메뉴 건너뛰기




Volumn 8, Issue 1, 2016, Pages 119-133

Epigenome-based personalized medicine in human cancer

Author keywords

DNA methylation; epigenome; histone modification; personalized medicine; targeted therapy

Indexed keywords

ALKYLATING AGENT; ANTINEOPLASTIC AGENT; AZACITIDINE; BROMODOMAIN INHIBITOR; DECITABINE; DOT1L INHIBITOR; EZH2 INHIBITOR; HISTONE DEACETYLASE INHIBITOR; TAZEMETOSTAT; UNCLASSIFIED DRUG;

EID: 84954520555     PISSN: 17501911     EISSN: 1750192X     Source Type: Journal    
DOI: 10.2217/epi.15.84     Document Type: Review
Times cited : (80)

References (160)
  • 1
    • 63849230673 scopus 로고    scopus 로고
    • Promoter methylation of HIN-1 in the progression to esophageal squamous cancer
    • Guo M, Ren J, Brock MV, Herman JG, Carraway HE. Promoter methylation of HIN-1 in the progression to esophageal squamous cancer. Epigenetics 3(6), 336-341 (2008).
    • (2008) Epigenetics , vol.3 , Issue.6 , pp. 336-341
    • Guo, M.1    Ren, J.2    Brock, M.V.3    Herman, J.G.4    Carraway, H.E.5
  • 2
    • 0025312728 scopus 로고
    • A genetic model for colorectal tumorigenesis
    • Fearon ER, Vogelstein B. A genetic model for colorectal tumorigenesis. Cell 61(5), 759-767 (1990).
    • (1990) Cell , vol.61 , Issue.5 , pp. 759-767
    • Fearon, E.R.1    Vogelstein, B.2
  • 3
    • 33748042301 scopus 로고    scopus 로고
    • Accumulation of promoter methylation suggests epigenetic progression in squamous cell carcinoma of the esophagus
    • Guo M, Ren J, House MG, Qi Y, Brock MV, Herman JG. Accumulation of promoter methylation suggests epigenetic progression in squamous cell carcinoma of the esophagus. Clin. Cancer Res. 12(15), 4515-4522 (2006).
    • (2006) Clin. Cancer Res. , vol.12 , Issue.15 , pp. 4515-4522
    • Guo, M.1    Ren, J.2    House, M.G.3    Qi, Y.4    Brock, M.V.5    Herman, J.G.6
  • 4
    • 84877574817 scopus 로고    scopus 로고
    • The future of epigenetic therapy in solid tumours-lessons from the past
    • Azad N, Zahnow CA, Rudin CM, Baylin SB. The future of epigenetic therapy in solid tumours-lessons from the past. Nat. Rev. Clin. Oncol. 10(5), 256-266 (2013).
    • (2013) Nat. Rev. Clin. Oncol. , vol.10 , Issue.5 , pp. 256-266
    • Azad, N.1    Zahnow, C.A.2    Rudin, C.M.3    Baylin, S.B.4
  • 5
    • 77954827460 scopus 로고    scopus 로고
    • The path to personalized medicine
    • Hamburg MA, Collins FS. The path to personalized medicine. N. Engl. J. Med. 363(4), 301-304 (2010).
    • (2010) N. Engl. J. Med. , vol.363 , Issue.4 , pp. 301-304
    • Ma, H.1    Collins, F.S.2
  • 6
    • 84923762812 scopus 로고    scopus 로고
    • A new initiative on precision medicine
    • Collins FS, Varmus H. A new initiative on precision medicine. N. Engl. J. Med. 372(9), 793-795 (2015).
    • (2015) N. Engl. J. Med. , vol.372 , Issue.9 , pp. 793-795
    • Collins, F.S.1    Varmus, H.2
  • 8
    • 84877280221 scopus 로고    scopus 로고
    • Momentum grows to make 'personalized' medicine more 'precise
    • Katsnelson A. Momentum grows to make 'personalized' medicine more 'precise'. Nat. Med. 19(3), 249 (2013).
    • (2013) Nat. Med. , vol.19 , Issue.3 , pp. 249
    • Katsnelson, A.1
  • 9
    • 84892485096 scopus 로고    scopus 로고
    • Precision medicine: An approach to R&D for delivering superior medicines to patients
    • Dolsten M, Sogaard M. Precision medicine: an approach to R&D for delivering superior medicines to patients. Clin. Transl. Med. 1(1), 7 (2012).
    • (2012) Clin. Transl. Med. , vol.1 , Issue.1 , pp. 7
    • Dolsten, M.1    Sogaard, M.2
  • 10
    • 33847323129 scopus 로고    scopus 로고
    • Epidermal growth factor receptor mutations in lung cancer
    • Sharma SV, Bell DW, Settleman J, Haber DA. Epidermal growth factor receptor mutations in lung cancer. Nat. Rev. Cancer 7(3), 169-181 (2007).
    • (2007) Nat. Rev. Cancer , vol.7 , Issue.3 , pp. 169-181
    • Sharma, S.V.1    Bell, D.W.2    Settleman, J.3    Haber, D.A.4
  • 13
    • 0037133565 scopus 로고    scopus 로고
    • Comprehensive analysis of CpG islands in human chromosomes 21 and 22
    • Takai D, Jones PA. Comprehensive analysis of CpG islands in human chromosomes 21 and 22. Proc. Natl Acad. Sci. USA 99(6), 3740-3745 (2002).
    • (2002) Proc. Natl Acad. Sci. USA , vol.99 , Issue.6 , pp. 3740-3745
    • Takai, D.1    Jones, P.A.2
  • 15
    • 84905190170 scopus 로고    scopus 로고
    • New answers to old questions from genome-wide maps of DNA methylation in hematopoietic cells
    • Jeong M, Goodell MA. New answers to old questions from genome-wide maps of DNA methylation in hematopoietic cells. Exp. Hematol. 42(8), 609-617 (2014).
    • (2014) Exp. Hematol. , vol.42 , Issue.8 , pp. 609-617
    • Jeong, M.1    Ma, G.2
  • 16
    • 84928530388 scopus 로고    scopus 로고
    • DNA methylome profiling beyond promoters-taking an epigenetic snapshot of the breast tumor microenvironment
    • Jeschke J, Collignon E, Fuks F. DNA methylome profiling beyond promoters-taking an epigenetic snapshot of the breast tumor microenvironment. FEBS J. 282(9), 1801-1814 (2015).
    • (2015) FEBS J , vol.282 , Issue.9 , pp. 1801-1814
    • Jeschke, J.1    Collignon, E.2    Fuks, F.3
  • 17
    • 59149084538 scopus 로고    scopus 로고
    • The human colon cancer methylome shows similar hypo-and hypermethylation at conserved tissue-specific CpG island shores
    • Irizarry RA, Ladd-Acosta C, Wen B et al. The human colon cancer methylome shows similar hypo-and hypermethylation at conserved tissue-specific CpG island shores. Nat. Genet. 41(2), 178-186 (2009).
    • (2009) Nat. Genet. , vol.41 , Issue.2 , pp. 178-186
    • Irizarry, R.A.1    Ladd-Acosta, C.2    Wen, B.3
  • 18
    • 70649095120 scopus 로고    scopus 로고
    • Differential methylation of tissue-and cancer-specific CpG island shores distinguishes human induced pluripotent stem cells, embryonic stem cells and fibroblasts
    • Doi A, Park IH, Wen B et al. Differential methylation of tissue-and cancer-specific CpG island shores distinguishes human induced pluripotent stem cells, embryonic stem cells and fibroblasts. Nat. Genet. 41(12), 1350-1353 (2009).
    • (2009) Nat. Genet. , vol.41 , Issue.12 , pp. 1350-1353
    • Doi, A.1    Park, I.H.2    Wen, B.3
  • 19
    • 1942469956 scopus 로고    scopus 로고
    • MGMT: Its role in cancer aetiology and cancer therapeutics
    • Gerson SL. MGMT: its role in cancer aetiology and cancer therapeutics. Nat. Rev. Cancer 4(4), 296-307 (2004).
    • (2004) Nat. Rev. Cancer , vol.4 , Issue.4 , pp. 296-307
    • Gerson, S.L.1
  • 20
    • 0034626988 scopus 로고    scopus 로고
    • Inactivation of the DNA-repair gene MGMT and the clinical response of gliomas to alkylating agents
    • Esteller M, Garcia-Foncillas J, Andion E et al. Inactivation of the DNA-repair gene MGMT and the clinical response of gliomas to alkylating agents. N. Engl. J. Med. 343(19), 1350-1354 (2000).
    • (2000) N. Engl. J. Med. , vol.343 , Issue.19 , pp. 1350-1354
    • Esteller, M.1    Garcia-Foncillas, J.2    Andion, E.3
  • 21
    • 0034721154 scopus 로고    scopus 로고
    • Chfr defines a mitotic stress checkpoint that delays entry into metaphase
    • Scolnick DM, Halazonetis TD. Chfr defines a mitotic stress checkpoint that delays entry into metaphase. Nature 406(6794), 430-435 (2000).
    • (2000) Nature , vol.406 , Issue.6794 , pp. 430-435
    • Scolnick, D.M.1    Halazonetis, T.D.2
  • 22
    • 84949505981 scopus 로고    scopus 로고
    • Methylation of CHFR sensitizes esophageal squamous cell cancer to docetaxel and paclitaxel
    • Yun T, Liu Y, Gao D et al. Methylation of CHFR sensitizes esophageal squamous cell cancer to docetaxel and paclitaxel. Genes Cancer 6(1-2), 38-48 (2015).
