small molecules based on a substructural analysis of the histone deacetylase inhibitors trichostatin and trapoxin. Org. Lett. 3, 4239-4242 (2001).
    • (2001) Org. Lett. , vol.3 , pp. 4239-4242
    • Sternson, S.M.1    Wong, J.C.2    Grozinger, C.M.3    Schreiber, S.L.4
  • 155
    • 1842578986 scopus 로고    scopus 로고
    • Molecular evolution of the histone deacetylase family: Functional implications of phylogenetic analysis
    • Gregoretti, I. V., Lee, Y. M., & Goodson, H. V. Molecular evolution of the histone deacetylase family: functional implications of phylogenetic analysis. J. Mol. Biol. 338, 17-31 (2004).
    • (2004) J. Mol. Biol. , vol.338 , pp. 17-31
    • Gregoretti, I.V.1    Lee, Y.M.2    Goodson, H.V.3
  • 156
    • 77249087051 scopus 로고    scopus 로고
    • Chemical phylogenetics of histone deacetylases
    • Bradner, J. E., et al. Chemical phylogenetics of histone deacetylases. Nat. Chem. Biol. 6, 238-243 (2010).
    • (2010) Nat. Chem. Biol. , vol.6 , pp. 238-243
    • Bradner, J.E.1
  • 157
    • 79952396960 scopus 로고    scopus 로고
    • Chemoproteomics profiling of HDAC inhibitors reveals selective targeting of HDAC complexes
    • Bantscheff, M., et al. Chemoproteomics profiling of HDAC inhibitors reveals selective targeting of HDAC complexes. Nat. Biotechnol. 29, 255-265 (2011).
    • (2011) Nat. Biotechnol. , vol.29 , pp. 255-265
    • Bantscheff, M.1
  • 158
    • 0038274087 scopus 로고    scopus 로고
    • Structural biasing elements for in cell histone deacetylase paralog selectivity
    • Wong, J. C., Hong, R., & Schreiber, S. L. Structural biasing elements for in cell histone deacetylase paralog selectivity. J. Am. Chem. Soc. 125, 5586-5587 (2003).
    • (2003) J. Am. Chem. Soc. , vol.125 , pp. 5586-5587
    • Wong, J.C.1    Hong, R.2    Schreiber, S.L.3
  • 159
    • 0000032126 scopus 로고    scopus 로고
    • Depudecin induces morphological reversion of transformed fibroblasts via the inhibition of histone deacetylase
    • Kwon, H. J., Owa, T., Hassig, C. A., Shimada, J., & Schreiber, S. L. Depudecin induces morphological reversion of transformed fibroblasts via the inhibition of histone deacetylase. Proc. Natl Acad. Sci. USA 95, 3356-3361 (1998).
    • (1998) Proc. Natl Acad. Sci. USA , vol.95 , pp. 3356-3361
    • Kwon, H.J.1    Owa, T.2    Hassig, C.A.3    Shimada, J.4    Schreiber, S.L.5
  • 160
    • 49849106385 scopus 로고    scopus 로고
    • KD5170, a novel mercaptoketone-based histone deacetylase inhibitor that exhibits broad spectrum antitumor activity in vitro and in vivo
    • Hassig, C. A., et al. KD5170, a novel mercaptoketone-based histone deacetylase inhibitor that exhibits broad spectrum antitumor activity in vitro and in vivo. Mol. Cancer Ther. 7, 1054-1065 (2008).
    • (2008) Mol. Cancer Ther. , vol.7 , pp. 1054-1065
    • Hassig, C.A.1
  • 161
    • 84865171888 scopus 로고    scopus 로고
    • Anticancer activity of MPT0E028, a novel potent histone deacetylase inhibitor, in human colorectal cancer HCT116 cells in vitro and in vivo
    • Huang, H. L., et al. Anticancer activity of MPT0E028, a novel potent histone deacetylase inhibitor, in human colorectal cancer HCT116 cells in vitro and in vivo. PLoS ONE 7, e43645 (2012).
    • (2012) PLoS ONE , vol.7 , pp. e43645
    • Huang, H.L.1
  • 163
    • 0026548820 scopus 로고
    • Morphological reversion of sis-Transformed NIH3T3 cells by trichostatin A
    • Sugita, K., Koizumi, K., & Yoshida, H Morphological Reversion of Sis-Transformed NIH3T3 Cells by Trichostatin A. Cancer Res. 52, 168-172 (1992).
    • (1992) Cancer Res. , vol.52 , pp. 168-172
    • Sugita, K.1    Koizumi, K.2    Yoshida, H.3
  • 164
    • 33847358736 scopus 로고    scopus 로고
    • Trichostatin A increases SMN expression and survival in a mouse model of spinal muscular atrophy
    • Avila, A. M., et al. Trichostatin A increases SMN expression and survival in a mouse model of spinal muscular atrophy. J. Clin. Invest. 117, 659-671 (2007).
    • (2007) J. Clin. Invest. , vol.117 , pp. 659-671
    • Avila, A.M.1
  • 165
    • 34547683194 scopus 로고    scopus 로고
    • Phase IIb multicenter trial of vorinostat in patients with persistent, progressive, or treatment refractory cutaneous T cell lymphoma
    • Olsen, E. A., et al. Phase IIb multicenter trial of vorinostat in patients with persistent, progressive, or treatment refractory cutaneous T cell lymphoma. J. Clin. Oncol. 25, 3109-3115 (2007).
    • (2007) J. Clin. Oncol. , vol.25 , pp. 3109-3115
    • Olsen, E.A.1
  • 166
    • 84949024001 scopus 로고    scopus 로고
    • Functional-genetic dissection of HDAC dependencies in mouse lymphoid and myeloid malignancies
    • Matthews, G. M., et al. Functional-genetic dissection of HDAC dependencies in mouse lymphoid and myeloid malignancies. Blood 126, 2392-2403 (2015).
    • (2015) Blood , vol.126 , pp. 2392-2403
    • Matthews, G.M.1
  • 167
    • 84895815366 scopus 로고    scopus 로고
    • Histone deacetylase 3 as a novel therapeutic target in multiple myeloma
    • Minami, J., et al. Histone deacetylase 3 as a novel therapeutic target in multiple myeloma. Leukemia 28, 680-689 (2014).
    • (2014) Leukemia , vol.28 , pp. 680-689
    • Minami, J.1
  • 168
    • 84880655164 scopus 로고    scopus 로고
    • Inhibition of histone deacetylase 3 causes replication stress in cutaneous T cell lymphoma
    • Wells, C. E., et al. Inhibition of histone deacetylase 3 causes replication stress in cutaneous T cell lymphoma. PLoS ONE 8, e68915 (2013).
    • (2013) PLoS ONE , vol.8 , pp. e68915
    • Wells, C.E.1
  • 169
    • 18244383806 scopus 로고    scopus 로고
    • Valproic acid defines a novel class of HDAC inhibitors inducing differentiation of transformed cells
    • Göttlicher, M., et al. Valproic acid defines a novel class of HDAC inhibitors inducing differentiation of transformed cells. EMBO J. 20, 6969-6978 (2001).
    • (2001) EMBO J. , vol.20 , pp. 6969-6978
    • Göttlicher, M.1
  • 170
    • 0017767153 scopus 로고
    • N Butyrate causes histone modification in HeLa and Friend erythroleukaemia cells
    • Riggs, M. G., Whittaker, R. G., Neumann, J. R., & Ingram, V. M. n Butyrate causes histone modification in HeLa and Friend erythroleukaemia cells. Nature 268, 462-464 (1977).
    • (1977) Nature , vol.268 , pp. 462-464
    • Riggs, M.G.1    Whittaker, R.G.2    Neumann, J.R.3    Ingram, V.M.4
  • 171
    • 79955701085 scopus 로고    scopus 로고
    • Hydralazine and magnesium valproate as epigenetic treatment for myelodysplastic syndrome Preliminary results of a phase II trial
    • Candelaria, M., et al. Hydralazine and magnesium valproate as epigenetic treatment for myelodysplastic syndrome. Preliminary results of a phase II trial. Ann. Hematol. 90, 379-387 (2011).
    • (2011) Ann. Hematol. , vol.90 , pp. 379-387
    • Candelaria, M.1
  • 172
    • 84857522968 scopus 로고    scopus 로고
    • Results from a pivotal, open-label, phase II study of romidepsin in relapsed or refractory peripheral T cell lymphoma after prior systemic therapy
    • Coiffier, B., et al. Results from a pivotal, open-label, phase II study of romidepsin in relapsed or refractory peripheral T cell lymphoma after prior systemic therapy. J. Clin. Oncol. 30, 631-636 (2012).
