메뉴 건너뛰기




Volumn 4 MAY, Issue , 2014, Pages

The therapeutic potential of class I selective histone deacetylase inhibitors in ovarian cancer

Author keywords

Epigenetic therapy; Histone deacetylase inhibitors; Histone deacetylases; Ovarian cancer; Targeted therapy

Indexed keywords

BELINOSTAT; BENZAMIDE; CARBOPLATIN; DEPSIPEPTIDE; ENTINOSTAT; GEMCITABINE; HISTONE DEACETYLASE; HISTONE DEACETYLASE 3; HISTONE DEACETYLASE INHIBITOR; PACLITAXEL; PROTEIN P21; ROMIDEPSIN; VALPROIC ACID; VORINOSTAT;

EID: 84904636264     PISSN: None     EISSN: 2234943X     Source Type: Journal    
DOI: 10.3389/fonc.2014.00111     Document Type: Review
Times cited : (40)

References (100)
  • 1
    • 84892805731 scopus 로고    scopus 로고
    • Cancer statistics, 2014
    • doi: 10.3322/caac.21208
    • Siegel R, Ma J, Zou Z, Jemal A. Cancer statistics, 2014. CA Cancer J Clin (2014) 64(1):9-29. doi: 10.3322/caac.21208
    • (2014) CA Cancer J Clin , vol.64 , Issue.1 , pp. 9-29
    • Siegel, R.1    Ma, J.2    Zou, Z.3    Jemal, A.4
  • 2
    • 79959328816 scopus 로고    scopus 로고
    • Molecular pathogenesis and extraovarian origin of epithelial ovarian cancer - shifting the paradigm
    • doi:10.1016/j.humpath.2011.03.003
    • Kurman RJ, Shih Ie M. Molecular pathogenesis and extraovarian origin of epithelial ovarian cancer - shifting the paradigm. Hum Pathol (2011) 42(7):918-31. doi:10.1016/j.humpath.2011.03.003
    • (2011) Hum Pathol , vol.42 , Issue.7 , pp. 918-931
    • Kurman, R.J.1    Shih Ie, M.2
  • 3
    • 79959838081 scopus 로고    scopus 로고
    • Integrated genomic analyses of ovarian carcinoma
    • Cancer Genome Atlas Research Network, doi:10.1038/nature10166
    • Cancer Genome Atlas Research Network. Integrated genomic analyses of ovarian carcinoma. Nature (2011) 474(7353):609-15. doi:10.1038/nature10166
    • (2011) Nature , vol.474 , Issue.7353 , pp. 609-615
  • 4
    • 84864026311 scopus 로고    scopus 로고
    • BRCA mutation frequency and patterns of treatment response in BRCA mutation-positive women with ovarian cancer: a report from the Australian Ovarian Cancer Study Group
    • doi:10.1200/JCO.2011.39.8545
    • Alsop K, Fereday S, Meldrum C, Defazio A, Emmanuel C, George J, et al. BRCA mutation frequency and patterns of treatment response in BRCA mutation-positive women with ovarian cancer: a report from the Australian Ovarian Cancer Study Group. J Clin Oncol (2012) 30(21):2654-63. doi:10.1200/JCO.2011.39.8545
    • (2012) J Clin Oncol , vol.30 , Issue.21 , pp. 2654-2663
    • Alsop, K.1    Fereday, S.2    Meldrum, C.3    Defazio, A.4    Emmanuel, C.5    George, J.6
  • 5
    • 77955039099 scopus 로고    scopus 로고
    • Oral poly(ADP-ribose) polymerase inhibitor olaparib in patients with BRCA1 or BRCA2 mutations and recurrent ovarian cancer: a proof-of-concept trial
    • doi:10.1016/S0140-6736(10)60893-8
    • Audeh MW, Carmichael J, Penson RT, Friedlander M, Powell B, Bell-McGuinn KM, et al. Oral poly(ADP-ribose) polymerase inhibitor olaparib in patients with BRCA1 or BRCA2 mutations and recurrent ovarian cancer: a proof-of-concept trial. Lancet (2010) 376(9737):245-51. doi:10.1016/S0140-6736(10)60893-8
    • (2010) Lancet , vol.376 , Issue.9737 , pp. 245-251
    • Audeh, M.W.1    Carmichael, J.2    Penson, R.T.3    Friedlander, M.4    Powell, B.5    Bell-McGuinn, K.M.6
  • 6
    • 67650471685 scopus 로고    scopus 로고
    • Inhibition of poly(ADP-ribose) polymerase in tumors from BRCA mutation carriers
    • doi:10.1056/NEJMoa0900212
    • Fong PC, Boss DS, Yap TA, Tutt A, Wu P, Mergui-Roelvink M, et al. Inhibition of poly(ADP-ribose) polymerase in tumors from BRCA mutation carriers. N Engl J Med (2009) 361(2):123-34. doi:10.1056/NEJMoa0900212
    • (2009) N Engl J Med , vol.361 , Issue.2 , pp. 123-134
    • Fong, P.C.1    Boss, D.S.2    Yap, T.A.3    Tutt, A.4    Wu, P.5    Mergui-Roelvink, M.6
  • 7
    • 77954032829 scopus 로고    scopus 로고
    • Poly(ADP)-ribose polymerase inhibition: frequent durable responses in BRCA carrier ovarian cancer correlating with platinum-free interval
    • doi:10.1200/JCO.2009.26.9589