    • (2015) Genes Cancer , vol.6 , Issue.1-2 , pp. 38-48
    • Yun, T.1    Liu, Y.2    Gao, D.3
  • 23
    • 84899984093 scopus 로고    scopus 로고
    • Emerging evidence for CHFR as a cancer biomarker: From tumor biology to precision medicine
    • Derks S, Cleven AH, Melotte V et al. Emerging evidence for CHFR as a cancer biomarker: from tumor biology to precision medicine. Cancer Metastasis Rev. 33(1), 161-171 (2014).
    • (2014) Cancer Metastasis Rev , vol.33 , Issue.1 , pp. 161-171
    • Derks, S.1    Cleven, A.H.2    Melotte, V.3
  • 24
    • 84929173745 scopus 로고    scopus 로고
    • Predictive value of CHFR and MLH1 methylation in human gastric cancer
    • Li Y, Yang Y, Lu Y et al. Predictive value of CHFR and MLH1 methylation in human gastric cancer. Gastric Cancer 18(2), 280-287 (2015).
    • (2015) Gastric Cancer , vol.18 , Issue.2 , pp. 280-287
    • Li, Y.1    Yang, Y.2    Lu, Y.3
  • 25
    • 0038075462 scopus 로고    scopus 로고
    • Disruption of the Fanconi anemia-BRCA pathway in cisplatin-sensitive ovarian tumors
    • Taniguchi T, Tischkowitz M, Ameziane N et al. Disruption of the Fanconi anemia-BRCA pathway in cisplatin-sensitive ovarian tumors. Nat. Med. 9(5), 568-574 (2003).
    • (2003) Nat. Med. , vol.9 , Issue.5 , pp. 568-574
    • Taniguchi, T.1    Tischkowitz, M.2    Ameziane, N.3
  • 26
    • 70350048828 scopus 로고    scopus 로고
    • Methylation and protein expression of DNA repair genes: Association with chemotherapy exposure and survival in sporadic ovarian and peritoneal carcinomas
    • Swisher EM, Gonzalez RM, Taniguchi T et al. Methylation and protein expression of DNA repair genes: association with chemotherapy exposure and survival in sporadic ovarian and peritoneal carcinomas. Mol. Cancer 8, 48 (2009).
    • (2009) Mol. Cancer , vol.8 , pp. 48
    • Swisher, E.M.1    Gonzalez, R.M.2    Taniguchi, T.3
  • 27
    • 85027954125 scopus 로고    scopus 로고
    • DNA methylation profiling in the clinic: Applications and challenges
    • Heyn H, Esteller M. DNA methylation profiling in the clinic: applications and challenges. Nat. Rev. Genet. 13(10), 679-692 (2012).
    • (2012) Nat. Rev. Genet. , vol.13 , Issue.10 , pp. 679-692
    • Heyn, H.1    Esteller, M.2
  • 28
    • 79960311900 scopus 로고    scopus 로고
    • Association of CHFR promoter methylation with disease recurrence in locally advanced colon cancer
    • Tanaka M, Chang P, Li Y et al. Association of CHFR promoter methylation with disease recurrence in locally advanced colon cancer. Clin. Cancer Res. 17(13), 4531-4540 (2011).
    • (2011) Clin. Cancer Res. , vol.17 , Issue.13 , pp. 4531-4540
    • Tanaka, M.1    Chang, P.2    Li, Y.3
  • 29
    • 78751629661 scopus 로고    scopus 로고
    • BRCA1 CpG island hypermethylation predicts sensitivity to poly(adenosine diphosphate)-ribose polymerase inhibitors
    • Veeck J, Ropero S, Setien F et al. BRCA1 CpG island hypermethylation predicts sensitivity to poly(adenosine diphosphate)-ribose polymerase inhibitors. J. Clin. Oncol. 28(29), e563-e564 (2010).
    • (2010) J. Clin. Oncol. , vol.28 , Issue.29 , pp. e563-e564
    • Veeck, J.1    Ropero, S.2    Setien, F.3
  • 30
    • 0033535612 scopus 로고    scopus 로고
    • A role for methylation of the hMLH1 promoter in loss of hMLH1 expression and drug resistance in ovarian cancer
    • Strathdee G, Mackean MJ, Illand M, Brown R. A role for methylation of the hMLH1 promoter in loss of hMLH1 expression and drug resistance in ovarian cancer. Oncogene 18(14), 2335-2341 (1999).
    • (1999) Oncogene , vol.18 , Issue.14 , pp. 2335-2341
    • Strathdee, G.1    MacKean, M.J.2    Illand, M.3    Brown, R.4
  • 31
    • 77952248685 scopus 로고    scopus 로고
    • DNA methylation profiling in doxorubicin treated primary locally advanced breast tumours identifies novel genes associated with survival and treatment response
    • Dejeux E, Ronneberg JA, Solvang H et al. DNA methylation profiling in doxorubicin treated primary locally advanced breast tumours identifies novel genes associated with survival and treatment response. Mol. Cancer 9, 68 (2010).
    • (2010) Mol. Cancer , vol.9 , pp. 68
    • Dejeux, E.1    Ronneberg, J.A.2    Solvang, H.3
  • 32
    • 84055222098 scopus 로고    scopus 로고
    • Epigenetic alteration of PRKCDBP in colorectal cancers and its implication in tumor cell resistance to TNF-induced apoptosis
    • Lee JH, Kang MJ, Han HY et al. Epigenetic alteration of PRKCDBP in colorectal cancers and its implication in tumor cell resistance to TNF-induced apoptosis. Clin. Cancer Res. 17(24), 7551-7562 (2011).
    • (2011) Clin. Cancer Res. , vol.17 , Issue.24 , pp. 7551-7562
    • Lee, J.H.1    Kang, M.J.2    Han, H.Y.3
  • 33
    • 33644824212 scopus 로고    scopus 로고
    • 14-3-3sigma methylation in pretreatment serum circulating DNA of cisplatin-plus-gemcitabine-treated advanced non-small-cell lung cancer patients predicts survival: The Spanish Lung Cancer Group
    • Ramirez JL, Rosell R, Taron M et al. 14-3-3sigma methylation in pretreatment serum circulating DNA of cisplatin-plus-gemcitabine-treated advanced non-small-cell lung cancer patients predicts survival: The Spanish Lung Cancer Group. J. Clin. Oncol. 23(36), 9105-9112 (2005).
    • (2005) J. Clin. Oncol. , vol.23 , Issue.36 , pp. 9105-9112
    • Ramirez, J.L.1    Rosell, R.2    Taron, M.3
  • 34
    • 84855755065 scopus 로고    scopus 로고
    • TFAP2E-DKK4 and chemoresistance in colorectal cancer
    • Ebert MP, Tanzer M, Balluff B et al. TFAP2E-DKK4 and chemoresistance in colorectal cancer. N. Engl. J. Med. 366(1), 44-53 (2012).
    • (2012) N. Engl. J. Med. , vol.366 , Issue.1 , pp. 44-53
    • Ebert, M.P.1    Tanzer, M.2    Balluff, B.3
  • 35
    • 28844468518 scopus 로고    scopus 로고
    • Role of DNA hypomethylation in the development of the resistance to doxorubicin in human MCF-7 breast adenocarcinoma cells
    • Chekhun VF, Kulik GI, Yurchenko OV et al. Role of DNA hypomethylation in the development of the resistance to doxorubicin in human MCF-7 breast adenocarcinoma cells. Cancer Lett. 231(1), 87-93 (2006).
    • (2006) Cancer Lett , vol.231 , Issue.1 , pp. 87-93
    • Chekhun, V.F.1    Kulik, G.I.2    Yurchenko, O.V.3
  • 36
    • 0035843169 scopus 로고    scopus 로고
    • Inactivation of the apoptosis effector Apaf-1 in malignant melanoma
    • Soengas MS, Capodieci P, Polsky D et al. Inactivation of the apoptosis effector Apaf-1 in malignant melanoma. Nature 409(6817), 207-211 (2001).
    • (2001) Nature , vol.409 , Issue.6817 , pp. 207-211
    • Soengas, M.S.1    Capodieci, P.2    Polsky, D.3
  • 37
    • 38549141939 scopus 로고    scopus 로고
    • Identification of CDK10 as an important determinant of resistance to endocrine therapy for breast cancer
    • Iorns E, Turner NC, Elliott R et al. Identification of CDK10 as an important determinant of resistance to endocrine therapy for breast cancer. Cancer Cell 13(2), 91-104 (2008).
    • (2008) Cancer Cell , vol.13 , Issue.2 , pp. 91-104
    • Iorns, E.1    Turner, N.C.2    Elliott, R.3
  • 38
    • 77949654035 scopus 로고    scopus 로고
    • IGFBP-3 hypermethylation-derived deficiency mediates cisplatin resistance in non-small-cell lung cancer
    • Ibanez De Caceres I, Cortes-Sempere M, Moratilla C et al. IGFBP-3 hypermethylation-derived deficiency mediates cisplatin resistance in non-small-cell lung cancer. Oncogene 29(11), 1681-1690 (2010).
    • (2010) Oncogene , vol.29 , Issue.11 , pp. 1681-1690
    • Ibanez De Caceres, I.1    Cortes-Sempere, M.2    Moratilla, C.3
  • 39
    • 77957560042 scopus 로고    scopus 로고
    • Metallothionein 1E is methylated in malignant melanoma and increases sensitivity to cisplatin-induced apoptosis
    • Faller WJ, Rafferty M, Hegarty S et al. Metallothionein 1E is methylated in malignant melanoma and increases sensitivity to cisplatin-induced apoptosis. Melanoma Res. 20(5), 392-400 (2010).
    • (2010) Melanoma Res , vol.20 , Issue.5 , pp. 392-400
    • Faller, W.J.1    Rafferty, M.2    Hegarty, S.3
  • 40
    • 40949114150 scopus 로고    scopus 로고
    • The transglutaminase 2 gene (TGM2), a potential molecular marker for chemotherapeutic drug sensitivity, is epigenetically silenced in breast cancer
    • Ai L, Kim WJ, Demircan B et al. The transglutaminase 2 gene (TGM2), a potential molecular marker for chemotherapeutic drug sensitivity, is epigenetically silenced in breast cancer. Carcinogenesis 29(3), 510-518 (2008).