    • (2012) J. Clin. Oncol. , vol.30 , pp. 631-636
    • Coiffier, B.1
  • 173
    • 0036735385 scopus 로고    scopus 로고
    • FK228 (depsipeptide) as a natural prodrug that inhibits class i histone deacetylases
    • Furumai, R., et al. FK228 (depsipeptide) as a natural prodrug that inhibits class I histone deacetylases. Cancer Res. 62, 4916-4921 (2002).
    • (2002) Cancer Res. , vol.62 , pp. 4916-4921
    • Furumai, R.1
  • 174
    • 77954879663 scopus 로고    scopus 로고
    • Final results from a multicenter, international, pivotal study of romidepsin in refractory cutaneous T cell lymphoma
    • Whittaker, S. J., et al. Final results from a multicenter, international, pivotal study of romidepsin in refractory cutaneous T cell lymphoma. J. Clin. Oncol. 28, 4485-4491 (2010).
    • (2010) J. Clin. Oncol. , vol.28 , pp. 4485-4491
    • Whittaker, S.J.1
  • 175
    • 0027185184 scopus 로고
    • Acetyldinaline: A new oral cytostatic drug with impressive differential activity against leukemic cells and normal stem cells-preclinical studies in a relevant rat model for human acute myelocytic leukemia
    • El Beltagi, H. M., Martens, A. C. M., Lelieveld, P., Haroun, E. A., & Hagenbeek, A. Acetyldinaline: a new oral cytostatic drug with impressive differential activity against leukemic cells and normal stem cells-preclinical studies in a relevant rat model for human acute myelocytic leukemia. Cancer Res. 53, 3008-3014 (1993).
    • (1993) Cancer Res. , vol.53 , pp. 3008-3014
    • El Beltagi, H.M.1    Martens, A.C.M.2    Lelieveld, P.3    Haroun, E.A.4    Hagenbeek, A.5
  • 176
    • 0033551152 scopus 로고    scopus 로고
    • A synthetic inhibitor of histone deacetylase, MS 27 275, with marked in vivo antitumor activity against human tumors
    • Saito, A., et al. A synthetic inhibitor of histone deacetylase, MS 27 275, with marked in vivo antitumor activity against human tumors Proc. Natl Acad. Sci. USA 96, 4592-4597 (1999).
    • (1999) Proc Natl Acad. Sci. USA , vol.96 , pp. 4592-4597
    • Saito, A.1
  • 177
    • 38749090156 scopus 로고    scopus 로고
    • Design and synthesis of 4-[(s triazin 2 ylamino)methyl]-N-(2 aminophenyl)-benzamides and their analogues as a novel class of histone deacetylase inhibitors
    • Paquin, I., et al. Design and synthesis of 4-[(s triazin 2 ylamino)methyl]-N-(2 aminophenyl)-benzamides and their analogues as a novel class of histone deacetylase inhibitors. Bioorg. Med. Chem. Lett. 18, 1067-1071 (2008).
    • (2008) Bioorg. Med. Chem. Lett. , vol.18 , pp. 1067-1071
    • Paquin, I.1
  • 178
    • 58149089923 scopus 로고    scopus 로고
    • Pimelic diphenylamide 106 is a slow, tight-binding inhibitor of class i histone deacetylases
    • Chou, C. J., Herman, D., & Gottesfeld, J. M. Pimelic diphenylamide 106 is a slow, tight-binding inhibitor of class I histone deacetylases. J. Biol. Chem. 283, 35402-35409 (2008).
    • (2008) J. Biol. Chem. , vol.283 , pp. 35402-35409
    • Chou, C.J.1    Herman, D.2    Gottesfeld, J.M.3
  • 179
    • 38749136234 scopus 로고    scopus 로고
    • Exploration of the internal cavity of histone deacetylase (HDAC) with selective HDAC1/ HDAC2 inhibitors (SHI 1:2)
    • Methot, J. L., et al. Exploration of the internal cavity of histone deacetylase (HDAC) with selective HDAC1/ HDAC2 inhibitors (SHI 1:2). Bioorg. Med. Chem. Lett. 18, 973-978 (2008).
    • (2008) Bioorg. Med. Chem. Lett. , vol.18 , pp. 973-978
    • Methot, J.L.1
  • 180
    • 84863194158 scopus 로고    scopus 로고
    • Inhibition of the function of class IIa HDACs by blocking their interaction with MEF2
    • Jayathilaka, N., et al. Inhibition of the function of class IIa HDACs by blocking their interaction with MEF2. Nucleic Acids Res. 40, 5378-5388 (2012).
    • (2012) Nucleic Acids Res. , vol.40 , pp. 5378-5388
    • Jayathilaka, N.1
  • 181
    • 71049156256 scopus 로고    scopus 로고
    • Identification of novel, selective, and stable inhibitors of class II histone deacetylases Validation studies of the enzymatic activity of HDAC4 by small molecules as a novel approach for cancer therapy
    • Ontoria, J. M., et al. Identification of novel, selective, and stable inhibitors of class II histone deacetylases. Validation studies of the enzymatic activity of HDAC4 by small molecules as a novel approach for cancer therapy. J. Med. Chem. 52, 6782-6783 (2009).
    • (2009) J. Med. Chem. , vol.52 , pp. 6782-6783
    • Ontoria, J.M.1
  • 182
    • 84879092346 scopus 로고    scopus 로고
    • Selective class IIa histone deacetylase inhibition via a non-chelating zinc-binding group
    • Lobera, M., et al. Selective class IIa histone deacetylase inhibition via a non-chelating zinc-binding group. Nat. Chem. Biol. 9, 319-325 (2013).
    • (2013) Nat. Chem. Biol. , vol.9 , pp. 319-325
    • Lobera, M.1
  • 183
    • 20944435415 scopus 로고    scopus 로고
    • Class II (IIa)-selective histone deacetylase inhibitors. 1 Synthesis and biological evaluation of novel (aryloxopropenyl)pyrrolyl hydroxamides
    • Mai, A., et al. Class II (IIa)-selective histone deacetylase inhibitors. 1. Synthesis and biological evaluation of novel (aryloxopropenyl)pyrrolyl hydroxamides. J. Med. Chem. 48, 3344-3353 (2005).
    • (2005) J. Med. Chem. , vol.48 , pp. 3344-3353
    • Mai, A.1
  • 184
    • 84942372058 scopus 로고    scopus 로고
    • The multifaceted functions of sirtuins in cancer
    • Chalkiadaki, A., & Guarente, L. The multifaceted functions of sirtuins in cancer. Nat. Rev. Cancer 15, 608-624 (2015).
    • (2015) Nat. Rev. Cancer , vol.15 , pp. 608-624
    • Chalkiadaki, A.1    Guarente, L.2
  • 185
    • 53149137486 scopus 로고    scopus 로고
    • Impaired DNA damage response, genome instability, and tumorigenesis in SIRT1 mutant mice
    • Wang, R. H., et al. Impaired DNA damage response, genome instability, and tumorigenesis in SIRT1 mutant mice. Cancer Cell 14, 312-323 (2008).
    • (2008) Cancer Cell , vol.14 , pp. 312-323
    • Wang, R.H.1
  • 186
    • 84898011296 scopus 로고    scopus 로고
    • Sorting out functions of sirtuins in cancer
    • Roth, M., & Chen, W. Y. Sorting out functions of sirtuins in cancer. Oncogene 33, 1609-1620 (2014).
    • (2014) Oncogene , vol.33 , pp. 1609-1620
    • Roth, M.1    Chen, W.Y.2
  • 187
    • 84867702707 scopus 로고    scopus 로고
    • Sirtuin activators and inhibitors
    • Villalba, J. M., & Alcain, F. J. Sirtuin activators and inhibitors. BioFactors 38, 349-359 (2012).
    • (2012) BioFactors , vol.38 , pp. 349-359
    • Villalba, J.M.1    Alcain, F.J.2
  • 188
    • 0141719702 scopus 로고    scopus 로고
    • Small molecule activators of sirtuins extend Saccharomyces cerevisiae lifespan
    • Howitz, K. T., et al. Small molecule activators of sirtuins extend Saccharomyces cerevisiae lifespan. Nature 425, 191-196 (2003).
    • (2003) Nature , vol.425 , pp. 191-196
    • Howitz, K.T.1
  • 189
    • 36749087548 scopus 로고    scopus 로고
    • Small molecule activators of SIRT1 as therapeutics for the treatment of type 2 diabetes
    • Milne, J. C., et al. Small molecule activators of SIRT1 as therapeutics for the treatment of type 2 diabetes. Nature 450, 712-716 (2007).