    • Fong PC, Yap TA, Boss DS, Carden CP, Mergui-Roelvink M, Gourley C, et al. Poly(ADP)-ribose polymerase inhibition: frequent durable responses in BRCA carrier ovarian cancer correlating with platinum-free interval. J Clin Oncol (2010) 28(15):2512-9. doi:10.1200/JCO.2009.26.9589
    • (2010) J Clin Oncol , vol.28 , Issue.15 , pp. 2512-2519
    • Fong, P.C.1    Yap, T.A.2    Boss, D.S.3    Carden, C.P.4    Mergui-Roelvink, M.5    Gourley, C.6
  • 8
    • 57649101120 scopus 로고    scopus 로고
    • Platinum compounds 30 years after the introduction of cisplatin: implications for the treatment of ovarian cancer
    • doi:10.1016/j.ygyno.2008.09.034
    • Muggia F. Platinum compounds 30 years after the introduction of cisplatin: implications for the treatment of ovarian cancer. Gynecol Oncol (2009) 112(1):275-81. doi:10.1016/j.ygyno.2008.09.034
    • (2009) Gynecol Oncol , vol.112 , Issue.1 , pp. 275-281
    • Muggia, F.1
  • 9
    • 60749137451 scopus 로고    scopus 로고
    • Beyond chemotherapy: targeted therapies in ovarian cancer
    • doi:10.1038/nrc2583
    • Yap TA, Carden CP, Kaye SB. Beyond chemotherapy: targeted therapies in ovarian cancer. Nat Rev Cancer (2009) 9(3):167-81. doi:10.1038/nrc2583
    • (2009) Nat Rev Cancer , vol.9 , Issue.3 , pp. 167-181
    • Yap, T.A.1    Carden, C.P.2    Kaye, S.B.3
  • 10
    • 19744368302 scopus 로고    scopus 로고
    • Cancer of the ovary
    • doi:10.1056/NEJMra041842
    • Cannistra SA. Cancer of the ovary. N Engl J Med (2004) 351(24):2519-29. doi:10.1056/NEJMra041842
    • (2004) N Engl J Med , vol.351 , Issue.24 , pp. 2519-2529
    • Cannistra, S.A.1
  • 11
    • 0031466222 scopus 로고    scopus 로고
    • Update of the NCCN ovarian cancer practice guidelines.
    • Ozols RF. Update of the NCCN ovarian cancer practice guidelines. Oncology (Williston Park) (1997) 11(11A):95-105.
    • (1997) Oncology (Williston Park) , vol.11 , Issue.11 , pp. 95-105
    • Ozols, R.F.1
  • 12
    • 0036222963 scopus 로고    scopus 로고
    • Systematic review of first-line chemotherapy for newly diagnosed postoperative patients with stage II, III, or IV epithelial ovarian cancer
    • doi:10.1006/gyno.2001.6552
    • Covens A, Carey M, Bryson P, Verma S, Fung Kee Fung M, Johnston M. Systematic review of first-line chemotherapy for newly diagnosed postoperative patients with stage II, III, or IV epithelial ovarian cancer. Gynecol Oncol (2002) 85(1):71-80. doi:10.1006/gyno.2001.6552
    • (2002) Gynecol Oncol , vol.85 , Issue.1 , pp. 71-80
    • Covens, A.1    Carey, M.2    Bryson, P.3    Verma, S.4    Fung Kee Fung, M.5    Johnston, M.6
  • 13
    • 84882998935 scopus 로고    scopus 로고
    • Long-term results of dose-dense paclitaxel and carboplatin versus conventional paclitaxel and carboplatin for treatment of advanced epithelial ovarian, fallopian tube, or primary peritoneal cancer (JGOG 3016): a randomised, controlled, open-label trial
    • doi:10.1016/S1470-2045(13)70363-2
    • Katsumata N, Yasuda M, Isonishi S, Takahashi F, Michimae H, Kimura E, et al. Long-term results of dose-dense paclitaxel and carboplatin versus conventional paclitaxel and carboplatin for treatment of advanced epithelial ovarian, fallopian tube, or primary peritoneal cancer (JGOG 3016): a randomised, controlled, open-label trial. Lancet Oncol (2013) 14(10):1020-6. doi:10.1016/S1470-2045(13)70363-2
    • (2013) Lancet Oncol , vol.14 , Issue.10 , pp. 1020-1026
    • Katsumata, N.1    Yasuda, M.2    Isonishi, S.3    Takahashi, F.4    Michimae, H.5    Kimura, E.6
  • 14
    • 30044438368 scopus 로고    scopus 로고
    • Intraperitoneal cisplatin and paclitaxel in ovarian cancer
    • doi:10.1056/NEJMoa052985
    • Armstrong DK, Bundy B, Wenzel L, Huang HQ, Baergen R, Lele S, et al. Intraperitoneal cisplatin and paclitaxel in ovarian cancer. N Engl J Med (2006) 354(1):34-43. doi:10.1056/NEJMoa052985
    • (2006) N Engl J Med , vol.354 , Issue.1 , pp. 34-43
    • Armstrong, D.K.1    Bundy, B.2    Wenzel, L.3    Huang, H.Q.4    Baergen, R.5    Lele, S.6
  • 15
    • 84892942381 scopus 로고    scopus 로고