    • (2008) Carcinogenesis , vol.29 , Issue.3 , pp. 510-518
    • Ai, L.1    Kim, W.J.2    Demircan, B.3
  • 41
    • 37049001640 scopus 로고    scopus 로고
    • Drug sensitivity prediction by CpG island methylation profile in the NCI-60 cancer cell line panel
    • Shen L, Kondo Y, Ahmed S et al. Drug sensitivity prediction by CpG island methylation profile in the NCI-60 cancer cell line panel. Cancer Res. 67(23), 11335-11343 (2007).
    • (2007) Cancer Res , vol.67 , Issue.23 , pp. 11335-11343
    • Shen, L.1    Kondo, Y.2    Ahmed, S.3
  • 42
    • 77951566673 scopus 로고    scopus 로고
    • DNA methyltransferase 3-like affects promoter methylation of thymine DNA glycosylase independently of DNMT1 and DNMT3B in cancer cells
    • Kim H, Park J, Jung Y et al. DNA methyltransferase 3-like affects promoter methylation of thymine DNA glycosylase independently of DNMT1 and DNMT3B in cancer cells. Int. J. Oncol. 36(6), 1563-1572 (2010).
    • (2010) Int. J. Oncol. , vol.36 , Issue.6 , pp. 1563-1572
    • Kim, H.1    Park, J.2    Jung, Y.3
  • 43
    • 84892949427 scopus 로고    scopus 로고
    • Harnessing the potential of epigenetic therapy to target solid tumors
    • Ahuja N, Easwaran H, Baylin SB. Harnessing the potential of epigenetic therapy to target solid tumors. J. Clin. Invest. 124(1), 56-63 (2014).
    • (2014) J. Clin. Invest. , vol.124 , Issue.1 , pp. 56-63
    • Ahuja, N.1    Easwaran, H.2    Baylin, S.B.3
  • 44
    • 84869853588 scopus 로고    scopus 로고
    • Phase 2 study of the lenalidomide and azacitidine combination in patients with higher-risk myelodysplastic syndromes
    • Sekeres MA, Tiu RV, Komrokji R et al. Phase 2 study of the lenalidomide and azacitidine combination in patients with higher-risk myelodysplastic syndromes. Blood 120(25), 4945-4951 (2012).
    • (2012) Blood , vol.120 , Issue.25 , pp. 4945-4951
    • Ma, S.1    Tiu, R.V.2    Komrokji, R.3
  • 45
    • 0035988868 scopus 로고    scopus 로고
    • Review of the clinical experience with 5-azacytidine and 5-aza-2-deoxycytidine in solid tumors
    • Aparicio A, Weber JS. Review of the clinical experience with 5-azacytidine and 5-aza-2-deoxycytidine in solid tumors. Curr. Opin. Invest. Drugs 3(4), 627-633 (2002).
    • (2002) Curr. Opin. Invest. Drugs , vol.3 , Issue.4 , pp. 627-633
    • Aparicio, A.1    Weber, J.S.2
  • 46
    • 84863337757 scopus 로고    scopus 로고
    • Transient low doses of DNA-demethylating agents exert durable antitumor effects on hematological and epithelial tumor cells
    • Tsai HC, Li H, Van Neste L et al. Transient low doses of DNA-demethylating agents exert durable antitumor effects on hematological and epithelial tumor cells. Cancer Cell 21(3), 430-446 (2012).
    • (2012) Cancer Cell , vol.21 , Issue.3 , pp. 430-446
    • Tsai, H.C.1    Li, H.2    Van Neste, L.3
  • 47
    • 26644460450 scopus 로고    scopus 로고
    • Zebularine: A unique molecule for an epigenetically based strategy in cancer chemotherapy. The magic of its chemistry and biology
    • Marquez VE, Barchi JJ Jr, Kelley JA et al. Zebularine: a unique molecule for an epigenetically based strategy in cancer chemotherapy. The magic of its chemistry and biology. Nucleosides, Nucleotides Nucleic Acids 24(5-7), 305-318 (2005).
    • (2005) Nucleosides.Nucleotides Nucleic Acids , vol.24 , Issue.5-7 , pp. 305-318
    • Marquez, V.E.1    Barchi, J. J.2    Kelley, J.A.3
  • 48
    • 1642499364 scopus 로고    scopus 로고
    • Continuous zebularine treatment effectively sustains demethylation in human bladder cancer cells
    • Cheng JC, Weisenberger DJ, Gonzales FA et al. Continuous zebularine treatment effectively sustains demethylation in human bladder cancer cells. Mol. Cell. Biol. 24(3), 1270-1278 (2004).
    • (2004) Mol. Cell. Biol. , vol.24 , Issue.3 , pp. 1270-1278
    • Cheng, J.C.1    Weisenberger, D.J.2    Gonzales, F.A.3
  • 49
    • 84945485095 scopus 로고    scopus 로고
    • Guadecitabine (SGI-110) priming sensitizes hepatocellular carcinoma cells to oxaliplatin
    • Epub ahead of print
    • Kuang Y, El-Khoueiry A, Taverna P, Ljungman M, Neamati N. Guadecitabine (SGI-110) priming sensitizes hepatocellular carcinoma cells to oxaliplatin. Mol. Oncol. doi:10.1016/j. molonc.2015.06.002 (2015) (Epub ahead of print).
    • (2015) Mol. Oncol.
    • Kuang, Y.1    El-Khoueiry, A.2    Taverna, P.3    Ljungman, M.4    Neamati, N.5
  • 50
    • 84899849424 scopus 로고    scopus 로고
    • Pharmacokinetics of different formulations of oral azacitidine (CC-486) and the effect of food and modified gastric pH on pharmacokinetics in subjects with hematologic malignancies
    • Laille E, Savona MR, Scott BL, Boyd TE, Dong Q, Skikne B. Pharmacokinetics of different formulations of oral azacitidine (CC-486) and the effect of food and modified gastric pH on pharmacokinetics in subjects with hematologic malignancies. J. Clin. Pharmacol. 54(6), 630-639 (2014).
    • (2014) J. Clin. Pharmacol. , vol.54 , Issue.6 , pp. 630-639
    • Laille, E.1    Savona, M.R.2    Scott, B.L.3    Boyd, T.E.4    Dong, Q.5    Skikne, B.6
  • 51
    • 79954416175 scopus 로고    scopus 로고
    • DNA hypermethylation as a chemotherapy target
    • Ren J, Singh BN, Huang Q et al. DNA hypermethylation as a chemotherapy target. Cell. Signal. 23(7), 1082-1093 (2011).
    • (2011) Cell. Signal. , vol.23 , Issue.7 , pp. 1082-1093
    • Ren, J.1    Singh, B.N.2    Huang, Q.3
  • 52
    • 79952532412 scopus 로고    scopus 로고
    • Cancer epigenetics: Linking basic biology to clinical medicine
    • Tsai HC, Baylin SB. Cancer epigenetics: linking basic biology to clinical medicine. Cell Res. 21(3), 502-517 (2011).
    • (2011) Cell Res , vol.21 , Issue.3 , pp. 502-517
    • Tsai, H.C.1    Baylin, S.B.2
  • 53
    • 0035943709 scopus 로고    scopus 로고
    • DNMT3A and DNMT3B are transcriptional repressors that exhibit unique localization properties to heterochromatin
    • Bachman KE, Rountree MR, Baylin SB. DNMT3A and DNMT3B are transcriptional repressors that exhibit unique localization properties to heterochromatin. J. Biol. Chem. 276(34), 32282-32287 (2001).
    • (2001) J. Biol. Chem. , vol.276 , Issue.34 , pp. 32282-32287
    • Bachman, K.E.1    Rountree, M.R.2    Baylin, S.B.3
  • 54
    • 0033919595 scopus 로고    scopus 로고
    • DNMT1 forms a complex with Rb E2F1 and HDAC1 and represses transcription from E2F-responsive promoters
    • Robertson KD, Ait-Si-Ali S, Yokochi T, Wade PA, Jones PL, Wolffe AP. DNMT1 forms a complex with Rb, E2F1 and HDAC1 and represses transcription from E2F-responsive promoters. Nat. Genet. 25(3), 338-342 (2000).
    • (2000) Nat. Genet. , vol.25 , Issue.3 , pp. 338-342
    • Robertson, K.D.1    Ait-Si-Ali, S.2    Yokochi, T.3    Wade, P.A.4    Jones, P.L.5    Wolffe, A.P.6
  • 55
    • 78650304236 scopus 로고    scopus 로고
    • Charting histone modifications and the functional organization of mammalian genomes
    • Zhou VW, Goren A, Bernstein BE. Charting histone modifications and the functional organization of mammalian genomes. Nat. Rev. Genet. 12(1), 7-18 (2011).
    • (2011) Nat. Rev. Genet. , vol.12 , Issue.1 , pp. 7-18
    • Zhou, V.W.1    Goren, A.2    Bernstein, B.E.3
  • 56
    • 60149091656 scopus 로고    scopus 로고
    • ChIP-seq accurately predicts tissue-specific activity of enhancers
    • Visel A, Blow MJ, Li Z et al. ChIP-seq accurately predicts tissue-specific activity of enhancers. Nature 457(7231), 854-858 (2009).
    • (2009) Nature , vol.457 , Issue.7231 , pp. 854-858
    • Visel, A.1    Blow, M.J.2    Li, Z.3
  • 59
    • 84863770814 scopus 로고    scopus 로고
    • Cancer genetics and epigenetics: Two sides of the same coin Cancer
    • You JS, Jones PA. Cancer genetics and epigenetics: two sides of the same coin Cancer Cell 22(1), 9-20 (2012).