    • (2007) Nature , vol.450 , pp. 712-716
    • Milne, J.C.1
  • 190
    • 81155148158 scopus 로고    scopus 로고
    • Preclinical evaluation of a novel SIRT1 modulator SRT1720 in multiple myeloma cells
    • Chauhan, D., et al. Preclinical evaluation of a novel SIRT1 modulator SRT1720 in multiple myeloma cells. Br. J. Haematol. 155, 588-598 (2011).
    • (2011) Br. J. Haematol. , vol.155 , pp. 588-598
    • Chauhan, D.1
  • 191
    • 84926628784 scopus 로고    scopus 로고
    • DOT1L inhibits SIRT1 mediated epigenetic silencing to maintain leukemic gene expression in MLL-rearranged leukemia
    • Chen, C. W., et al. DOT1L inhibits SIRT1 mediated epigenetic silencing to maintain leukemic gene expression in MLL-rearranged leukemia. Nat. Med. 21, 335-343 (2015).
    • (2015) Nat. Med. , vol.21 , pp. 335-343
    • Chen, C.W.1
  • 192
    • 0037160097 scopus 로고    scopus 로고
    • Inhibition of silencing and accelerated aging by nicotinamide, a putative negative regulator of yeast sir2 and human SIRT1
    • Bitterman, K. J., Anderson, R. M., Cohen, H. Y., Latorre-Esteves, M., & Sinclair, D. A. Inhibition of silencing and accelerated aging by nicotinamide, a putative negative regulator of yeast sir2 and human SIRT1. J. Biol. Chem. 277, 45099-45107 (2002).
    • (2002) J. Biol. Chem. , vol.277 , pp. 45099-45107
    • Bitterman, K.J.1    Anderson, R.M.2    Cohen, H.Y.3    Latorre-Esteves, M.4    Sinclair, D.A.5
  • 193
    • 37349110743 scopus 로고    scopus 로고
    • Mechanism-based inhibition of Sir2 deacetylases by thioacetyl-lysine peptide
    • Smith, B. C., & Denu, J. M. Mechanism-based inhibition of Sir2 deacetylases by thioacetyl-lysine peptide. Biochemistry 46, 14478-14486 (2007).
    • (2007) Biochemistry , vol.46 , pp. 14478-14486
    • Smith, B.C.1    Denu, J.M.2
  • 194
    • 28144438533 scopus 로고    scopus 로고
    • Design, syn thesis, and biological evaluation of sirtinol analogues as class III histone/ protein deacetylase (Sirtuin) inhibitors
    • Mai, A., et al. Design, synthesis, and biological evaluation of sirtinol analogues as class III histone/ protein deacetylase (Sirtuin) inhibitors. J. Med. Chem. 48, 7789-7795 (2005).
    • (2005) J. Med. Chem. , vol.48 , pp. 7789-7795
    • Mai, A.1
  • 195
    • 0035914304 scopus 로고    scopus 로고
    • Identification of a class of small molecule inhibitors of the sirtuin family of NAD-dependent deacetylases by phenotypic screening
    • Grozinger, C. M., Chao, E. D., Blackwell, H. E., Moazed, D., & Schreiber, S. L. Identification of a class of small molecule inhibitors of the sirtuin family of NAD-dependent deacetylases by phenotypic screening. J. Biol. Chem. 276, 38837-38843 (2001).
    • (2001) J. Biol. Chem. , vol.276 , pp. 38837-38843
    • Grozinger, C.M.1    Chao, E.D.2    Blackwell, H.E.3    Moazed, D.4    Schreiber, S.L.5
  • 196
    • 9744284968 scopus 로고    scopus 로고
    • An in silico approach to discovering novel inhibitors of human sirtuin type 2
    • Tervo, A. J., et al. An in silico approach to discovering novel inhibitors of human sirtuin type 2. J. Med. Chem. 47, 6292-6298 (2004).
    • (2004) J. Med. Chem. , vol.47 , pp. 6292-6298
    • Tervo, A.J.1
  • 197
    • 53649086367 scopus 로고    scopus 로고
    • Mechanisms and molecular probes of sirtuins
    • Smith, B. C., Hallows, W. C., & Denu, J. M. Mechanisms and molecular probes of sirtuins. Chem. Biol. 15, 1002-1013 (2008).
    • (2008) Chem. Biol. , vol.15 , pp. 1002-1013
    • Smith, B.C.1    Hallows, W.C.2    Denu, J.M.3
  • 198
    • 84881613507 scopus 로고    scopus 로고
    • Pleiotropic effects of the sirtuin inhibitor sirtinol involves concentration-dependent modulation of multiple nuclear receptor-mediated pathways in androgen-responsive prostate cancer cell LNCaP
    • Wang, T. T., Schoene, N. W., Kim, E. K., & Kim, Y. S. Pleiotropic effects of the sirtuin inhibitor sirtinol involves concentration-dependent modulation of multiple nuclear receptor-mediated pathways in androgen-responsive prostate cancer cell LNCaP. Mol. Carcinog. 52, 676-685 (2013).
    • (2013) Mol. Carcinog. , vol.52 , pp. 676-685
    • Wang, T.T.1    Schoene, N.W.2    Kim, E.K.3    Kim, Y.S.4
  • 200
    • 33646254136 scopus 로고    scopus 로고
    • Antitumor activity of a small-molecule inhibitor of human silent information regulator 2 enzymes
    • Heltweg, B., et al. Antitumor activity of a small-molecule inhibitor of human silent information regulator 2 enzymes. Cancer Res. 66, 4368-4377 (2006).
    • (2006) Cancer Res. , vol.66 , pp. 4368-4377
    • Heltweg, B.1
  • 201
    • 84899500104 scopus 로고    scopus 로고
    • Development of pyrazolone and isoxazol 5 one cambinol analogues as sirtuin inhibitors
    • Mahajan, S. S., et al. Development of pyrazolone and isoxazol 5 one cambinol analogues as sirtuin inhibitors. J. Med. Chem. 57, 3283-3294 (2014).
    • (2014) J. Med. Chem. , vol.57 , pp. 3283-3294
    • Mahajan, S.S.1
  • 202
    • 42949114938 scopus 로고    scopus 로고
    • Discovery. in vivo activity, and mechanism of action of a small-molecule p53 activator
    • Lain, S., et al. Discovery. in vivo activity, and mechanism of action of a small-molecule p53 activator. Cancer Cell 13, 454-463 (2008).
    • (2008) Cancer Cell , vol.13 , pp. 454-463
    • Lain, S.1
  • 203
    • 84863011183 scopus 로고    scopus 로고
    • Activation of p53 by SIRT1 inhibition enhances elimination of CML leukemia stem cells in combination with imatinib
    • Li, L., et al. Activation of p53 by SIRT1 inhibition enhances elimination of CML leukemia stem cells in combination with imatinib. Cancer Cell 21, 266-281 (2012).
    • (2012) Cancer Cell , vol.21 , pp. 266-281
    • Li, L.1
  • 204
    • 84876473075 scopus 로고    scopus 로고
    • Tenovin D3, a novel small-molecule inhibitor of sirtuin SirT2, increases p21 (CDKN1A) expression in a p53 independent manner
    • McCarthy, A. R., et al. Tenovin D3, a novel small-molecule inhibitor of sirtuin SirT2, increases p21 (CDKN1A) expression in a p53 independent manner. Mol. Cancer Ther. 12, 352-360 (2013).
    • (2013) Mol. Cancer Ther. , vol.12 , pp. 352-360
    • McCarthy, A.R.1
  • 205
    • 84888375767 scopus 로고    scopus 로고
    • The role of p21waf1/cip1 and p27Kip1 in HDACi-mediated tumor cell death and cell cycle arrest in the Emu-myc model of B cell lymphoma
    • Newbold, A., Salmon, J. M., Martin, B. P., Stanley, K., & Johnstone, R. W. The role of p21waf1/cip1 and p27Kip1 in HDACi-mediated tumor cell death and cell cycle arrest in the Emu-myc model of B cell lymphoma. Oncogene 33, 5415-5423 (2014).
    • (2014) Oncogene , vol.33 , pp. 5415-5423
    • Newbold, A.1    Salmon, J.M.2    Martin, B.P.3    Stanley, K.4    Johnstone, R.W.5
  • 206
    • 35548936745 scopus 로고    scopus 로고
    • Structure-Activity studies on suramin analogues as inhibitors of NAD+-dependent histone deacetylases (sirtuins)
    • Trapp, J., et al. Structure-Activity studies on suramin analogues as inhibitors of NAD+-dependent histone deacetylases (sirtuins). ChemMedChem 2, 1419-1431 (2007).