    • New and emerging HDAC inhibitors for cancer treatment
    • doi:10.1172/JCI69738
    • West AC, Johnstone RW. New and emerging HDAC inhibitors for cancer treatment. J Clin Invest (2014) 124(1):30-9. doi:10.1172/JCI69738
    • (2014) J Clin Invest , vol.124 , Issue.1 , pp. 30-39
    • West, A.C.1    Johnstone, R.W.2
  • 16
    • 67349285731 scopus 로고    scopus 로고
    • Enhancing the apoptotic and therapeutic effects of HDAC inhibitors
    • doi:10.1016/j.canlet.2009.02.042
    • Frew AJ, Johnstone RW, Bolden JE. Enhancing the apoptotic and therapeutic effects of HDAC inhibitors. Cancer Lett (2009) 280(2):125-33. doi:10.1016/j.canlet.2009.02.042
    • (2009) Cancer Lett , vol.280 , Issue.2 , pp. 125-133
    • Frew, A.J.1    Johnstone, R.W.2    Bolden, J.E.3
  • 17
    • 57749170458 scopus 로고    scopus 로고
    • The many roles of histone deacetylases in development and physiology: implications for disease and therapy
    • doi:10.1038/nrg2485
    • Haberland M, Montgomery RL, Olson EN. The many roles of histone deacetylases in development and physiology: implications for disease and therapy. Nat Rev Genet (2009) 10(1):32-42. doi:10.1038/nrg2485
    • (2009) Nat Rev Genet , vol.10 , Issue.1 , pp. 32-42
    • Haberland, M.1    Montgomery, R.L.2    Olson, E.N.3
  • 18
    • 41549156540 scopus 로고    scopus 로고
    • Deletion of histone deacetylase 3 reveals critical roles in S phase progression and DNA damage control
    • doi:10.1016/j.molcel.2008.02.030
    • Bhaskara S, Chyla BJ, Amann JM, Knutson SK, Cortez D, Sun ZW, et al. Deletion of histone deacetylase 3 reveals critical roles in S phase progression and DNA damage control. Mol Cell (2008) 30(1):61-72. doi:10.1016/j.molcel.2008.02.030
    • (2008) Mol Cell , vol.30 , Issue.1 , pp. 61-72
    • Bhaskara, S.1    Chyla, B.J.2    Amann, J.M.3    Knutson, S.K.4    Cortez, D.5    Sun, Z.W.6
  • 19
    • 77953170728 scopus 로고    scopus 로고
    • Inhibition of histone deacetylase in cancer cells slows down replication forks, activates dormant origins, and induces DNA damage
    • doi:10.1158/0008-5472.CAN-09-3028
    • Conti C, Leo E, Eichler GS, Sordet O, Martin MM, Fan A, et al. Inhibition of histone deacetylase in cancer cells slows down replication forks, activates dormant origins, and induces DNA damage. Cancer Res (2010) 70(11):4470-80. doi:10.1158/0008-5472.CAN-09-3028
    • (2010) Cancer Res , vol.70 , Issue.11 , pp. 4470-4480
    • Conti, C.1    Leo, E.2    Eichler, G.S.3    Sordet, O.4    Martin, M.M.5    Fan, A.6
  • 20
    • 78249276172 scopus 로고    scopus 로고
    • Hdac3 is essential for the maintenance of chromatin structure and genome stability
    • doi:10.1016/j.ccr.2010.10.022
    • Bhaskara S, Knutson SK, Jiang G, Chandrasekharan MB, Wilson AJ, Zheng S, et al. Hdac3 is essential for the maintenance of chromatin structure and genome stability. Cancer Cell (2010) 18(5):436-47. doi:10.1016/j.ccr.2010.10.022
    • (2010) Cancer Cell , vol.18 , Issue.5 , pp. 436-447
    • Bhaskara, S.1    Knutson, S.K.2    Jiang, G.3    Chandrasekharan, M.B.4    Wilson, A.J.5    Zheng, S.6
  • 21
    • 0036827874 scopus 로고    scopus 로고
    • Novel detection and differential utilization of a c-myc transcriptional block in colon cancer chemoprevention
    • Wilson AJ, Velcich A, Arango D, Kurland AR, Shenoy SM, Pezo RC, et al. Novel detection and differential utilization of a c-myc transcriptional block in colon cancer chemoprevention. Cancer Res (2002) 62(21):6006-10.
    • (2002) Cancer Res , vol.62 , Issue.21 , pp. 6006-6010
    • Wilson, A.J.1    Velcich, A.2    Arango, D.3    Kurland, A.R.4    Shenoy, S.M.5    Pezo, R.C.6
  • 22
    • 31544455853 scopus 로고    scopus 로고
    • Histone deacetylase inhibitor FK228 suppresses the Ras-MAP kinase signaling pathway by upregulating Rap1 and induces apoptosis in malignant melanoma
    • Kobayashi Y, Ohtsuki M, Murakami T, Kobayashi T, Sutheesophon K, Kitayama H, et al. Histone deacetylase inhibitor FK228 suppresses the Ras-MAP kinase signaling pathway by upregulating Rap1 and induces apoptosis in malignant melanoma. Oncogene (2006) 25(4):512-24.