    • (2012) Cell , vol.22 , Issue.1 , pp. 9-20
    • You, J.S.1    Jones, P.A.2
  • 60
    • 84921788105 scopus 로고    scopus 로고
    • Epigenetics of gastric cancer
    • Guo M, Yan W. Epigenetics of gastric cancer. Methods Mol. Biol. 1238, 783-799 (2015).
    • (2015) Methods Mol. Biol. , vol.1238 , pp. 783-799
    • Guo, M.1    Yan, W.2
  • 62
    • 33748451151 scopus 로고    scopus 로고
    • Anticancer activities of histone deacetylase inhibitors
    • Bolden JE, Peart MJ, Johnstone RW. Anticancer activities of histone deacetylase inhibitors. Nat. Rev. Drug Discov. 5(9), 769-784 (2006).
    • (2006) Nat. Rev. Drug Discov. , vol.5 , Issue.9 , pp. 769-784
    • Bolden, J.E.1    Peart, M.J.2    Johnstone, R.W.3
  • 63
    • 84921810598 scopus 로고    scopus 로고
    • Epigenetics of colorectal cancer
    • Yan W, Guo M. Epigenetics of colorectal cancer. Methods Mol. Biol. 1238, 405-424 (2015).
    • (2015) Methods Mol. Biol. , vol.1238 , pp. 405-424
    • Yan, W.1    Guo, M.2
  • 64
    • 84939522027 scopus 로고    scopus 로고
    • FDA approval: Belinostat for the treatment of patients with relapsed or refractory peripheral T-cell lymphoma
    • Lee HZ, Kwitkowski VE, Del Valle PL et al. FDA approval: belinostat for the treatment of patients with relapsed or refractory peripheral T-cell lymphoma. Clin. Cancer Res. 21(12), 2666-2670 (2015).
    • (2015) Clin. Cancer Res. , vol.21 , Issue.12 , pp. 2666-2670
    • Lee, H.Z.1    Kwitkowski, V.E.2    Del Valle, P.L.3
  • 66
    • 77957937450 scopus 로고    scopus 로고
    • Epigenetic modifications as therapeutic targets
    • Kelly TK, De Carvalho DD, Jones PA. Epigenetic modifications as therapeutic targets. Nat. Biotechnol. 28(10), 1069-1078 (2010).
    • (2010) Nat. Biotechnol. , vol.28 , Issue.10 , pp. 1069-1078
    • Kelly, T.K.1    De Carvalho, D.D.2    Jones, P.A.3
  • 67
    • 43749109171 scopus 로고    scopus 로고
    • A novel histone deacetylase 8 (HDAC8)-specific inhibitor PCI-34051 induces apoptosis in T-cell lymphomas
    • Balasubramanian S, Ramos J, Luo W, Sirisawad M, Verner E, Buggy JJ. A novel histone deacetylase 8 (HDAC8)-specific inhibitor PCI-34051 induces apoptosis in T-cell lymphomas. Leukemia 22(5), 1026-1034 (2008).
    • (2008) Leukemia , vol.22 , Issue.5 , pp. 1026-1034
    • Balasubramanian, S.1    Ramos, J.2    Luo, W.3    Sirisawad, M.4    Verner, E.5    Buggy, J.J.6
  • 68
    • 79960058024 scopus 로고    scopus 로고
    • Selective killing of mixed lineage leukemia cells by a potent small-molecule DOT1L inhibitor
    • Daigle SR, Olhava EJ, Therkelsen CA et al. Selective killing of mixed lineage leukemia cells by a potent small-molecule DOT1L inhibitor. Cancer Cell 20(1), 53-65 (2011).
    • (2011) Cancer Cell , vol.20 , Issue.1 , pp. 53-65
    • Daigle, S.R.1    Olhava, E.J.2    Therkelsen, C.A.3
  • 69
    • 84894493778 scopus 로고    scopus 로고
    • Targeting epigenetic regulators for cancer therapy
    • Wee S, Dhanak D, Li H et al. Targeting epigenetic regulators for cancer therapy. Ann. NY Acad. Sci. 1309, 30-36 (2014).
    • (2014) Ann. NY Acad. Sci. , vol.1309 , pp. 30-36
    • Wee, S.1    Dhanak, D.2    Li, H.3
  • 70
    • 34247625135 scopus 로고    scopus 로고
    • Pharmacologic disruption of Polycomb-repressive complex 2-mediated gene repression selectively induces apoptosis in cancer cells
    • Tan J, Yang X, Zhuang L et al. Pharmacologic disruption of Polycomb-repressive complex 2-mediated gene repression selectively induces apoptosis in cancer cells. Genes Dev. 21(9), 1050-1063 (2007).
    • (2007) Genes Dev , vol.21 , Issue.9 , pp. 1050-1063
    • Tan, J.1    Yang, X.2    Zhuang, L.3
  • 73
    • 84900463546 scopus 로고    scopus 로고
    • Targeting the BET family for the treatment of leukemia
    • Lucas X, Gunther S. Targeting the BET family for the treatment of leukemia. Epigenomics 6(2), 153-155 (2014).
    • (2014) Epigenomics , vol.6 , Issue.2 , pp. 153-155
    • Lucas, X.1    Gunther, S.2
  • 74
    • 80054984945 scopus 로고    scopus 로고
    • Inhibition of BET recruitment to chromatin as an effective treatment for MLL-fusion leukaemia
    • Dawson MA, Prinjha RK, Dittmann A et al. Inhibition of BET recruitment to chromatin as an effective treatment for MLL-fusion leukaemia. Nature 478(7370), 529-533 (2011).
    • (2011) Nature , vol.478 , Issue.7370 , pp. 529-533
    • Ma, D.1    Prinjha, R.K.2    Dittmann, A.3
  • 75
    • 80052955256 scopus 로고    scopus 로고
    • BET bromodomain inhibition as a therapeutic strategy to target c-Myc
    • Delmore JE, Issa GC, Lemieux ME et al. BET bromodomain inhibition as a therapeutic strategy to target c-Myc. Cell 146(6), 904-917 (2011).
    • (2011) Cell , vol.146 , Issue.6 , pp. 904-917
    • Delmore, J.E.1    Issa, G.C.2    Lemieux, M.E.3
  • 76
    • 27644589675 scopus 로고    scopus 로고
    • The diverse functions of histone lysine methylation
    • Martin C, Zhang Y. The diverse functions of histone lysine methylation. Nat. Rev. Mol. Cell Biol. 6(11), 838-849 (2005).
    • (2005) Nat. Rev. Mol. Cell Biol. , vol.6 , Issue.11 , pp. 838-849
    • Martin, C.1    Zhang, Y.2
  • 77
    • 32844454603 scopus 로고    scopus 로고
    • Histone demethylation by a family of JmjC domain-containing proteins
    • Tsukada Y, Fang J, Erdjument-Bromage H et al. Histone demethylation by a family of JmjC domain-containing proteins. Nature 439(7078), 811-816 (2006).
    • (2006) Nature , vol.439 , Issue.7078 , pp. 811-816
    • Tsukada, Y.1    Fang, J.2    Erdjument-Bromage, H.3
  • 78
    • 34249026300 scopus 로고    scopus 로고
    • High-resolution profiling of histone methylations in the human genome
    • Barski A, Cuddapah S, Cui K et al. High-resolution profiling of histone methylations in the human genome. Cell 129(4), 823-837 (2007).
    • (2007) Cell , vol.129 , Issue.4 , pp. 823-837
    • Barski, A.1    Cuddapah, S.2    Cui, K.3
  • 79
  • 80
    • 35848946372 scopus 로고    scopus 로고
    • Primers on chromatin
    • Lall S. Primers on chromatin. Nat. Struct. Mol. Biol. 14(11), 1110-1115 (2007).
    • (2007) Nat. Struct. Mol. Biol. , vol.14 , Issue.11 , pp. 1110-1115
    • Lall, S.1
  • 81
    • 84870375316 scopus 로고    scopus 로고
    • Histone lysine methylation dynamics: Establishment, regulation, and biological impact
    • Black JC, Van Rechem C, Whetstine JR. Histone lysine methylation dynamics: establishment, regulation, and biological impact. Mol. Cell 48(4), 491-507 (2012).
    • (2012) Mol. Cell , vol.48 , Issue.4 , pp. 491-507
    • Black, J.C.1    Van Rechem, C.2    Whetstine, J.R.3
  • 82
    • 79959267832 scopus 로고    scopus 로고
    • Validation of the histone methyltransferase EZH2 as a therapeutic target for various types of human cancer and as a prognostic marker
    • Takawa M, Masuda K, Kunizaki M et al. Validation of the histone methyltransferase EZH2 as a therapeutic target for various types of human cancer and as a prognostic marker. Cancer Sci. 102(7), 1298-1305 (2011).
    • (2011) Cancer Sci , vol.102 , Issue.7 , pp. 1298-1305
    • Takawa, M.1    Masuda, K.2    Kunizaki, M.3
  • 83
    • 84923109013 scopus 로고    scopus 로고
    • Targeting histone methyltransferase EZH2 as cancer treatment
    • Kondo Y. Targeting histone methyltransferase EZH2 as cancer treatment. J. Biochem. 156(5), 249-257 (2014).
    • (2014) J. Biochem. , vol.156 , Issue.5 , pp. 249-257
    • Kondo, Y.1
  • 84
    • 84893521949 scopus 로고    scopus 로고
    • EZH2: Biology, disease, and structure-based drug discovery
    • Tan JZ, Yan Y, Wang XX, Jiang Y, Xu HE. EZH2: biology, disease, and structure-based drug discovery. Acta Pharmacol. Sin. 35(2), 161-174 (2014).