    • (2007) ChemMedChem , vol.2 , pp. 1419-1431
    • Trapp, J.1
  • 208
    • 0034128221 scopus 로고    scopus 로고
    • Suramin therapy for patients with symptomatic hormone-refractory prostate cancer: Results of a randomized phase III trial comparing suramin plus hydrocortisone to placebo plus hydrocortisone
    • Small, E. J., et al. Suramin therapy for patients with symptomatic hormone-refractory prostate cancer: results of a randomized phase III trial comparing suramin plus hydrocortisone to placebo plus hydrocortisone. J. Clin. Oncol. 18, 1440-1450 (2000).
    • (2000) J. Clin. Oncol. , vol.18 , pp. 1440-1450
    • Small, E.J.1
  • 209
    • 33645221885 scopus 로고    scopus 로고
    • Inhibition of SIRT1 catalytic activity increases p53 acetylation but does not alter cell survival following DNA damage
    • Solomon, J. M., et al. Inhibition of SIRT1 catalytic activity increases p53 acetylation but does not alter cell survival following DNA damage. Mol. Cell. Biol. 26, 28-38 (2006).
    • (2006) Mol. Cell. Biol. , vol.26 , pp. 28-38
    • Solomon, J.M.1
  • 210
    • 34547599329 scopus 로고    scopus 로고
    • Sirtuin 2 inhibitors rescue alpha-synuclein-mediated toxicity in models of Parkinson?s disease
    • Outeiro, T. F., et al. Sirtuin 2 inhibitors rescue alpha-synuclein-mediated toxicity in models of Parkinson?s disease. Science 317, 516-519 (2007).
    • (2007) Science , vol.317 , pp. 516-519
    • Outeiro, T.F.1
  • 211
    • 0033519641 scopus 로고    scopus 로고
    • Structure and ligand of a histone acetyltransferase bromodomain
    • Dhalluin, C., et al. Structure and ligand of a histone acetyltransferase bromodomain. Nature 399, 491-496 (1999).
    • (1999) Nature , vol.399 , pp. 491-496
    • Dhalluin, C.1
  • 212
    • 0034717183 scopus 로고    scopus 로고
    • Structure and function of a human TAFII250 double bromodomain module
    • Jacobson, R. H., Ladurner, A. G., King, D. S., & Tjian, R. Structure and function of a human TAFII250 double bromodomain module. Science 288, 1422-1425 (2000).
    • (2000) Science , vol.288 , pp. 1422-1425
    • Jacobson, R.H.1    Ladurner, A.G.2    King, D.S.3    Tjian, R.4
  • 213
    • 0041806599 scopus 로고    scopus 로고
    • The double bromodomain protein Brd4 binds to acetylated chromatin during interphase and mitosis
    • Dey, A., Chitsaz, F., Abbasi, A., Misteli, T., & Ozato, K. The double bromodomain protein Brd4 binds to acetylated chromatin during interphase and mitosis. Proc. Natl Acad. Sci. USA 100, 8758-8763 (2003).
    • (2003) Proc. Natl Acad. Sci. USA , vol.100 , pp. 8758-8763
    • Dey, A.1    Chitsaz, F.2    Abbasi, A.3    Misteli, T.4    Ozato, K.5
  • 214
    • 73949087197 scopus 로고    scopus 로고
    • Brd4 marks select genes on mitotic chromatin and directs postmitotic transcription
    • Dey, A., Nishiyama, A., Karpova, T., McNally, J., & Ozato, K. Brd4 marks select genes on mitotic chromatin and directs postmitotic transcription. Mol. Biol. Cell 20, 4899-4909 (2009).
    • (2009) Mol. Biol. Cell , vol.20 , pp. 4899-4909
    • Dey, A.1    Nishiyama, A.2    Karpova, T.3    McNally, J.4    Ozato, K.5
  • 215
    • 80455122751 scopus 로고    scopus 로고
    • Gene bookmarking accelerates the kinetics of post-mitotic transcriptional re activation
    • Zhao, R., Nakamura, T., Fu, Y., Lazar, Z., & Spector, D. L. Gene bookmarking accelerates the kinetics of post-mitotic transcriptional re activation. Nat. Cell Biol. 13, 1295-1304 (2011).
    • (2011) Nat. Cell Biol. , vol.13 , pp. 1295-1304
    • Zhao, R.1    Nakamura, T.2    Fu, Y.3    Lazar, Z.4    Spector, D.L.5
  • 216
    • 70349669271 scopus 로고    scopus 로고
    • Cooperative binding of two acetylation marks on a histone tail by a single bromodomain
    • Morinière, J., et al. Cooperative binding of two acetylation marks on a histone tail by a single bromodomain. Nature 461, 664-668 (2009).
    • (2009) Nature , vol.461 , pp. 664-668
    • Morinière, J.1
  • 217
    • 84937639611 scopus 로고    scopus 로고
    • BET bromodomain inhibition suppresses the function of hematopoietic transcription factors in acute myeloid leukemia
    • Roe, J. S., Mercan, F., Rivera, K., Pappin, D. J., & Vakoc, C. R. BET bromodomain inhibition suppresses the function of hematopoietic transcription factors in acute myeloid leukemia. Mol. Cell 58, 1028-1039 (2015).
    • (2015) Mol. Cell , vol.58 , pp. 1028-1039
    • Roe, J.S.1    Mercan, F.2    Rivera, K.3    Pappin, D.J.4    Vakoc, C.R.5
  • 218
    • 41649120757 scopus 로고    scopus 로고
    • BRD-NUT oncoproteins: A family of closely related nuclear proteins that block epithelial differentiation and maintain the growth of carcinoma cells
    • French, C. A., et al. BRD-NUT oncoproteins: a family of closely related nuclear proteins that block epithelial differentiation and maintain the growth of carcinoma cells. Oncogene 27, 2237-2242 (2008).
    • (2008) Oncogene , vol.27 , pp. 2237-2242
    • French, C.A.1
  • 219
    • 78650847770 scopus 로고    scopus 로고
    • Selective inhibition of BET bromodomains
    • Filippakopoulos, P., et al. Selective inhibition of BET bromodomains. Nature 468, 1067-1073 (2010).
    • (2010) Nature , vol.468 , pp. 1067-1073
    • Filippakopoulos, P.1
  • 220
    • 0021949612 scopus 로고
    • Alprazolam: Review of pharmacological, pharmacokinetic, and clinical data
    • VonVoigtlander, P. F., & Straw, R. N. Alprazolam: review of pharmacological, pharmacokinetic, and clinical data. Drug Dev. Res. 6, 1-12 (1985).
    • (1985) Drug Dev. Res. , vol.6 , pp. 1-12
    • Von Voigtlander, P.F.1    Straw, R.N.2
  • 221
    • 84857913614 scopus 로고    scopus 로고
    • Benzodiazepines and benzotriazepines as protein interaction inhibitors targeting bromodomains of the BET family
    • Filippakopoulos, P., et al. Benzodiazepines and benzotriazepines as protein interaction inhibitors targeting bromodomains of the BET family. Bioorg. Med. Chem. 20, 1878-1886 (2012).
    • (2012) Bioorg. Med. Chem. , vol.20 , pp. 1878-1886
    • Filippakopoulos, P.1
  • 222
    • 84892164371 scopus 로고    scopus 로고
    • Genome-wide localization of small molecules
    • Anders, L., et al. Genome-wide localization of small molecules. Nat. Biotechnol. 32, 92-96 (2014).
    • (2014) Nat. Biotechnol. , vol.32 , pp. 92-96
    • Anders, L.1
  • 223
    • 80055000824 scopus 로고    scopus 로고
    • RNAi screen identifies Brd4 as a therapeutic target in acute myeloid leukaemia
    • Zuber, J., et al. RNAi screen identifies Brd4 as a therapeutic target in acute myeloid leukaemia. Nature 478, 524-528 (2011).
    • (2011) Nature , vol.478 , pp. 524-528
    • Zuber, J.1
  • 224
    • 80054984945 scopus 로고    scopus 로고
    • Inhibition of BET recruitment to chromatin as an effective treatment for MLL-fusion leukaemia
    • Dawson, M. A., et al. Inhibition of BET recruitment to chromatin as an effective treatment for MLL-fusion leukaemia. Nature 478, 529-533 (2011).
    • (2011) Nature , vol.478 , pp. 529-533
    • Dawson, M.A.1
  • 225
    • 23744467035 scopus 로고    scopus 로고
    • Recruitment of P TEFb for stimulation of transcriptional elongation by the bromodomain protein Brd4
    • Yang, Z., et al. Recruitment of P TEFb for stimulation of transcriptional elongation by the bromodomain protein Brd4. Mol. Cell 19, 535-545 (2005).