    • (2006) Oncogene , vol.25 , Issue.4 , pp. 512-524
    • Kobayashi, Y.1    Ohtsuki, M.2    Murakami, T.3    Kobayashi, T.4    Sutheesophon, K.5    Kitayama, H.6
  • 23
    • 1842850710 scopus 로고    scopus 로고
    • Involvement of histone deacetylation in ras-induced down-regulation of the metastasis suppressor RECK
    • doi:10.1016/j.cellsig.2003.11.001
    • Chang HC, Liu LT, Hung WC. Involvement of histone deacetylation in ras-induced down-regulation of the metastasis suppressor RECK. Cell Signal (2004) 16(6):675-9. doi:10.1016/j.cellsig.2003.11.001
    • (2004) Cell Signal , vol.16 , Issue.6 , pp. 675-679
    • Chang, H.C.1    Liu, L.T.2    Hung, W.C.3
  • 24
    • 34247107123 scopus 로고    scopus 로고
    • Histone deacetylase inhibitor trichostatin a potentiates doxorubicin-induced apoptosis by up-regulating PTEN expression
    • doi:10.1002/cncr.22585
    • Pan L, Lu J, Wang X, Han L, Zhang Y, Han S, et al. Histone deacetylase inhibitor trichostatin a potentiates doxorubicin-induced apoptosis by up-regulating PTEN expression. Cancer (2007) 109(8):1676-88. doi:10.1002/cncr.22585
    • (2007) Cancer , vol.109 , Issue.8 , pp. 1676-1688
    • Pan, L.1    Lu, J.2    Wang, X.3    Han, L.4    Zhang, Y.5    Han, S.6
  • 25
    • 56049090769 scopus 로고    scopus 로고
    • Acetylation of non-histone proteins modulates cellular signalling at multiple levels
    • doi:10.1016/j.biocel.2008.08.027
    • Spange S, Wagner T, Heinzel T, Kramer OH. Acetylation of non-histone proteins modulates cellular signalling at multiple levels. Int J Biochem Cell Biol (2009) 41(1):185-98. doi:10.1016/j.biocel.2008.08.027
    • (2009) Int J Biochem Cell Biol , vol.41 , Issue.1 , pp. 185-198
    • Spange, S.1    Wagner, T.2    Heinzel, T.3    Kramer, O.H.4
  • 26
    • 58149310725 scopus 로고    scopus 로고
    • Histone deacetylase inhibitors and genomic instability
    • doi:10.1016/j.canlet.2008.06.005
    • Eot-Houllier G, Fulcrand G, Magnaghi-Jaulin L, Jaulin C. Histone deacetylase inhibitors and genomic instability. Cancer Lett (2009) 274(2):169-76. doi:10.1016/j.canlet.2008.06.005
    • (2009) Cancer Lett , vol.274 , Issue.2 , pp. 169-176
    • Eot-Houllier, G.1    Fulcrand, G.2    Magnaghi-Jaulin, L.3    Jaulin, C.4
  • 27
    • 28044471827 scopus 로고    scopus 로고
    • Acetylation and deacetylation of non-histone proteins.
    • doi:10.1016/j.gene.2005.09.010.
    • Glozak MA, Sengupta N, Zhang X, Seto E. Acetylation and deacetylation of non-histone proteins. Gene (2005) 363:15-23. doi:10.1016/j.gene.2005.09.010
    • (2005) Gene , vol.363 , pp. 15-23
    • Glozak, M.A.1    Sengupta, N.2    Zhang, X.3    Seto, E.4
  • 28
    • 0029932598 scopus 로고    scopus 로고
    • A mammalian histone deacetylase related to the yeast transcriptional regulator Rpd3p
    • doi:10.1126/science.272.5260.408
    • Taunton J, Hassig CA, Schreiber SL. A mammalian histone deacetylase related to the yeast transcriptional regulator Rpd3p. Science (1996) 272(5260):408-11. doi:10.1126/science.272.5260.408
    • (1996) Science , vol.272 , Issue.5260 , pp. 408-411
    • Taunton, J.1    Hassig, C.A.2    Schreiber, S.L.3
  • 29
    • 33748451151 scopus 로고    scopus 로고
    • Anticancer activities of histone deacetylase inhibitors
    • doi:10.1038/nrd2133
    • Bolden JE, Peart MJ, Johnstone RW. Anticancer activities of histone deacetylase inhibitors. Nat Rev Drug Discov (2006) 5(9):769-84. doi:10.1038/nrd2133
    • (2006) Nat Rev Drug Discov , vol.5 , Issue.9 , pp. 769-784
    • Bolden, J.E.1    Peart, M.J.2    Johnstone, R.W.3
  • 30
    • 0037115711 scopus 로고    scopus 로고
    • Assembly of the SMRT-histone deacetylase 3 repression complex requires the TCP-1 ring complex
    • doi:10.1101/gad.1037502
    • Guenther MG, Yu J, Kao GD, Yen TJ, Lazar MA. Assembly of the SMRT-histone deacetylase 3 repression complex requires the TCP-1 ring complex. Genes Dev (2002) 16(24):3130-5. doi:10.1101/gad.1037502
    • (2002) Genes Dev , vol.16 , Issue.24 , pp. 3130-3135