    • (2014) Acta Pharmacol. Sin. , vol.35 , Issue.2 , pp. 161-174
    • Tan, J.Z.1    Yan, Y.2    Wang, X.X.3    Jiang, Y.4    Xu, H.E.5
  • 85
    • 55949136562 scopus 로고    scopus 로고
    • Roles of the EZH2 histone methyltransferase in cancer epigenetics
    • Simon JA, Lange CA. Roles of the EZH2 histone methyltransferase in cancer epigenetics. Mutat. Res. 647(1-2), 21-29 (2008).
    • (2008) Mutat. Res. , vol.647 , Issue.1-2 , pp. 21-29
    • Simon, J.A.1    Lange, C.A.2
  • 86
    • 69949148388 scopus 로고    scopus 로고
    • Protein methyltransferases as a target class for drug discovery
    • Copeland RA, Solomon ME, Richon VM. Protein methyltransferases as a target class for drug discovery. Nat. Rev. Drug Discov. 8(9), 724-732 (2009).
    • (2009) Nat. Rev. Drug Discov. , vol.8 , Issue.9 , pp. 724-732
    • Copeland, R.A.1    Solomon, M.E.2    Richon, V.M.3
  • 87
    • 84867632489 scopus 로고    scopus 로고
    • A selective inhibitor of EZH2 blocks H3K27 methylation and kills mutant lymphoma cells
    • Knutson SK, Wigle TJ, Warholic NM et al. A selective inhibitor of EZH2 blocks H3K27 methylation and kills mutant lymphoma cells. Nat. Chem. Biol. 8(11), 890-896 (2012).
    • (2012) Nat. Chem. Biol. , vol.8 , Issue.11 , pp. 890-896
    • Knutson, S.K.1    Wigle, T.J.2    Warholic, N.M.3
  • 88
    • 84870573126 scopus 로고    scopus 로고
    • EZH2 inhibition as a therapeutic strategy for lymphoma with EZH2-activating mutations
    • Mccabe MT, Ott HM, Ganji G et al. EZH2 inhibition as a therapeutic strategy for lymphoma with EZH2-activating mutations. Nature 492(7427), 108-112 (2012).
    • (2012) Nature , vol.492 , Issue.7427 , pp. 108-112
    • McCabe, M.T.1    Ott, H.M.2    Ganji, G.3
  • 89
    • 84921318899 scopus 로고    scopus 로고
    • Selective inhibition of EZH2 by EPZ-6438 leads to potent antitumor activity in EZH2-mutant non-Hodgkin lymphoma
    • Knutson SK, Kawano S, Minoshima Y et al. Selective inhibition of EZH2 by EPZ-6438 leads to potent antitumor activity in EZH2-mutant non-Hodgkin lymphoma. Mol. Cancer Ther. 13(4), 842-854 (2014).
    • (2014) Mol. Cancer Ther. , vol.13 , Issue.4 , pp. 842-854
    • Knutson, S.K.1    Kawano, S.2    Minoshima, Y.3
  • 90
    • 84900414359 scopus 로고    scopus 로고
    • EZH2: Not EZHY (easy) to deal
    • Deb G, Singh AK, Gupta S. EZH2: not EZHY (easy) to deal. Mol. Cancer Res. 12(5), 639-653 (2014).
    • (2014) Mol. Cancer Res. , vol.12 , Issue.5 , pp. 639-653
    • Deb, G.1    Singh, A.K.2    Gupta, S.3
  • 91
    • 79959960773 scopus 로고    scopus 로고
    • The diverse functions of DOT1 and H3K79 methylation
    • Nguyen AT, Zhang Y. The diverse functions of DOT1 and H3K79 methylation. Genes Dev. 25(13), 1345-1358 (2011).
    • (2011) Genes Dev , vol.25 , Issue.13 , pp. 1345-1358
    • Nguyen, A.T.1    Zhang, Y.2
  • 92
    • 4644220954 scopus 로고    scopus 로고
    • MLL: A histone methyltransferase disrupted in leukemia
    • Hess JL. MLL: a histone methyltransferase disrupted in leukemia. Trends Mol. Med. 10(10), 500-507 (2004).
    • (2004) Trends Mol. Med. , vol.10 , Issue.10 , pp. 500-507
    • Hess, J.L.1
  • 93
    • 84886864184 scopus 로고    scopus 로고
    • Potent inhibition of DOT1L as treatment of MLL-fusion leukemia
    • Daigle SR, Olhava EJ, Therkelsen CA et al. Potent inhibition of DOT1L as treatment of MLL-fusion leukemia. Blood 122(6), 1017-1025 (2013).
    • (2013) Blood , vol.122 , Issue.6 , pp. 1017-1025
    • Daigle, S.R.1    Olhava, E.J.2    Therkelsen, C.A.3
  • 94
    • 33846522525 scopus 로고    scopus 로고
    • The mixed-lineage leukemia fusion partner AF4 stimulates RNA polymerase II transcriptional elongation and mediates coordinated chromatin remodeling
    • Bitoun E, Oliver PL, Davies KE. The mixed-lineage leukemia fusion partner AF4 stimulates RNA polymerase II transcriptional elongation and mediates coordinated chromatin remodeling. Hum. Mol. Genet. 16(1), 92-106 (2007).
    • (2007) Hum. Mol. Genet. , vol.16 , Issue.1 , pp. 92-106
    • Bitoun, E.1    Oliver, P.L.2    Davies, K.E.3
  • 95
    • 80053140577 scopus 로고    scopus 로고
    • Function of leukemogenic mixed lineage leukemia 1 (MLL) fusion proteins through distinct partner protein complexes
    • Biswas D, Milne TA, Basrur V et al. Function of leukemogenic mixed lineage leukemia 1 (MLL) fusion proteins through distinct partner protein complexes. Proc. Natl Acad. Sci. USA 108(38), 15751-15756 (2011).
    • (2011) Proc. Natl Acad. Sci. USA , vol.108 , Issue.38 , pp. 15751-15756
    • Biswas, D.1    Milne, T.A.2    Basrur, V.3
  • 96
    • 39649084473 scopus 로고    scopus 로고
    • A role for the MLL fusion partner ENL in transcriptional elongation and chromatin modification
    • Mueller D, Bach C, Zeisig D et al. A role for the MLL fusion partner ENL in transcriptional elongation and chromatin modification. Blood 110(13), 4445-4454 (2007).
    • (2007) Blood , vol.110 , Issue.13 , pp. 4445-4454
    • Mueller, D.1    Bach, C.2    Zeisig, D.3
  • 98
    • 76349118080 scopus 로고    scopus 로고
    • A higher-order complex containing AF4 and ENL family proteins with P-TEFb facilitates oncogenic and physiologic MLL-dependent transcription
    • Yokoyama A, Lin M, Naresh A, Kitabayashi I, Cleary ML. A higher-order complex containing AF4 and ENL family proteins with P-TEFb facilitates oncogenic and physiologic MLL-dependent transcription. Cancer Cell 17(2), 198-212 (2010).
    • (2010) Cancer Cell , vol.17 , Issue.2 , pp. 198-212
    • Yokoyama, A.1    Lin, M.2    Naresh, A.3    Kitabayashi, I.4    Cleary, M.L.5
  • 99
    • 77954143236 scopus 로고    scopus 로고
    • Characterization of the DOT1L network: Implications of diverse roles for DOT1L
    • Park G, Gong Z, Chen J, Kim JE. Characterization of the DOT1L network: implications of diverse roles for DOT1L. Protein J. 29(3), 213-223 (2010).
    • (2010) Protein J , vol.29 , Issue.3 , pp. 213-223
    • Park, G.1    Gong, Z.2    Chen, J.3    Kim, J.E.4
  • 100
    • 33745873838 scopus 로고    scopus 로고
    • DOT1A-AF9 complex mediates histone H3 Lys-79 hypermethylation and repression of ENaCalpha in an aldosterone-sensitive manner
    • Zhang W, Xia X, Reisenauer MR, Hemenway CS, Kone BC. DOT1A-AF9 complex mediates histone H3 Lys-79 hypermethylation and repression of ENaCalpha in an aldosterone-sensitive manner. J. Biol. Chem. 281(26), 18059-18068 (2006).
    • (2006) J. Biol. Chem. , vol.281 , Issue.26 , pp. 18059-18068
    • Zhang, W.1    Xia, X.2    Reisenauer, M.R.3    Hemenway, C.S.4    Kone, B.C.5
  • 101
    • 77949409084 scopus 로고    scopus 로고
    • Linking H3K79 trimethylation to Wnt signaling through a novel DOT1-containing complex (DotCom)
    • Mohan M, Herz HM, Takahashi YH et al. Linking H3K79 trimethylation to Wnt signaling through a novel DOT1-containing complex (DotCom). Genes Dev. 24(6), 574-589 (2010).
    • (2010) Genes Dev , vol.24 , Issue.6 , pp. 574-589
    • Mohan, M.1    Herz, H.M.2    Takahashi, Y.H.3
  • 102
    • 79959493965 scopus 로고    scopus 로고
    • DOT1L, the H3K79 methyltransferase, is required for MLL-AF9-mediated leukemogenesis
    • Nguyen AT, Taranova O, He J, Zhang Y. DOT1L, the H3K79 methyltransferase, is required for MLL-AF9-mediated leukemogenesis. Blood 117(25), 6912-6922 (2011).
    • (2011) Blood , vol.117 , Issue.25 , pp. 6912-6922
    • Nguyen, A.T.1    Taranova, O.2    He, J.3    Zhang, Y.4
  • 103
    • 78650205549 scopus 로고    scopus 로고
    • MLL-AF9 and MLL-ENL alter the dynamic association of transcriptional regulators with genes critical for leukemia
    • e71-75
    • Monroe SC, Jo SY, Sanders DS et al. MLL-AF9 and MLL-ENL alter the dynamic association of transcriptional regulators with genes critical for leukemia. Exp. Hematol. 39(1), 77-86 e71-75 (2011).