    • (2005) Mol. Cell , vol.19 , pp. 535-545
    • Yang, Z.1
  • 226
    • 38549113034 scopus 로고    scopus 로고
    • Brd4 recruits P TEFb to chromosomes at late mitosis to promote G1 gene expression and cell cycle progression
    • Yang, Z., He, N., & Zhou, Q. Brd4 recruits P TEFb to chromosomes at late mitosis to promote G1 gene expression and cell cycle progression. Mol. Cell. Biol. 28, 967-976 (2008).
    • (2008) Mol. Cell. Biol. , vol.28 , pp. 967-976
    • Yang, Z.1    He, N.2    Zhou, Q.3
  • 227
    • 77249119762 scopus 로고    scopus 로고
    • The landscape of somatic copy-number alteration across human cancers
    • Beroukhim, R., et al. The landscape of somatic copy-number alteration across human cancers. Nature 463, 899-905 (2010).
    • (2010) Nature , vol.463 , pp. 899-905
    • Beroukhim, R.1
  • 228
    • 77951920690 scopus 로고    scopus 로고
    • C Myc regulates transcriptional pause release
    • Rahl, P. B., et al. c Myc regulates transcriptional pause release. Cell 141, 432-445 (2010).
    • (2010) Cell , vol.141 , pp. 432-445
    • Rahl, P.B.1
  • 229
    • 80052955256 scopus 로고    scopus 로고
    • BET bromodomain inhibition as a therapeutic strategy to target c Myc
    • Delmore, J. E., et al. BET bromodomain inhibition as a therapeutic strategy to target c Myc. Cell 146, 904-917 (2011).
    • (2011) Cell , vol.146 , pp. 904-917
    • Delmore, J.E.1
  • 230
    • 80053651202 scopus 로고    scopus 로고
    • Targeting MYC dependence in cancer by inhibiting BET bromodomains
    • Mertz, J. A., et al. Targeting MYC dependence in cancer by inhibiting BET bromodomains. Proc. Natl Acad. Sci. USA 108, 16669-16674 (2011).
    • (2011) Proc. Natl Acad. Sci. USA , vol.108 , pp. 16669-16674
    • Mertz, J.A.1
  • 231
    • 84867316537 scopus 로고    scopus 로고
    • BET bromodomain inhibition targets both c Myc and IL7R in high-risk acute lymphoblastic leukemia
    • Ott, C. J., et al. BET bromodomain inhibition targets both c Myc and IL7R in high-risk acute lymphoblastic leukemia. Blood 120, 2843-2852 (2012).
    • (2012) Blood , vol.120 , pp. 2843-2852
    • Ott, C.J.1
  • 232
    • 84876222028 scopus 로고    scopus 로고
    • Selective inhibition of tumor oncogenes by disruption of super-enhancers
    • Lovén, J., et al. Selective inhibition of tumor oncogenes by disruption of super-enhancers. Cell 153, 320-334 (2013).
    • (2013) Cell , vol.153 , pp. 320-334
    • Lovén, J.1
  • 233
    • 0001087931 scopus 로고
    • Extreme instability of myc mRNA in normal and transformed human cells
    • Dani, C., et al. Extreme instability of myc mRNA in normal and transformed human cells. Proc. Natl Acad. Sci. USA 81, 7046-7050 (1984).
    • (1984) Proc. Natl Acad. Sci. USA , vol.81 , pp. 7046-7050
    • Dani, C.1
  • 234
    • 84891953718 scopus 로고    scopus 로고
    • Discovery and characterization of super-enhancer-Associated dependencies in diffuse large B cell lymphoma
    • Chapuy, B., et al. Discovery and characterization of super-enhancer-Associated dependencies in diffuse large B cell lymphoma. Cancer Cell 24, 777-790 (2013).
    • (2013) Cancer Cell , vol.24 , pp. 777-790
    • Chapuy, B.1
  • 235
    • 84899927814 scopus 로고    scopus 로고
    • Brd4 maintains constitutively active NF kappaB in cancer cells by binding to acetylated RelA
    • Zou, Z., et al. Brd4 maintains constitutively active NF kappaB in cancer cells by binding to acetylated RelA. Oncogene 33, 2395-2404 (2014).
    • (2014) Oncogene , vol.33 , pp. 2395-2404
    • Zou, Z.1
  • 236
    • 84931560527 scopus 로고    scopus 로고
    • Hijacking the E3 ubiquitin ligase cereblon to efficiently target BRD4
    • Lu, J., et al. Hijacking the E3 ubiquitin ligase cereblon to efficiently target BRD4. Chem. Biol. 22, 755-763 (2015).
    • (2015) Chem. Biol. , vol.22 , pp. 755-763
    • Lu, J.1
  • 237
    • 79958078949 scopus 로고    scopus 로고
    • Discovery and characterization of small molecule inhibitors of the BET family bromodomains
    • Chung, C. W., et al. Discovery and characterization of small molecule inhibitors of the BET family bromodomains. J. Med. Chem. 54, 3827-3838 (2011).
    • (2011) J. Med. Chem. , vol.54 , pp. 3827-3838
    • Chung, C.W.1
  • 238
    • 80053924872 scopus 로고    scopus 로고
    • 3, 5 Dimethylisoxazoles act as acetyl-lysine-mimetic bromodomain ligands
    • Hewings, D. S., et al. 3, 5 Dimethylisoxazoles act as acetyl-lysine-mimetic bromodomain ligands. J. Med. Chem. 54, 6761-6770 (2011).
    • (2011) J. Med. Chem. , vol.54 , pp. 6761-6770
    • Hewings, D.S.1
  • 239
    • 79954552505 scopus 로고    scopus 로고
    • The super elongation complex (SEC) and MLL in development and disease
    • Smith, E., Lin, C., & Shilatifard, A. The super elongation complex (SEC) and MLL in development and disease. Genes Dev. 25, 661-672 (2011).
    • (2011) Genes Dev. , vol.25 , pp. 661-672
    • Smith, E.1    Lin, C.2    Shilatifard, A.3
  • 240
    • 77953246813 scopus 로고    scopus 로고
    • The PAF complex synergizes with MLL fusion proteins at HOX loci to promote leukemogenesis
    • Muntean, A. G., et al. The PAF complex synergizes with MLL fusion proteins at HOX loci to promote leukemogenesis. Cancer Cell 17, 609-621 (2010).
    • (2010) Cancer Cell , vol.17 , pp. 609-621
    • Muntean, A.G.1
  • 241
    • 84878565710 scopus 로고    scopus 로고
    • PFI 1, a highly selective protein interaction inhibitor, targeting BET bromodomains
    • Picaud, S., et al. PFI 1, a highly selective protein interaction inhibitor, targeting BET bromodomains. Cancer Res. 73, 3336-3346 (2013).
    • (2013) Cancer Res. , vol.73 , pp. 3336-3346
    • Picaud, S.1
  • 242
    • 84896611007 scopus 로고    scopus 로고
    • Dual kinase-bromodomain inhibitors for rationally designed polypharmacology
    • Ciceri, P., et al. Dual kinase-bromodomain inhibitors for rationally designed polypharmacology. Nat. Chem. Biol. 10, 305-312 (2014).
    • (2014) Nat. Chem. Biol. , vol.10 , pp. 305-312
    • Ciceri, P.1
  • 243
    • 84961933254 scopus 로고    scopus 로고
    • Safety, tolerability, and preliminary activity of CUDC 907, a first in class, oral, dual inhibitor of HDAC and PI3K, in patients with relapsed or refractory lymphoma or multiple myeloma: An open-label, dose-escalation, phase 1 trial
    • Younes, A., et al. Safety, tolerability, and preliminary activity of CUDC 907, a first in class, oral, dual inhibitor of HDAC and PI3K, in patients with relapsed or refractory lymphoma or multiple myeloma: an open-label, dose-escalation, phase 1 trial. Lancet Oncol. 17, 622-631 (2016).
    • (2016) Lancet Oncol. , vol.17 , pp. 622-631
    • Younes, A.1
  • 244
    • 84964691616 scopus 로고    scopus 로고
    • IDH mutations in cancer and progress toward development of targeted therapeutics
    • Dang, L., Yen, K., & Attar, E. C. IDH mutations in cancer and progress toward development of targeted therapeutics. Ann. Oncol. 27, 599-608 (2016).