    • Guenther, M.G.1    Yu, J.2    Kao, G.D.3    Yen, T.J.4    Lazar, M.A.5
  • 31
    • 0035724413 scopus 로고    scopus 로고
    • The SMRT and N-CoR corepressors are activating cofactors for histone deacetylase 3
    • doi:10.1128/MCB.21.18.6091-6101.2001
    • Guenther MG, Barak O, Lazar MA. The SMRT and N-CoR corepressors are activating cofactors for histone deacetylase 3. Mol Cell Biol (2001) 21(18):6091-101. doi:10.1128/MCB.21.18.6091-6101.2001
    • (2001) Mol Cell Biol , vol.21 , Issue.18 , pp. 6091-6101
    • Guenther, M.G.1    Barak, O.2    Lazar, M.A.3
  • 32
    • 0037088643 scopus 로고    scopus 로고
    • Functional domains of histone deacetylase-3
    • doi:10.1074/jbc.M105993200
    • Yang WM, Tsai SC, Wen YD, Fejer G, Seto E. Functional domains of histone deacetylase-3. J Biol Chem (2002) 277(11):9447-54. doi:10.1074/jbc.M105993200
    • (2002) J Biol Chem , vol.277 , Issue.11 , pp. 9447-9454
    • Yang, W.M.1    Tsai, S.C.2    Wen, Y.D.3    Fejer, G.4    Seto, E.5
  • 33
    • 33846159473 scopus 로고    scopus 로고
    • Cleavage and cytoplasmic relocalization of histone deacetylase 3 are important for apoptosis progression
    • doi:10.1128/MCB.00869-06
    • Escaffit F, Vaute O, Chevillard-Briet M, Segui B, Takami Y, Nakayama T, et al. Cleavage and cytoplasmic relocalization of histone deacetylase 3 are important for apoptosis progression. Mol Cell Biol (2007) 27(2):554-67. doi:10.1128/MCB.00869-06
    • (2007) Mol Cell Biol , vol.27 , Issue.2 , pp. 554-567
    • Escaffit, F.1    Vaute, O.2    Chevillard-Briet, M.3    Segui, B.4    Takami, Y.5    Nakayama, T.6
  • 34
    • 0141570564 scopus 로고    scopus 로고
    • Role of class I and class II histone deacetylases in carcinoma cells using siRNA
    • doi:10.1016/j.bbrc.2003.09.043
    • Glaser KB, Li J, Staver MJ, Wei RQ, Albert DH, Davidsen SK. Role of class I and class II histone deacetylases in carcinoma cells using siRNA. Biochem Biophys Res Commun (2003) 310(2):529-36. doi:10.1016/j.bbrc.2003.09.043
    • (2003) Biochem Biophys Res Commun , vol.310 , Issue.2 , pp. 529-536
    • Glaser, K.B.1    Li, J.2    Staver, M.J.3    Wei, R.Q.4    Albert, D.H.5    Davidsen, S.K.6
  • 35
    • 33744956666 scopus 로고    scopus 로고
    • Histone deacetylase 3 (HDAC3) and other class I HDACs regulate colon cell maturation and p21 expression and are deregulated in human colon cancer
    • 13548-58, doi:10.1074/jbc.M510023200
    • Wilson AJ, Byun DS, Popova N, Murray LB, L'Italien K, Sowa Y, et al. Histone deacetylase 3 (HDAC3) and other class I HDACs regulate colon cell maturation and p21 expression and are deregulated in human colon cancer. J Biol Chem (2006) 281(19):13548-58. doi:10.1074/jbc.M510023200
    • (2006) J Biol Chem , vol.281 , Issue.19
    • Wilson, A.J.1    Byun, D.S.2    Popova, N.3    Murray, L.B.4    L'Italien, K.5    Sowa, Y.6
  • 36
    • 34548226848 scopus 로고    scopus 로고
    • Drug-induced inactivation or gene silencing of class I histone deacetylases suppresses ovarian cancer cell growth: implications for therapy
    • doi:10.4161/cbt.6.5.4007
    • Khabele D, Son DS, Parl AK, Goldberg GL, Augenlicht LH, Mariadason JM, et al. Drug-induced inactivation or gene silencing of class I histone deacetylases suppresses ovarian cancer cell growth: implications for therapy. Cancer Biol Ther (2007) 6(5):795-801. doi:10.4161/cbt.6.5.4007
    • (2007) Cancer Biol Ther , vol.6 , Issue.5 , pp. 795-801
    • Khabele, D.1    Son, D.S.2    Parl, A.K.3    Goldberg, G.L.4    Augenlicht, L.H.5    Mariadason, J.M.6
  • 37
    • 50949104573 scopus 로고    scopus 로고
    • Expression of class I histone deacetylases indicates poor prognosis in endometrioid subtypes of ovarian and endometrial carcinomas
    • Weichert W, Denkert C, Noske A, Darb-Esfahani S, Dietel M, Kalloger SE, et al. Expression of class I histone deacetylases indicates poor prognosis in endometrioid subtypes of ovarian and endometrial carcinomas. Neoplasia (2008) 10(9):1021-7.