    • (2011) Exp. Hematol. , vol.39 , Issue.1 , pp. 77-86
    • Monroe, S.C.1    Jo, S.Y.2    Sanders, D.S.3
  • 104
    • 29244433742 scopus 로고    scopus 로고
    • Leukemogenic MLL fusion proteins bind across a broad region of the Hox a9 locus, promoting transcription and multiple histone modifications
    • Milne TA, Martin ME, Brock HW, Slany RK, Hess JL. Leukemogenic MLL fusion proteins bind across a broad region of the Hox a9 locus, promoting transcription and multiple histone modifications. Cancer Res. 65(24), 11367-11374 (2005).
    • (2005) Cancer Res , vol.65 , Issue.24 , pp. 11367-11374
    • Milne, T.A.1    Martin, M.E.2    Brock, H.W.3    Slany, R.K.4    Hess, J.L.5
  • 105
    • 84863115862 scopus 로고    scopus 로고
    • Deficiency of H3K79 histone methyltransferase DOT-like protein (DOT1L) inhibits cell proliferation
    • Kim W, Kim R, Park G, Park JW, Kim JE. Deficiency of H3K79 histone methyltransferase DOT-like protein (DOT1L) inhibits cell proliferation. J. Biol. Chem. 287(8), 5588-5599 (2012).
    • (2012) J. Biol. Chem. , vol.287 , Issue.8 , pp. 5588-5599
    • Kim, W.1    Kim, R.2    Park, G.3    Park, J.W.4    Kim, J.E.5
  • 106
    • 84916897484 scopus 로고    scopus 로고
    • Inhibition of histone H3K79 methylation selectively inhibits proliferation, self-renewal and metastatic potential of breast cancer
    • Zhang L, Deng L, Chen F et al. Inhibition of histone H3K79 methylation selectively inhibits proliferation, self-renewal and metastatic potential of breast cancer. Oncotarget 5(21), 10665-10677 (2014).
    • (2014) Oncotarget , vol.5 , Issue.21 , pp. 10665-10677
    • Zhang, L.1    Deng, L.2    Chen, F.3
  • 107
    • 84923254803 scopus 로고    scopus 로고
    • Mixed lineage rearranged leukaemia: Pathogenesis and targeting DOT1L
    • Stein EM, Tallman MS. Mixed lineage rearranged leukaemia: pathogenesis and targeting DOT1L. Curr. Opin. Hematol. 22(2), 92-96 (2015).
    • (2015) Curr. Opin. Hematol. , vol.22 , Issue.2 , pp. 92-96
    • Stein, E.M.1    Tallman, M.S.2
  • 108
    • 84871737742 scopus 로고    scopus 로고
    • Catalytic site remodelling of the DOT1L methyltransferase by selective inhibitors
    • Yu W, Chory EJ, Wernimont AK 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    Chory, E.J.2    Wernimont, A.K.3
  • 109
    • 84868101932 scopus 로고    scopus 로고
    • Conformational adaptation drives potent, selective and durable inhibition of the human protein methyltransferase DOT1L
    • Basavapathruni A, Jin L, Daigle SR et al. Conformational adaptation drives potent, selective and durable inhibition of the human protein methyltransferase DOT1L. Chem. Biol. Drug Des. 80(6), 971-980 (2012).
    • (2012) Chem. Biol. Drug Des. , vol.80 , Issue.6 , pp. 971-980
    • Basavapathruni, A.1    Jin, L.2    Daigle, S.R.3
  • 110
    • 11144332565 scopus 로고    scopus 로고
    • Histone demethylation mediated by the nuclear amine oxidase homolog LSD1
    • Shi Y, Lan F, Matson C et al. Histone demethylation mediated by the nuclear amine oxidase homolog LSD1. Cell 119(7), 941-953 (2004).
    • (2004) Cell , vol.119 , Issue.7 , pp. 941-953
    • Shi, Y.1    Lan, F.2    Matson, C.3
  • 111
    • 0035852637 scopus 로고    scopus 로고
    • CoREST is an integral component of the CoREST-human histone deacetylase complex
    • You A, Tong JK, Grozinger CM, Schreiber SL. CoREST is an integral component of the CoREST-human histone deacetylase complex. Proc. Natl Acad. Sci. USA 98(4), 1454-1458 (2001).
    • (2001) Proc. Natl Acad. Sci. USA , vol.98 , Issue.4 , pp. 1454-1458
    • You, A.1    Tong, J.K.2    Grozinger, C.M.3    Schreiber, S.L.4
  • 112
    • 78649664485 scopus 로고    scopus 로고
    • Structural insights into histone lysine demethylation
    • Hou H, Yu H. Structural insights into histone lysine demethylation. Curr. Opin. Struct. Biol. 20(6), 739-748 (2010).
    • (2010) Curr. Opin. Struct. Biol. , vol.20 , Issue.6 , pp. 739-748
    • Hou, H.1    Yu, H.2
  • 113
    • 68749108259 scopus 로고    scopus 로고
    • LSD1 is a subunit of the NuRD complex and targets the metastasis programs in breast cancer
    • Wang Y, Zhang H, Chen Y et al. LSD1 is a subunit of the NuRD complex and targets the metastasis programs in breast cancer. Cell 138(4), 660-672 (2009).
    • (2009) Cell , vol.138 , Issue.4 , pp. 660-672
    • Wang, Y.1    Zhang, H.2    Chen, Y.3
  • 114
    • 24144462170 scopus 로고    scopus 로고
    • LSD1 demethylates repressive histone marks to promote androgen-receptordependent transcription
    • Metzger E, Wissmann M, Yin N et al. LSD1 demethylates repressive histone marks to promote androgen-receptordependent transcription. Nature 437(7057), 436-439 (2005).
    • (2005) Nature , vol.437 , Issue.7057 , pp. 436-439
    • Metzger, E.1    Wissmann, M.2    Yin, N.3
  • 115
    • 33845762289 scopus 로고    scopus 로고
    • Androgen receptor coactivators lysine-specific histone demethylase 1 and four and a half LIM domain protein 2 predict risk of prostate cancer recurrence
    • Kahl P, Gullotti L, Heukamp LC et al. Androgen receptor coactivators lysine-specific histone demethylase 1 and four and a half LIM domain protein 2 predict risk of prostate cancer recurrence. Cancer Res. 66(23), 11341-11347 (2006).
    • (2006) Cancer Res , vol.66 , Issue.23 , pp. 11341-11347
    • Kahl, P.1    Gullotti, L.2    Heukamp, L.C.3
  • 116
    • 78649815150 scopus 로고    scopus 로고
    • Overexpression of LSD1 contributes to human carcinogenesis through chromatin regulation in various cancers
    • Hayami S, Kelly JD, Cho HS et al. Overexpression of LSD1 contributes to human carcinogenesis through chromatin regulation in various cancers. Int. J. Cancer 128(3), 574-586 (2011).
    • (2011) Int. J. Cancer , vol.128 , Issue.3 , pp. 574-586
    • Hayami, S.1    Kelly, J.D.2    Cho, H.S.3
  • 117
    • 80255137124 scopus 로고    scopus 로고
    • Role of androgen receptor and associated lysine-demethylase coregulators, LSD1 and JMJD2A, in localized and advanced human bladder cancer
    • Kauffman EC, Robinson BD, Downes MJ et al. Role of androgen receptor and associated lysine-demethylase coregulators, LSD1 and JMJD2A, in localized and advanced human bladder cancer. Mol. Carcinog. 50(12), 931-944 (2011).
    • (2011) Mol. Carcinog. , vol.50 , Issue.12 , pp. 931-944
    • Kauffman, E.C.1    Robinson, B.D.2    Downes, M.J.3
  • 118
    • 62449197931 scopus 로고    scopus 로고
    • Lysine-specific demethylase 1 is strongly expressed in poorly differentiated neuroblastoma: Implications for therapy
    • Schulte JH, Lim S, Schramm A et al. Lysine-specific demethylase 1 is strongly expressed in poorly differentiated neuroblastoma: implications for therapy. Cancer Res. 69(5), 2065-2071 (2009).
    • (2009) Cancer Res , vol.69 , Issue.5 , pp. 2065-2071
    • Schulte, J.H.1    Lim, S.2    Schramm, A.3
  • 119
    • 33745187327 scopus 로고    scopus 로고
    • Histone H3 lysine 4 demethylation is a target of nonselective antidepressive medications
    • Lee MG, Wynder C, Schmidt DM, McCafferty DG, Shiekhattar R. Histone H3 lysine 4 demethylation is a target of nonselective antidepressive medications. Chem. Biol. 13(6), 563-567 (2006).
    • (2006) Chem. Biol. , vol.13 , Issue.6 , pp. 563-567
    • Lee, M.G.1    Wynder, C.2    Schmidt, D.M.3    McCafferty, D.G.4    Shiekhattar, R.5
  • 120
    • 84923271907 scopus 로고    scopus 로고
    • NCL1, a highly selective lysine-specific demethylase 1 inhibitor, suppresses prostate cancer without adverse effect
    • Etani T, Suzuki T, Naiki T et al. NCL1, a highly selective lysine-specific demethylase 1 inhibitor, suppresses prostate cancer without adverse effect. Oncotarget 6(5), 2865-2878 (2015).
    • (2015) Oncotarget , vol.6 , Issue.5 , pp. 2865-2878
    • Etani, T.1    Suzuki, T.2    Naiki, T.3
  • 121
    • 33747455678 scopus 로고    scopus 로고
    • JmjC-domain-containing proteins and histone demethylation
    • Klose RJ, Kallin EM, Zhang Y. JmjC-domain-containing proteins and histone demethylation. Nat. Rev. Genet. 7(9), 715-727 (2006).