    • (2016) Ann. Oncol. , vol.27 , pp. 599-608
    • Dang, L.1    Yen, K.2    Attar, E.C.3
  • 245
    • 65349196261 scopus 로고    scopus 로고
    • Constrained (l-)-S adenosyl-l homocysteine (SAH) analogues as DNA methyltransferase inhibitors
    • Isakovic, L., et al. Constrained (l-)-S adenosyl-l homocysteine (SAH) analogues as DNA methyltransferase inhibitors. Bioorg. Med. Chem. Lett. 19, 2742-2746 (2009).
    • (2009) Bioorg. Med. Chem. Lett. , vol.19 , pp. 2742-2746
    • Isakovic, L.1
  • 246
    • 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
    • Jüttermann, R., Li, E., & Jaenisch, R. 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, 11797-11801 (1994).
    • (1994) Proc. Natl Acad. Sci. USA , vol.91 , pp. 11797-11801
    • Jüttermann, R.1    Li, E.2    Jaenisch, R.3
  • 247
    • 0020582853 scopus 로고
    • On the mechanism of inhibition of DNA-cytosine methyltransferases by cytosine analogs
    • Santi, D. V., Garrett, C. E., & Barr, P. J. On the mechanism of inhibition of DNA-cytosine methyltransferases by cytosine analogs. Cell 33, 9-10 (1983).
    • (1983) Cell , vol.33 , pp. 9-10
    • Santi, D.V.1    Garrett, C.E.2    Barr, P.J.3
  • 248
    • 22144461094 scopus 로고    scopus 로고
    • Mechanism of inhibition of DNA methyltransferases by cytidine analogs in cancer therapy
    • Gowher, H., & Jeltsch, A. Mechanism of inhibition of DNA methyltransferases by cytidine analogs in cancer therapy. Cancer Biol. Ther. 3, 1062-1068 (2004).
    • (2004) Cancer Biol. Ther. , vol.3 , pp. 1062-1068
    • Gowher, H.1    Jeltsch, A.2
  • 249
    • 0014872738 scopus 로고
    • Cytotoxicity and mode of action of 5 azacytidine on L1210 leukemia
    • Li, L. H., Olin, E. J., Buskirk, H. H., & Reineke, L. M. Cytotoxicity and mode of action of 5 azacytidine on L1210 leukemia. Cancer Res. 30, 2760-2769 (1970).
    • (1970) Cancer Res. , vol.30 , pp. 2760-2769
    • Li, L.H.1    Olin, E.J.2    Buskirk, H.H.3    Reineke, L.M.4
  • 250
    • 84861892178 scopus 로고    scopus 로고
    • RNA-dependent inhibition of ribonucleotide reductase is a major pathway for 5 azacytidine activity in acute myeloid leukemia
    • Aimiuwu, J., et al. RNA-dependent inhibition of ribonucleotide reductase is a major pathway for 5 azacytidine activity in acute myeloid leukemia. Blood 119, 5229-5238 (2012).
    • (2012) Blood , vol.119 , pp. 5229-5238
    • Aimiuwu, J.1
  • 251
    • 70350234722 scopus 로고    scopus 로고
    • Azacytidine inhibits RNA methylation at DNMT2 target sites in human cancer cell lines
    • Schaefer, M., Hagemann, S., Hanna, K., & Lyko, F. Azacytidine inhibits RNA methylation at DNMT2 target sites in human cancer cell lines. Cancer Res. 69, 8127-8132 (2009).
    • (2009) Cancer Res. , vol.69 , pp. 8127-8132
    • Schaefer, M.1    Hagemann, S.2    Hanna, K.3    Lyko, F.4
  • 252
    • 33646071894 scopus 로고    scopus 로고
    • Decitabine improves patient outcomes in myelodysplastic syndromes: Results of a phase III randomized study
    • Kantarjian, H., et al. Decitabine improves patient outcomes in myelodysplastic syndromes: results of a phase III randomized study. Cancer 106, 1794-1803 (2006).
    • (2006) Cancer , vol.106 , pp. 1794-1803
    • Kantarjian, H.1
  • 253
    • 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., 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. 10, 223-232 (2009).
    • (2009) Lancet Oncol. , vol.10 , pp. 223-232
    • Fenaux, P.1
  • 254
    • 77952203695 scopus 로고    scopus 로고
    • S110, a 5 aza 2? Deoxycytidine-containing dinucleotide, is an effective DNA methylation inhibitor in vivo and can reduce tumor growth
    • Chuang, J. C., et al. S110, a 5 aza 2? deoxycytidine-containing dinucleotide, is an effective DNA methylation inhibitor in vivo and can reduce tumor growth. Mol. Cancer Ther. 9, 1443-1450 (2010).
    • (2010) Mol. Cancer Ther. , vol.9 , pp. 1443-1450
    • Chuang, J.C.1
  • 255
    • 84908247072 scopus 로고    scopus 로고
    • TET2 mutations predict response to hypomethylating agents in myelodysplastic syndrome patients
    • Bejar, R., et al. TET2 mutations predict response to hypomethylating agents in myelodysplastic syndrome patients. Blood 124, 2705-2712 (2014).
    • (2014) Blood , vol.124 , pp. 2705-2712
    • Bejar, R.1
  • 256
    • 0022453948 scopus 로고
    • Studies of mutagenicity and clastogenicity of 5 azacytidine in human lymphoblasts and Salmonella typhimurium
    • Call, K. M., Jensen, J. C., Liber, H. L., & Thilly, W. G. Studies of mutagenicity and clastogenicity of 5 azacytidine in human lymphoblasts and Salmonella typhimurium. Mutat. Res. 160, 249-257 (1986).
    • (1986) Mutat. Res. , vol.160 , pp. 249-257
    • Call, K.M.1    Jensen, J.C.2    Liber, H.L.3    Thilly, W.G.4
  • 257
    • 0023936754 scopus 로고
    • Hydralazine and procainamide inhibit T cell DNA methylation and induce autoreactivity
    • Cornacchia, E., et al. Hydralazine and procainamide inhibit T cell DNA methylation and induce autoreactivity. J. Immunol. 140, 2197-2200 (1988).
    • (1988) J. Immunol. , vol.140 , pp. 2197-2200
    • Cornacchia, E.1
  • 258
    • 76449100936 scopus 로고    scopus 로고
    • Novel and selective DNA methyltransferase inhibitors: Docking-based virtual screening and experimental evaluation
    • Kuck, D., Singh, N., Lyko, F., & Medina-Franco, J. L. Novel and selective DNA methyltransferase inhibitors: docking-based virtual screening and experimental evaluation. Bioorg. Med. Chem. 18, 822-829 (2010).
    • (2010) Bioorg. Med. Chem. , vol.18 , pp. 822-829
    • Kuck, D.1    Singh, N.2    Lyko, F.3    Medina-Franco, J.L.4
  • 259
    • 31544439666 scopus 로고    scopus 로고
    • Discovery of two novel, small-molecule inhibitors of DNA methylation
    • Siedlecki, P., et al. Discovery of two novel, small-molecule inhibitors of DNA methylation. J. Med. Chem. 49, 678-683 (2006).
    • (2006) J. Med. Chem. , vol.49 , pp. 678-683
    • Siedlecki, P.1
  • 260
    • 66249127951 scopus 로고    scopus 로고
    • A new class of quinoline-based DNA hypomethylating agents reactivates tumor suppressor genes by blocking DNA methyltransferase 1 activity and inducing its degradation
    • Datta, J., et al. A new class of quinoline-based DNA hypomethylating agents reactivates tumor suppressor genes by blocking DNA methyltransferase 1 activity and inducing its degradation. Cancer Res. 69, 4277-4285 (2009).
    • (2009) Cancer Res. , vol.69 , pp. 4277-4285
    • Datta, J.1
  • 261
    • 22244435605 scopus 로고    scopus 로고
    • Epigenetic reactivation of tumor suppressor genes by a novel small-molecule inhibitor of human DNA methyltransferases
    • Brueckner, B., et al. Epigenetic reactivation of tumor suppressor genes by a novel small-molecule inhibitor of human DNA methyltransferases. Cancer Res. 65, 6305-6311 (2005).
    • (2005) Cancer Res. , vol.65 , pp. 6305-6311
    • Brueckner, B.1
  • 262
    • 84856990498 scopus 로고    scopus 로고
    • Structure-based mechanistic insights into DNMT1 mediated maintenance DNA methylation
    • Song, J., Teplova, M., Ishibe-Murakami, S., & Patel, D. J. Structure-based mechanistic insights into DNMT1 mediated maintenance DNA methylation. Science 335, 709-712 (2012).
    • (2012) Science , vol.335 , pp. 709-712
    • Song, J.1    Teplova, M.2    Ishibe-Murakami, S.3    Patel, D.J.4
  • 263
    • 0019190864 scopus 로고
    • Hydralazine-pyrimidine interactions may explain hydralazine-induced lupus erythematosus
    • Dubroff, L. M., & Reid, R. J. Jr. Hydralazine-pyrimidine interactions may explain hydralazine-induced lupus erythematosus. Science 208, 404-406 (1980).