    • (2008) Neoplasia , vol.10 , Issue.9 , pp. 1021-1027
    • Weichert, W.1    Denkert, C.2    Noske, A.3    Darb-Esfahani, S.4    Dietel, M.5    Kalloger, S.E.6
  • 38
    • 42149162792 scopus 로고    scopus 로고
    • HDACs and HDAC inhibitors in colon cancer
    • doi:10.4161/epi.3.1.5736
    • Mariadason JM. HDACs and HDAC inhibitors in colon cancer. Epigenetics (2008) 3(1):28-37. doi:10.4161/epi.3.1.5736
    • (2008) Epigenetics , vol.3 , Issue.1 , pp. 28-37
    • Mariadason, J.M.1
  • 39
    • 49249116407 scopus 로고    scopus 로고
    • A work in progress: the clinical development of histone deacetylase inhibitors
    • doi:10.4161/epi.3.3.6253
    • Marsoni S, Damia G, Camboni G. A work in progress: the clinical development of histone deacetylase inhibitors. Epigenetics (2008) 3(3):164-71. doi:10.4161/epi.3.3.6253
    • (2008) Epigenetics , vol.3 , Issue.3 , pp. 164-171
    • Marsoni, S.1    Damia, G.2    Camboni, G.3
  • 40
    • 34547864236 scopus 로고    scopus 로고
    • Histone deacetylase inhibitors: molecular mechanisms of action
    • doi:10.1038/sj.onc.1210620
    • Xu WS, Parmigiani RB, Marks PA. Histone deacetylase inhibitors: molecular mechanisms of action. Oncogene (2007) 26(37):5541-52. doi:10.1038/sj.onc.1210620
    • (2007) Oncogene , vol.26 , Issue.37 , pp. 5541-5552
    • Xu, W.S.1    Parmigiani, R.B.2    Marks, P.A.3
  • 41
    • 36148950997 scopus 로고    scopus 로고
    • FDA approval summary: vorinostat for treatment of advanced primary cutaneous T-cell lymphoma
    • doi:10.1634/theoncologist.12-10-1247
    • Mann BS, Johnson JR, Cohen MH, Justice R, Pazdur R. FDA approval summary: vorinostat for treatment of advanced primary cutaneous T-cell lymphoma. Oncologist (2007) 12(10):1247-52. doi:10.1634/theoncologist.12-10-1247
    • (2007) Oncologist , vol.12 , Issue.10 , pp. 1247-1252
    • Mann, B.S.1    Johnson, J.R.2    Cohen, M.H.3    Justice, R.4    Pazdur, R.5
  • 42
    • 34247860871 scopus 로고    scopus 로고
    • Vorinostat for treatment of cutaneous manifestations of advanced primary cutaneous T-cell lymphoma
    • doi:10.1158/1078-0432.CCR-06-2672
    • Mann BS, Johnson JR, He K, Sridhara R, Abraham S, Booth BP, et al. Vorinostat for treatment of cutaneous manifestations of advanced primary cutaneous T-cell lymphoma. Clin Cancer Res (2007) 13(8):2318-22. doi:10.1158/1078-0432.CCR-06-2672
    • (2007) Clin Cancer Res , vol.13 , Issue.8 , pp. 2318-2322
    • Mann, B.S.1    Johnson, J.R.2    He, K.3    Sridhara, R.4    Abraham, S.5    Booth, B.P.6
  • 43
    • 73949149251 scopus 로고    scopus 로고
    • Phase II multi-institutional trial of the histone deacetylase inhibitor romidepsin as monotherapy for patients with cutaneous T-cell lymphoma
    • doi:10.1200/JCO.2008.21.6150
    • Piekarz RL, Frye R, Turner M, Wright JJ, Allen SL, Kirschbaum MH, et al. Phase II multi-institutional trial of the histone deacetylase inhibitor romidepsin as monotherapy for patients with cutaneous T-cell lymphoma. J Clin Oncol (2009) 27(32):5410-7. doi:10.1200/JCO.2008.21.6150
    • (2009) J Clin Oncol , vol.27 , Issue.32 , pp. 5410-5417
    • Piekarz, R.L.1    Frye, R.2    Turner, M.3    Wright, J.J.4    Allen, S.L.5    Kirschbaum, M.H.6
  • 44
    • 79952977561 scopus 로고    scopus 로고
    • Phase 2 trial of romidepsin in patients with peripheral T-cell lymphoma
    • doi:10.1182/blood-2010-10-312603
    • Piekarz RL, Frye R, Prince HM, Kirschbaum MH, Zain J, Allen SL, et al. Phase 2 trial of romidepsin in patients with peripheral T-cell lymphoma. Blood (2011) 117(22):5827-34. doi:10.1182/blood-2010-10-312603
    • (2011) Blood , vol.117 , Issue.22 , pp. 5827-5834
    • Piekarz, R.L.1    Frye, R.2    Prince, H.M.3    Kirschbaum, M.H.4    Zain, J.5    Allen, S.L.6
  • 45
    • 84863419977 scopus 로고    scopus 로고
    • A phase II evaluation of belinostat and carboplatin in the treatment of recurrent or persistent platinum-resistant ovarian, fallopian tube, or primary peritoneal carcinoma: a Gynecologic Oncology Group study
    • doi:10.1016/j.ygyno.2012.02.019
    • Dizon DS, Blessing JA, Penson RT, Drake RD, Walker JL, Johnston CM, et al. A phase II evaluation of belinostat and carboplatin in the treatment of recurrent or persistent platinum-resistant ovarian, fallopian tube, or primary peritoneal carcinoma: a Gynecologic Oncology Group study. Gynecol Oncol (2012) 125(2):367-71. doi:10.1016/j.ygyno.2012.02.019
    • (2012) Gynecol Oncol , vol.125 , Issue.2 , pp. 367-371
    • Dizon, D.S.1    Blessing, J.A.2    Penson, R.T.3    Drake, R.D.4    Walker, J.L.5    Johnston, C.M.6
  • 46
    • 84863615068 scopus 로고    scopus 로고