    • (2006) Nat. Rev. Genet. , vol.7 , Issue.9 , pp. 715-727
    • Klose, R.J.1    Kallin, E.M.2    Zhang, Y.3
  • 122
    • 84889573275 scopus 로고    scopus 로고
    • Histone lysine demethylases as targets for anticancer therapy
    • Hojfeldt JW, Agger K, Helin K. Histone lysine demethylases as targets for anticancer therapy. Nat. Rev. Drug Discov. 12(12), 917-930 (2013).
    • (2013) Nat. Rev. Drug Discov. , vol.12 , Issue.12 , pp. 917-930
    • Hojfeldt, J.W.1    Agger, K.2    Helin, K.3
  • 123
    • 84877861510 scopus 로고    scopus 로고
    • KDM4/JMJD2 histone demethylases: Epigenetic regulators in cancer cells
    • Berry WL, Janknecht R. KDM4/JMJD2 histone demethylases: epigenetic regulators in cancer cells. Cancer Res. 73(10), 2936-2942 (2013).
    • (2013) Cancer Res , vol.73 , Issue.10 , pp. 2936-2942
    • Berry, W.L.1    Janknecht, R.2
  • 124
    • 33746332412 scopus 로고    scopus 로고
    • The putative oncogene GASC1 demethylates tri-and dimethylated lysine 9 on histone H3
    • Cloos PA, Christensen J, Agger K et al. The putative oncogene GASC1 demethylates tri-and dimethylated lysine 9 on histone H3. Nature 442(7100), 307-311 (2006).
    • (2006) Nature , vol.442 , Issue.7100 , pp. 307-311
    • Cloos, P.A.1    Christensen, J.2    Agger, K.3
  • 125
    • 84907526739 scopus 로고    scopus 로고
    • Contrasting roles of histone 3 lysine 27 demethylases in acute lymphoblastic leukaemia
    • Ntziachristos P, Tsirigos A, Welstead GG et al. Contrasting roles of histone 3 lysine 27 demethylases in acute lymphoblastic leukaemia. Nature 514(7523), 513-517 (2014).
    • (2014) Nature , vol.514 , Issue.7523 , pp. 513-517
    • Ntziachristos, P.1    Tsirigos, A.2    Welstead, G.G.3
  • 126
    • 80052849547 scopus 로고    scopus 로고
    • Targeting histone lysine demethylases by truncating the histone 3 tail to obtain selective substrate-based inhibitors
    • Lohse B, Nielsen AL, Kristensen JB et al. Targeting histone lysine demethylases by truncating the histone 3 tail to obtain selective substrate-based inhibitors. Angew. Chem. Int. Ed. Engl. 50(39), 9100-9103 (2011).
    • (2011) Angew. Chem. Int. Ed. Engl. , vol.50 , Issue.39 , pp. 9100-9103
    • Lohse, B.1    Nielsen, A.L.2    Kristensen, J.B.3
  • 127
    • 84889595831 scopus 로고    scopus 로고
    • A small molecule modulates Jumonji histone demethylase activity and selectively inhibits cancer growth
    • Wang L, Chang J, Varghese D et al. A small molecule modulates Jumonji histone demethylase activity and selectively inhibits cancer growth. Nat. Commun. 4, 2035 (2013).
    • (2013) Nat. Commun. , vol.4 , pp. 2035
    • Wang, L.1    Chang, J.2    Varghese, D.3
  • 128
    • 78649528262 scopus 로고    scopus 로고
    • Quantitative highthroughput screening identifies 8-hydroxyquinolines as cell-active histone demethylase inhibitors
    • King ON, Li XS, Sakurai M et al. Quantitative highthroughput screening identifies 8-hydroxyquinolines as cell-active histone demethylase inhibitors. PLoS ONE 5(11), e15535 (2010).
    • (2010) PLoS ONE , vol.5 , Issue.11 , pp. e15535
    • King, O.N.1    Li, X.S.2    Sakurai, M.3
  • 129
    • 79955035374 scopus 로고    scopus 로고
    • Inhibition of histone demethylases by 4-carboxy-2,2-bipyridyl compounds
    • Chang KH, King ON, Tumber A et al. Inhibition of histone demethylases by 4-carboxy-2,2-bipyridyl compounds. ChemMedChem 6(5), 759-764 (2011).
    • (2011) ChemMedChem , vol.6 , Issue.5 , pp. 759-764
    • Chang, K.H.1    King, O.N.2    Tumber, A.3
  • 130
    • 84896706026 scopus 로고    scopus 로고
    • A cell-permeable ester derivative of the JmjC histone demethylase inhibitor IOX1
    • Schiller R, Scozzafava G, Tumber A et al. A cell-permeable ester derivative of the JmjC histone demethylase inhibitor IOX1. ChemMedChem 9(3), 566-571 (2014).
    • (2014) Chem Med Chem , vol.9 , Issue.3 , pp. 566-571
    • Schiller, R.1    Scozzafava, G.2    Tumber, A.3
  • 131
    • 17144439749 scopus 로고
    • Radiographic diagnosis of sliding hiatal hernias in children
    • Steller P. [Radiographic diagnosis of sliding hiatal hernias in children]. Bratisl. Lek. Listy 54(2), 224-229 (1970).
    • (1970) Bratisl. Lek. Listy , vol.54 , Issue.2 , pp. 224-229
    • Steller, P.1
  • 132
    • 84899973908 scopus 로고    scopus 로고
    • Targeting bromodomains: Epigenetic readers of lysine acetylation
    • Filippakopoulos P, Knapp S. Targeting bromodomains: epigenetic readers of lysine acetylation. Nat. Rev. Drug Discov. 13(5), 337-356 (2014).
    • (2014) Nat. Rev. Drug Discov. , vol.13 , Issue.5 , pp. 337-356
    • Filippakopoulos, P.1    Knapp, S.2
  • 133
    • 84878450298 scopus 로고    scopus 로고
    • Bromodomains as therapeutic targets in cancer
    • Barbieri I, Cannizzaro E, Dawson MA. Bromodomains as therapeutic targets in cancer. Brief. Funct. Genom. 12(3), 219-230 (2013).
    • (2013) Brief. Funct. Genom. , vol.12 , Issue.3 , pp. 219-230
    • Barbieri, I.1    Cannizzaro, E.2    Ma, D.3
  • 134
    • 84913610674 scopus 로고    scopus 로고
    • Epigenetic drug therapy based on bromodomain inhibition
    • Bayarsaihan D, Shin DG. Epigenetic drug therapy based on bromodomain inhibition. Epigenomics 6(5), 473-476 (2014).
    • (2014) Epigenomics , vol.6 , Issue.5 , pp. 473-476
    • Bayarsaihan, D.1    Shin, D.G.2
  • 135
    • 84993709912 scopus 로고    scopus 로고
    • Inhibition of bromodomain and extra-terminal proteins (BET) as a potential therapeutic approach in haematological malignancies: Emerging preclinical and clinical evidence
    • Chaidos A, Caputo V, Karadimitris A. Inhibition of bromodomain and extra-terminal proteins (BET) as a potential therapeutic approach in haematological malignancies: emerging preclinical and clinical evidence. Ther. Adv. Hematol. 6(3), 128-141 (2015).
    • (2015) Ther. Adv. Hematol. , vol.6 , Issue.3 , pp. 128-141
    • Chaidos, A.1    Caputo, V.2    Karadimitris, A.3
  • 136
    • 85027953829 scopus 로고    scopus 로고
    • Bromodomains: Pockets with therapeutic potential
    • Papavassiliou KA, Papavassiliou AG. Bromodomains: pockets with therapeutic potential. Trends Mol. Med. 20(9), 477-478 (2014).
    • (2014) Trends Mol. Med. , vol.20 , Issue.9 , pp. 477-478
    • Papavassiliou, K.A.1    Papavassiliou, A.G.2
  • 137
    • 78650847770 scopus 로고    scopus 로고
    • Selective inhibition of BET bromodomains
    • Filippakopoulos P, Qi J, Picaud S et al. Selective inhibition of BET bromodomains. Nature 468(7327), 1067-1073 (2010).
    • (2010) Nature , vol.468 , Issue.7327 , pp. 1067-1073
    • Filippakopoulos, P.1    Qi, J.2    Picaud, S.3
  • 138
    • 79958078949 scopus 로고    scopus 로고
    • Discovery and characterization of small molecule inhibitors of the BET family bromodomains
    • Chung CW, Coste H, White JH et al. Discovery and characterization of small molecule inhibitors of the BET family bromodomains. J. Med. Chem. 54(11), 3827-3838 (2011).
    • (2011) J. Med. Chem. , vol.54 , Issue.11 , pp. 3827-3838
    • Chung, C.W.1    Coste, H.2    White, J.H.3
  • 139
    • 14744276712 scopus 로고    scopus 로고
    • Selective small molecules blocking HIV-1 Tat and coactivator PCAF association
    • Zeng L, Li J, Muller M et al. Selective small molecules blocking HIV-1 Tat and coactivator PCAF association. J. Am. Chem. Soc. 127(8), 2376-2377 (2005).
    • (2005) J. Am. Chem. Soc. , vol.127 , Issue.8 , pp. 2376-2377
    • Zeng, L.1    Li, J.2    Muller, M.3
  • 140
    • 58149174109 scopus 로고    scopus 로고
    • Phase i study of epigenetic modulation with 5-azacytidine and valproic acid in patients with advanced cancers
    • Braiteh F, Soriano AO, Garcia-Manero G et al. Phase I study of epigenetic modulation with 5-azacytidine and valproic acid in patients with advanced cancers. Clin. Cancer Res. 14(19), 6296-6301 (2008).
    • (2008) Clin. Cancer Res. , vol.14 , Issue.19 , pp. 6296-6301
    • Braiteh, F.1    Soriano, A.O.2    Garcia-Manero, G.3
  • 141
    • 0034326784 scopus 로고    scopus 로고
    • Reversal of drug resistance in human tumor xenografts by 2-deoxy-5-azacytidine-induced demethylation of the hMLH1 gene promoter
    • Plumb JA, Strathdee G, Sludden J, Kaye SB, Brown R. Reversal of drug resistance in human tumor xenografts by 2-deoxy-5-azacytidine-induced demethylation of the hMLH1 gene promoter. Cancer Res. 60(21), 6039-6044 (2000).