    • (1980) Science , vol.208 , pp. 404-406
    • Dubroff, L.M.1    Reid, R.J.2
  • 264
    • 28844497663 scopus 로고    scopus 로고
    • Procainamide is a specific inhibitor of DNA methyltransferase 1
    • Lee, B. H., Yegnasubramanian, S., Lin, X., & Nelson, W. G. Procainamide is a specific inhibitor of DNA methyltransferase 1. J. Biol. Chem. 280, 40749-40756 (2005).
    • (2005) J. Biol. Chem. , vol.280 , pp. 40749-40756
    • Lee, B.H.1    Yegnasubramanian, S.2    Lin, X.3    Nelson, W.G.4
  • 265
    • 10744225448 scopus 로고    scopus 로고
    • Reactivation of tumor suppressor genes by the cardiovascular drugs hydralazine and procainamide and their potential use in cancer therapy
    • Segura-Pacheco, B., et al. Reactivation of tumor suppressor genes by the cardiovascular drugs hydralazine and procainamide and their potential use in cancer therapy. Clin. Cancer Res. 9, 1596-1603 (2003).
    • (2003) Clin. Cancer Res. , vol.9 , pp. 1596-1603
    • Segura-Pacheco, B.1
  • 266
    • 80053383964 scopus 로고    scopus 로고
    • Molecular dynamics simulations of human DNA methyltransferase 3B with selective inhibitor nanaomycin A
    • Caulfield, T., & Medina-Franco, J. L. Molecular dynamics simulations of human DNA methyltransferase 3B with selective inhibitor nanaomycin A. J. Struct. Biol. 176, 185-191 (2011).
    • (2011) J. Struct. Biol. , vol.176 , pp. 185-191
    • Caulfield, T.1    Medina-Franco, J.L.2
  • 267
    • 77956153243 scopus 로고    scopus 로고
    • The language of histone crosstalk
    • Lee, J. S., Smith, E., & Shilatifard, A. The language of histone crosstalk. Cell 142, 682-685 (2010).
    • (2010) Cell , vol.142 , pp. 682-685
    • Lee, J.S.1    Smith, E.2    Shilatifard, A.3
  • 268
    • 84879341188 scopus 로고    scopus 로고
    • A dual role for Hdac1: Oncosuppressor in tumorigenesis, oncogene in tumor maintenance
    • Santoro, F., et al. A dual role for Hdac1: oncosuppressor in tumorigenesis, oncogene in tumor maintenance. Blood 121, 3459-3468 (2013).
    • (2013) Blood , vol.121 , pp. 3459-3468
    • Santoro, F.1
  • 269
    • 84877609321 scopus 로고    scopus 로고
    • Dosage-dependent tumor suppression by histone deacetylases 1 and 2 through regulation of c Myc collaborating genes and p53 function
    • Heideman, M. R., et al. Dosage-dependent tumor suppression by histone deacetylases 1 and 2 through regulation of c Myc collaborating genes and p53 function. Blood 121, 2038-2050 (2013).
    • (2013) Blood , vol.121 , pp. 2038-2050
    • Heideman, M.R.1
  • 270
    • 0034234237 scopus 로고    scopus 로고
    • CBP/p300 in cell growth, transformation, and development
    • Goodman, R. H., & Smolik, S. CBP/p300 in cell growth, transformation, and development. Genes Dev. 14, 1553-1577 (2000).
    • (2000) Genes Dev. , vol.14 , pp. 1553-1577
    • Goodman, R.H.1    Smolik, S.2
  • 271
    • 0030797585 scopus 로고    scopus 로고
    • Activation of p53 sequence-specific DNA binding by acetylation of the p53 C terminal domain
    • Gu, W., & Roeder, R. G. Activation of p53 sequence-specific DNA binding by acetylation of the p53 C terminal domain. Cell 90, 595-606 (1997).
    • (1997) Cell , vol.90 , pp. 595-606
    • Gu, W.1    Roeder, R.G.2
  • 272
    • 77949893772 scopus 로고    scopus 로고
    • Tip60 promotes prostate cancer cell proliferation by translocation of androgen receptor into the nucleus
    • Shiota, M., et al. Tip60 promotes prostate cancer cell proliferation by translocation of androgen receptor into the nucleus. Prostate 70, 540-554 (2010).
    • (2010) Prostate , vol.70 , pp. 540-554
    • Shiota, M.1
  • 273
    • 34548312368 scopus 로고    scopus 로고
    • Tip60 is a haplo-insufficient tumour suppressor required for an oncogene-induced DNA damage response
    • Gorrini, C., et al. Tip60 is a haplo-insufficient tumour suppressor required for an oncogene-induced DNA damage response. Nature 448, 1063-1067 (2007).
    • (2007) Nature , vol.448 , pp. 1063-1067
    • Gorrini, C.1
  • 274
    • 84896711459 scopus 로고    scopus 로고
    • Second primary malignancies with lenalidomide therapy for newly diagnosed myeloma: A meta-Analysis of individual patient data
    • Palumbo, A., et al. Second primary malignancies with lenalidomide therapy for newly diagnosed myeloma: a meta-Analysis of individual patient data. Lancet Oncol. 15, 333-342 (2014).
    • (2014) Lancet Oncol. , vol.15 , pp. 333-342
    • Palumbo, A.1
  • 275
    • 84908265816 scopus 로고    scopus 로고
    • Histone deacetylases and their inhibitors in cancer, neurological diseases and immune disorders
    • Falkenberg, K. J., & Johnstone, R. W. Histone deacetylases and their inhibitors in cancer, neurological diseases and immune disorders. Nat. Rev. Drug Discov. 13, 673-691 (2014).
    • (2014) Nat. Rev. Drug Discov. , vol.13 , pp. 673-691
    • Falkenberg, K.J.1    Johnstone, R.W.2
  • 276
    • 84866183822 scopus 로고    scopus 로고
    • HDAC8 mutations in Cornelia de Lange syndrome affect the cohesin acetylation cycle
    • Deardorff, M. A., et al. HDAC8 mutations in Cornelia de Lange syndrome affect the cohesin acetylation cycle. Nature 489, 313-317 (2012).
    • (2012) Nature , vol.489 , pp. 313-317
    • Deardorff, M.A.1
  • 277
    • 84890080778 scopus 로고    scopus 로고
    • The up regulation of histone deacetylase 8 promotes proliferation and inhibits apoptosis in hepatocellular carcinoma
    • Wu, J., et al. The up regulation of histone deacetylase 8 promotes proliferation and inhibits apoptosis in hepatocellular carcinoma. Dig. Dis. Sci. 58, 3545-3553 (2013).
    • (2013) Dig. Dis. Sci. , vol.58 , pp. 3545-3553
    • Wu, J.1
  • 278
    • 77954831404 scopus 로고    scopus 로고
    • An acetylation switch modulates the transcriptional activity of estrogen-related receptor alpha
    • Wilson, B. J., Tremblay, A. M., Deblois, G., Sylvain-Drolet, G., & Giguère, V. An acetylation switch modulates the transcriptional activity of estrogen-related receptor alpha. Mol. Endocrinol. 24, 1349-1358 (2010).
    • (2010) Mol. Endocrinol. , vol.24 , pp. 1349-1358
    • Wilson, B.J.1    Tremblay, A.M.2    Deblois, G.3    Sylvain-Drolet, G.4    Giguère, V.5
  • 279
    • 43749109171 scopus 로고    scopus 로고
    • A novel histone deacetylase 8 (HDAC8)-specific inhibitor PCI 34051 induces apoptosis in T cell lymphomas
    • Balasubramanian, S., et al. A novel histone deacetylase 8 (HDAC8)-specific inhibitor PCI 34051 induces apoptosis in T cell lymphomas. Leukemia 22, 1026-1034 (2008).
    • (2008) Leukemia , vol.22 , pp. 1026-1034
    • Balasubramanian, S.1
  • 280
    • 84961291039 scopus 로고    scopus 로고
    • Selective inhibition of HDAC8 decreases neuroblastoma growth in vitro and in vivo and enhances retinoic acid-mediated differentiation
    • Rettig, I., et al. Selective inhibition of HDAC8 decreases neuroblastoma growth in vitro and in vivo and enhances retinoic acid-mediated differentiation. Cell Death Dis. 6, e1657 (2015).