    • Phase II activity of belinostat (PXD-101), carboplatin, and paclitaxel in women with previously treated ovarian cancer
    • doi:10.1097/IGC.0b013e31825736fd
    • Dizon DS, Damstrup L, Finkler NJ, Lassen U, Celano P, Glasspool R, et al. Phase II activity of belinostat (PXD-101), carboplatin, and paclitaxel in women with previously treated ovarian cancer. Int J Gynecol Cancer (2012) 22(6):979-86. doi:10.1097/IGC.0b013e31825736fd
    • (2012) Int J Gynecol Cancer , vol.22 , Issue.6 , pp. 979-986
    • Dizon, D.S.1    Damstrup, L.2    Finkler, N.J.3    Lassen, U.4    Celano, P.5    Glasspool, R.6
  • 47
    • 84876268623 scopus 로고    scopus 로고
    • Increased incidence of severe gastrointestinal events with first-line paclitaxel, carboplatin, and vorinostat chemotherapy for advanced-stage epithelial ovarian, primary peritoneal, and fallopian tube cancer
    • doi:10.1097/IGC.0b013e31828566f1
    • Mendivil AA, Micha JP, Brown JV III, Rettenmaier MA, Abaid LN, Lopez KL, et al. Increased incidence of severe gastrointestinal events with first-line paclitaxel, carboplatin, and vorinostat chemotherapy for advanced-stage epithelial ovarian, primary peritoneal, and fallopian tube cancer. Int J Gynecol Cancer (2013) 23(3):533-9. doi:10.1097/IGC.0b013e31828566f1
    • (2013) Int J Gynecol Cancer , vol.23 , Issue.3 , pp. 533-539
    • Mendivil, A.A.1    Micha, J.P.2    Brown J.V, I.I.I.3    Rettenmaier, M.A.4    Abaid, L.N.5    Lopez, K.L.6
  • 48
    • 43049091153 scopus 로고    scopus 로고
    • A phase II study of vorinostat in the treatment of persistent or recurrent epithelial ovarian or primary peritoneal carcinoma: a Gynecologic Oncology Group study
    • doi:10.1016/j.ygyno.2008.01.009
    • Modesitt SC, Sill M, Hoffman JS, Bender DP. A phase II study of vorinostat in the treatment of persistent or recurrent epithelial ovarian or primary peritoneal carcinoma: a Gynecologic Oncology Group study. Gynecol Oncol (2008) 109(2):182-6. doi:10.1016/j.ygyno.2008.01.009
    • (2008) Gynecol Oncol , vol.109 , Issue.2 , pp. 182-186
    • Modesitt, S.C.1    Sill, M.2    Hoffman, J.S.3    Bender, D.P.4
  • 50
    • 80052844657 scopus 로고    scopus 로고
    • The DNA damage mark pH2AX differentiates the cytotoxic effects of small molecule HDAC inhibitors in ovarian cancer cells
    • doi:10.4161/cbt.12.6.15956
    • Wilson AJ, Holson E, Wagner F, Zhang YL, Fass DM, Haggarty SJ, et al. The DNA damage mark pH2AX differentiates the cytotoxic effects of small molecule HDAC inhibitors in ovarian cancer cells. Cancer Biol Ther (2011) 12(6):484-93. doi:10.4161/cbt.12.6.15956
    • (2011) Cancer Biol Ther , vol.12 , Issue.6 , pp. 484-493
    • Wilson, A.J.1    Holson, E.2    Wagner, F.3    Zhang, Y.L.4    Fass, D.M.5    Haggarty, S.J.6
  • 51
    • 0036735385 scopus 로고    scopus 로고
    • FK228 (depsipeptide) as a natural prodrug that inhibits class I histone deacetylases
    • Furumai R, Matsuyama A, Kobashi N, Lee KH, Nishiyama M, Nakajima H, et al. FK228 (depsipeptide) as a natural prodrug that inhibits class I histone deacetylases. Cancer Res (2002) 62(17):4916-21.
    • (2002) Cancer Res , vol.62 , Issue.17 , pp. 4916-4921
    • Furumai, R.1    Matsuyama, A.2    Kobashi, N.3    Lee, K.H.4    Nishiyama, M.5    Nakajima, H.6
  • 52
    • 80055098514 scopus 로고    scopus 로고
    • Thailandepsins: bacterial products with potent histone deacetylase inhibitory activities and broad-spectrum antiproliferative activities
    • doi:10.1021/np200324x
    • Wang C, Henkes LM, Doughty LB, He M, Wang D, Meyer-Almes FJ, et al. Thailandepsins: bacterial products with potent histone deacetylase inhibitory activities and broad-spectrum antiproliferative activities. J Nat Prod (2011) 74(10):2031-8. doi:10.1021/np200324x
    • (2011) J Nat Prod , vol.74 , Issue.10 , pp. 2031-2038
    • Wang, C.1    Henkes, L.M.2    Doughty, L.B.3    He, M.4    Wang, D.5    Meyer-Almes, F.J.6
  • 53
    • 84859932807 scopus 로고    scopus 로고
    • Thailandepsins are new small molecule class I HDAC inhibitors with potent cytotoxic activity in ovarian cancer cells: a preclinical study of epigenetic ovarian cancer therapy
    • doi:10.1186/1757-2215-5-12
    • Wilson AJ, Cheng YQ, Khabele D. Thailandepsins are new small molecule class I HDAC inhibitors with potent cytotoxic activity in ovarian cancer cells: a preclinical study of epigenetic ovarian cancer therapy. J Ovarian Res (2012) 5(1):12. doi:10.1186/1757-2215-5-12
    • (2012) J Ovarian Res , vol.5 , Issue.1 , pp. 12
    • Wilson, A.J.1    Cheng, Y.Q.2    Khabele, D.3
  • 54
    • 1842578986 scopus 로고    scopus 로고