    • (2000) Cancer Res , vol.60 , Issue.21 , pp. 6039-6044
    • Plumb, J.A.1    Strathdee, G.2    Sludden, J.3    Kaye, S.B.4    Brown, R.5
  • 142
    • 84655163371 scopus 로고    scopus 로고
    • Combination epigenetic therapy has efficacy in patients with refractory advanced non-small cell lung cancer
    • Juergens RA, Wrangle J, Vendetti FP et al. Combination epigenetic therapy has efficacy in patients with refractory advanced non-small cell lung cancer. Cancer Discov. 1(7), 598-607 (2011).
    • (2011) Cancer Discov , vol.1 , Issue.7 , pp. 598-607
    • Juergens, R.A.1    Wrangle, J.2    Vendetti, F.P.3
  • 143
    • 84892614202 scopus 로고    scopus 로고
    • Gemtuzumab ozogamicin in combination with vorinostat and azacitidine in older patients with relapsed or refractory acute myeloid leukemia: A Phase I/II study
    • Walter RB, Medeiros BC, Gardner KM et al. Gemtuzumab ozogamicin in combination with vorinostat and azacitidine in older patients with relapsed or refractory acute myeloid leukemia: a Phase I/II study. Haematologica 99(1), 54-59 (2014).
    • (2014) Haematologica , vol.99 , Issue.1 , pp. 54-59
    • Walter, R.B.1    Medeiros, B.C.2    Gardner, K.M.3
  • 144
    • 84942860046 scopus 로고    scopus 로고
    • Preclinical activity of combined HDAC and KDM1A inhibition in glioblastoma
    • Epub ahead of print
    • Singh MM, Johnson B, Venkatarayan A et al. Preclinical activity of combined HDAC and KDM1A inhibition in glioblastoma. Neuro Oncol. doi:10.1093/neuonc/nov041 (2015) (Epub ahead of print).
    • (2015) Neuro Oncol
    • Singh, M.M.1    Johnson, B.2    Venkatarayan, A.3
  • 145
    • 84927547043 scopus 로고    scopus 로고
    • EZH2 inhibition sensitizes BRG1 and EGFR mutant lung tumours to TopoII inhibitors
    • Fillmore CM, Xu C, Desai PT et al. EZH2 inhibition sensitizes BRG1 and EGFR mutant lung tumours to TopoII inhibitors. Nature 520(7546), 239-242 (2015).
    • (2015) Nature , vol.520 , Issue.7546 , pp. 239-242
    • Fillmore, C.M.1    Xu, C.2    Desai, P.T.3
  • 146
    • 84907291965 scopus 로고    scopus 로고
    • DOT1L inhibitor EPZ-5676 displays synergistic antiproliferative activity in combination with standard of care drugs and hypomethylating agents in MLL-rearranged leukemia cells
    • Klaus CR, Iwanowicz D, Johnston D et al. DOT1L inhibitor EPZ-5676 displays synergistic antiproliferative activity in combination with standard of care drugs and hypomethylating agents in MLL-rearranged leukemia cells. J. Pharmacol. Exp. Ther. 350(3), 646-656 (2014).
    • (2014) J. Pharmacol. Exp. Ther. , vol.350 , Issue.3 , pp. 646-656
    • Klaus, C.R.1    Iwanowicz, D.2    Johnston, D.3
  • 148
    • 53949090314 scopus 로고    scopus 로고
    • Restoration of tumor suppressor miR-34 inhibits human p53-mutant gastric cancer tumorspheres
    • Ji Q, Hao X, Meng Y et al. Restoration of tumor suppressor miR-34 inhibits human p53-mutant gastric cancer tumorspheres. BMC Cancer 8, 266 (2008).
    • (2008) BMC Cancer , vol.8 , pp. 266
    • Ji, Q.1    Hao, X.2    Meng, Y.3
  • 149
    • 84862860233 scopus 로고    scopus 로고
    • Anti-miR-155 oligonucleotide enhances chemosensitivity of U251 cell to taxol by inducing apoptosis
    • Meng W, Jiang L, Lu L et al. Anti-miR-155 oligonucleotide enhances chemosensitivity of U251 cell to taxol by inducing apoptosis. Cell Biol. Int. 36(7), 653-659 (2012).
    • (2012) Cell Biol. Int. , vol.36 , Issue.7 , pp. 653-659
    • Meng, W.1    Jiang, L.2    Lu, L.3
  • 150
    • 77957361864 scopus 로고    scopus 로고
    • Targeting microRNAs in cancer: Rationale, strategies and challenges
    • Garzon R, Marcucci G, Croce CM. Targeting microRNAs in cancer: rationale, strategies and challenges. Nat. Rev. Drug Discov. 9(10), 775-789 (2010).
    • (2010) Nat. Rev. Drug Discov. , vol.9 , Issue.10 , pp. 775-789
    • Garzon, R.1    Marcucci, G.2    Croce, C.M.3
  • 151
    • 83255192141 scopus 로고    scopus 로고
    • Developing therapeutic microRNAs for cancer
    • Bader AG, Brown D, Stoudemire J, Lammers P. Developing therapeutic microRNAs for cancer. Gene Ther. 18(12), 1121-1126 (2011).
    • (2011) Gene Ther , vol.18 , Issue.12 , pp. 1121-1126
    • Bader, A.G.1    Brown, D.2    Stoudemire, J.3    Lammers, P.4
  • 152
    • 84878858306 scopus 로고    scopus 로고
    • Therapeutic delivery of microRNA-29b by cationic lipoplexes for lung cancer Molecular therapy
    • Wu Y, Crawford M, Mao Y et al. Therapeutic delivery of microRNA-29b by cationic lipoplexes for lung cancer. Molecular therapy. Nucleic Acids 2, e84 (2013).
    • (2013) Nucleic Acids , vol.2 , pp. e84
    • Wu, Y.1    Crawford, M.2    Mao, Y.3
  • 153
    • 33645135862 scopus 로고    scopus 로고
    • Anti-miRNA oligonucleotides (AMOs): Ammunition to target miRNAs implicated in human disease
    • Weiler J, Hunziker J, Hall J. Anti-miRNA oligonucleotides (AMOs): ammunition to target miRNAs implicated in human disease Gene Ther. 13(6), 496-502 (2006).
    • (2006) Gene Ther , vol.13 , Issue.6 , pp. 496-502
    • Weiler, J.1    Hunziker, J.2    Hall, J.3
  • 154
  • 155
    • 34548316982 scopus 로고    scopus 로고
    • MicroRNA sponges: Competitive inhibitors of small RNAs in mammalian cells
    • Ebert MS, Neilson JR, Sharp PA. MicroRNA sponges: competitive inhibitors of small RNAs in mammalian cells. Nat. Methods 4(9), 721-726 (2007).
    • (2007) Nat. Methods , vol.4 , Issue.9 , pp. 721-726
    • Ebert, M.S.1    Neilson, J.R.2    Sharp, P.A.3
  • 156
    • 72949098057 scopus 로고    scopus 로고
    • Therapeutic microRNA strategies in human cancer
    • Li C, Feng Y, Coukos G, Zhang L. Therapeutic microRNA strategies in human cancer. AAPS J. 11(4), 747-757 (2009).
    • (2009) AAPS J , vol.11 , Issue.4 , pp. 747-757
    • Li, C.1    Feng, Y.2    Coukos, G.3    Zhang, L.4
  • 157
    • 84873351133 scopus 로고    scopus 로고
    • The long non-coding RNAs, a new cancer diagnostic and therapeutic gold mine
    • Qi P, Du X. The long non-coding RNAs, a new cancer diagnostic and therapeutic gold mine. Mod. Pathol. 26(2), 155-165 (2013).
    • (2013) Mod. Pathol. , vol.26 , Issue.2 , pp. 155-165
    • Qi, P.1    Du, X.2
  • 158
    • 78049308492 scopus 로고    scopus 로고
    • RNAi screen indicates widespread biological function for human natural antisense transcripts
    • Epub ahead of print
    • Faghihi MA, Kocerha J, Modarresi F et al. RNAi screen indicates widespread biological function for human natural antisense transcripts. PLoS ONE doi: 10.1371/journal. pone.0013177 (2010) (Epub ahead of print).
    • (2010) PLoS ONE
    • Ma, F.1    Kocerha, J.2    Modarresi, F.3
  • 159
    • 84860353437 scopus 로고    scopus 로고
    • Inhibition of natural antisense transcripts in vivo results in gene-specific transcriptional upregulation
    • Modarresi F, Faghihi MA, Lopez-Toledano MA et al. Inhibition of natural antisense transcripts in vivo results in gene-specific transcriptional upregulation. Nat. Biotechnol. 30(5), 453-459 (2012).
    • (2012) Nat. Biotechnol. , vol.30 , Issue.5 , pp. 453-459
    • Modarresi, F.1    Ma, F.2    Ma, L.3
  • 160
    • 77949512140 scopus 로고    scopus 로고
    • RNA targeting therapeutics: Molecular mechanisms of antisense oligonucleotides as a therapeutic platform
    • Bennett CF, Swayze EE. RNA targeting therapeutics: molecular mechanisms of antisense oligonucleotides as a therapeutic platform. Annu. Rev. Pharmacol. Toxicol. 50, 259-293 (2010).
    • (2010) Annu. Rev. Pharmacol. Toxicol. , vol.50 , pp. 259-293
    • Bennett, C.F.1    Swayze, E.E.2


* 이 정보는 Elsevier사의 SCOPUS DB에서 KISTI가 분석하여 추출한 것입니다.