    • (2015) Cell Death Dis. , vol.6 , pp. e1657
    • Rettig, I.1
  • 281
    • 84873173538 scopus 로고    scopus 로고
    • Histone deacetylase 6 plays a role as a distinct regulator of diverse cellular processes
    • Li, Y., Shin, D., & Kwon, S. H. Histone deacetylase 6 plays a role as a distinct regulator of diverse cellular processes. FEBS J. 280, 775-793 (2013).
    • (2013) FEBS J. , vol.280 , pp. 775-793
    • Li, Y.1    Shin, D.2    Kwon, S.H.3
  • 282
    • 22844432021 scopus 로고    scopus 로고
    • Inhibition of histone deacetylase 6 acetylates and disrupts the chaperone function of heat shock protein 90: A novel basis for antileukemia activity of histone deacetylase inhibitors
    • Bali, P., et al. Inhibition of histone deacetylase 6 acetylates and disrupts the chaperone function of heat shock protein 90: a novel basis for antileukemia activity of histone deacetylase inhibitors. J. Biol. Chem. 280, 26729-26734 (2005).
    • (2005) J. Biol. Chem. , vol.280 , pp. 26729-26734
    • Bali, P.1
  • 283
    • 0346020435 scopus 로고    scopus 로고
    • The deacetylase HDAC6 regulates aggresome formation and cell viability in response to misfolded protein stress
    • Kawaguchi, Y., et al. The deacetylase HDAC6 regulates aggresome formation and cell viability in response to misfolded protein stress. Cell 115, 727-738 (2003).
    • (2003) Cell , vol.115 , pp. 727-738
    • Kawaguchi, Y.1
  • 284
    • 34548416641 scopus 로고    scopus 로고
    • HDAC6 controls major cell response pathways to cytotoxic accumulation of protein aggregates
    • Boyault, C., et al. HDAC6 controls major cell response pathways to cytotoxic accumulation of protein aggregates. Genes Dev. 21, 2172-2181 (2007).
    • (2007) Genes Dev. , vol.21 , pp. 2172-2181
    • Boyault, C.1
  • 285
    • 0344640906 scopus 로고    scopus 로고
    • Domain-selective small-molecule inhibitor of histone deacetylase 6 (HDAC6)-mediated tubulin deacetylation
    • Haggarty, S. J., Koeller, K. M., Wong, J. C., Grozinger, C. M., & Schreiber, S. L. Domain-selective small-molecule inhibitor of histone deacetylase 6 (HDAC6)-mediated tubulin deacetylation. Proc. Natl Acad. Sci. USA 100, 4389-4394 (2003).
    • (2003) Proc. Natl Acad. Sci. USA , vol.100 , pp. 4389-4394
    • Haggarty, S.J.1    Koeller, K.M.2    Wong, J.C.3    Grozinger, C.M.4    Schreiber, S.L.5
  • 286
    • 84858640254 scopus 로고    scopus 로고
    • Preclinical activity, pharmacodynamic, and pharmacokinetic properties of a selective HDAC6 inhibitor, ACY 1215, in combination with bortezomib in multiple myeloma
    • Santo, L., et al. Preclinical activity, pharmacodynamic, and pharmacokinetic properties of a selective HDAC6 inhibitor, ACY 1215, in combination with bortezomib in multiple myeloma. Blood 119, 2579-2589 (2012).
    • (2012) Blood , vol.119 , pp. 2579-2589
    • Santo, L.1
  • 287
    • 84984679985 scopus 로고    scopus 로고
    • ClinicalTrials.gov
    • US National Library of Medicine. ClinicalTrials.gov https://clinicaltrials.gov/ct2/show/NCT01684150 (2016).
    • (2016) US National Library of Medicine.
  • 288
    • 84984679985 scopus 로고    scopus 로고
    • ClinicalTrials.gov
    • US National Library of Medicine. ClinicalTrials.gov https://clinicaltrials.gov/ct2/show/NCT02082977 (2016).
    • (2016) US National Library of Medicine.
  • 289
    • 84984679985 scopus 로고    scopus 로고
    • ClinicalTrials.gov
    • US National Library of Medicine. ClinicalTrials.gov https://clinicaltrials.gov/ct2/show/NCT01897571 (2016).
    • (2016) US National Library of Medicine.
  • 290
    • 84984679985 scopus 로고    scopus 로고
    • ClinicalTrials.gov
    • US National Library of Medicine. ClinicalTrials.gov https://clinicaltrials.gov/ct2/show/NCT02395601 (2016).
    • (2016) US National Library of Medicine.
  • 291
    • 84984679985 scopus 로고    scopus 로고
    • ClinicalTrials.gov
    • US National Library of Medicine. ClinicalTrials.gov https://clinicaltrials.gov/ct2/show/NCT02273102 (2016).
    • (2016) US National Library of Medicine.
  • 292
    • 84984679985 scopus 로고    scopus 로고
    • ClinicalTrials.gov.
    • US National Library of Medicine. ClinicalTrials.gov. https://clinicaltrials.gov/ct2/show/NCT02177812 (2016).
    • (2016) US National Library of Medicine.
  • 293
    • 85013674820 scopus 로고    scopus 로고
    • ClinicalTrialsRegister.eu
    • EU Clinical Trials Register. ClinicalTrialsRegister.eu https://www.clinicaltrialsregister.eu/ctr-search/ trial/2013-002447-29/ES (2013).
    • (2013) EU Clinical Trials Register.
  • 294
    • 84984679985 scopus 로고    scopus 로고
    • ClinicalTrials.gov
    • US National Library of Medicine. ClinicalTrials.gov https://clinicaltrials.gov/ct2/show/NCT01713582 (2016).
    • (2016) US National Library of Medicine.
  • 295
    • 84984679985 scopus 로고    scopus 로고
    • ClinicalTrials.gov
    • US National Library of Medicine. ClinicalTrials.gov https://clinicaltrials.gov/ct2/show/NCT01949883 (2016).
    • (2016) US National Library of Medicine.
  • 296
    • 84984679985 scopus 로고    scopus 로고
    • ClinicalTrials.gov
    • US National Library of Medicine. ClinicalTrials.gov https://clinicaltrials.gov/ct2/show/NCT02419417 (2016).
    • (2016) US National Library of Medicine.
  • 297
    • 84984679985 scopus 로고    scopus 로고
    • ClinicalTrials.gov
    • US National Library of Medicine. ClinicalTrials.gov https://clinicaltrials.gov/ct2/show/NCT01587703 (2016).
    • (2016) US National Library of Medicine.
  • 298
    • 84984679985 scopus 로고    scopus 로고
    • ClinicalTrials.gov
    • US National Library of Medicine. ClinicalTrials.gov https://clinicaltrials.gov/ct2/show/NCT02391480 (2016).
    • (2016) US National Library of Medicine.
  • 299
    • 84984679985 scopus 로고    scopus 로고
    • ClinicalTrials.gov
    • US National Library of Medicine. ClinicalTrials.gov https://clinicaltrials.gov/ct2/show/NCT02308761 (2016).
    • (2016) US National Library of Medicine.
  • 300
    • 84984679985 scopus 로고    scopus 로고
    • ClinicalTrials.gov
    • US National Library of Medicine. ClinicalTrials.gov https://clinicaltrials.gov/ct2/show/NCT02468687 (2016).
    • (2016) US National Library of Medicine.
  • 301
    • 84940562761 scopus 로고    scopus 로고
    • Belinostat in patients with relapsed or refractory peripheral T cell lymphoma: Results of the pivotal Phase II BELIEF (CLN 19) Study
    • O?Connor, O. A., et al. Belinostat in patients with relapsed or refractory peripheral T cell lymphoma: results of the pivotal Phase II BELIEF (CLN 19) Study. J. Clin. Oncol. 33, 2492-2499 (2015).
    • (2015) J. Clin. Oncol. , vol.33 , pp. 2492-2499
    • O'Connor, O.A.1
  • 302
    • 84908133944 scopus 로고    scopus 로고
    • Panobinostat plus bortezomib and dexamethasone versus placebo plus bortezomib and dexamethasone in patients with relapsed or relapsed and refractory multiple myeloma: A multicentre, randomised, double-blind phase 3 trial
    • San-Miguel, J. F., et al. Panobinostat plus bortezomib and dexamethasone versus placebo plus bortezomib and dexamethasone in patients with relapsed or relapsed and refractory multiple myeloma: a multicentre, randomised, double-blind phase 3 trial. Lancet Oncol. 15, 1195-1206 (2014).
    • (2014) Lancet Oncol. , vol.15 , pp. 1195-1206
    • San-Miguel, J.F.1


* 이 정보는 Elsevier사의 SCOPUS DB에서 KISTI가 분석하여 추출한 것입니다.