    • Molecular evolution of the histone deacetylase family: functional implications of phylogenetic analysis
    • doi:10.1016/j.jmb.2004.02.006
    • Gregoretti IV, Lee YM, Goodson HV. Molecular evolution of the histone deacetylase family: functional implications of phylogenetic analysis. J Mol Biol (2004) 338(1):17-31. doi:10.1016/j.jmb.2004.02.006
    • (2004) J Mol Biol , vol.338 , Issue.1 , pp. 17-31
    • Gregoretti, I.V.1    Lee, Y.M.2    Goodson, H.V.3
  • 55
    • 80051818192 scopus 로고    scopus 로고
    • Histone deacetylase inhibitor sodium butyrate enhances cellular radiosensitivity by inhibiting both DNA nonhomologous end joining and homologous recombination
    • doi:10.1016/j.dnarep.2011.07.003
    • Koprinarova M, Botev P, Russev G. Histone deacetylase inhibitor sodium butyrate enhances cellular radiosensitivity by inhibiting both DNA nonhomologous end joining and homologous recombination. DNA Repair (Amst) (2011) 10(9):970-7. doi:10.1016/j.dnarep.2011.07.003
    • (2011) DNA Repair (Amst) , vol.10 , Issue.9 , pp. 970-977
    • Koprinarova, M.1    Botev, P.2    Russev, G.3
  • 56
    • 37649015347 scopus 로고    scopus 로고
    • HDAC inhibitor PCI-24781 decreases RAD51 expression and inhibits homologous recombination
    • 19482-7, doi:10.1073/pnas.0707828104
    • Adimoolam S, Sirisawad M, Chen J, Thiemann P, Ford JM, Buggy JJ. HDAC inhibitor PCI-24781 decreases RAD51 expression and inhibits homologous recombination. Proc Natl Acad Sci U S A (2007) 104(49):19482-7. doi:10.1073/pnas.0707828104
    • (2007) Proc Natl Acad Sci U S A. , vol.104 , Issue.49
    • Adimoolam, S.1    Sirisawad, M.2    Chen, J.3    Thiemann, P.4    Ford, J.M.5    Buggy, J.J.6
  • 57
    • 84888132744 scopus 로고    scopus 로고
    • Inhibition of homologous recombination with vorinostat synergistically enhances ganciclovir cytotoxicity
    • doi:10.1016/j.dnarep.2013.10.008
    • Ladd B, Ackroyd JJ, Hicks JK, Canman CE, Flanagan SA, Shewach DS. Inhibition of homologous recombination with vorinostat synergistically enhances ganciclovir cytotoxicity. DNA Repair (Amst) (2013) 12(12):1114-21. doi:10.1016/j.dnarep.2013.10.008
    • (2013) DNA Repair (Amst) , vol.12 , Issue.12 , pp. 1114-1121
    • Ladd, B.1    Ackroyd, J.J.2    Hicks, J.K.3    Canman, C.E.4    Flanagan, S.A.5    Shewach, D.S.6
  • 58
    • 84901684118 scopus 로고    scopus 로고
    • Suberoylanilide hydroxamic acid (SAHA) enhances olaparib activity by targeting homologous recombination DNA repair in ovarian cancer.
    • doi:10.1016/j.ygyno.2014.03.007.
    • Konstantinopoulos PA, Wilson AJ, Saskowski J, Wass E, Khabele D. Suberoylanilide hydroxamic acid (SAHA) enhances olaparib activity by targeting homologous recombination DNA repair in ovarian cancer. Gynecol Oncol (2014). doi:10.1016/j.ygyno.2014.03.007
    • (2014) Gynecol Oncol
    • Konstantinopoulos, P.A.1    Wilson, A.J.2    Saskowski, J.3    Wass, E.4    Khabele, D.5
  • 59
    • 77952585540 scopus 로고    scopus 로고
    • Phase II trial of the histone deacetylase inhibitor belinostat in women with platinum resistant epithelial ovarian cancer and micropapillary (LMP) ovarian tumours
    • doi:10.1016/j.ejca.2010.02.047
    • Mackay HJ, Hirte H, Colgan T, Covens A, MacAlpine K, Grenci P, et al. Phase II trial of the histone deacetylase inhibitor belinostat in women with platinum resistant epithelial ovarian cancer and micropapillary (LMP) ovarian tumours. Eur J Cancer (2010) 46(9):1573-9. doi:10.1016/j.ejca.2010.02.047
    • (2010) Eur J Cancer , vol.46 , Issue.9 , pp. 1573-1579
    • Mackay, H.J.1    Hirte, H.2    Colgan, T.3    Covens, A.4    MacAlpine, K.5    Grenci, P.6
  • 60
    • 79952279235 scopus 로고    scopus 로고
    • The effects of the histone deacetylase inhibitor romidepsin (FK228) are enhanced by aspirin (ASA) in COX-1 positive ovarian cancer cells through augmentation of p21
    • doi:10.4161/cbt.9.11.11873
    • Son DS, Wilson AJ, Parl AK, Khabele D. The effects of the histone deacetylase inhibitor romidepsin (FK228) are enhanced by aspirin (ASA) in COX-1 positive ovarian cancer cells through augmentation of p21. Cancer Biol Ther (2010) 9(11):928-35. doi:10.4161/cbt.9.11.11873
    • (2010) Cancer Biol Ther , vol.9 , Issue.11 , pp. 928-935
    • Son, D.S.1    Wilson, A.J.2    Parl, A.K.3    Khabele, D.4
  • 61
    • 67349222049 scopus 로고    scopus 로고
    • Histone deacetylase inhibitors as anti-neoplastic agents
    • doi:10.1016/j.canlet.2009.03.013
    • Batty N, Malouf GG, Issa JP. Histone deacetylase inhibitors as anti-neoplastic agents. Cancer Lett (2009) 280(2):192-200. doi:10.1016/j.canlet.2009.03.013
    • (2009) Cancer Lett , vol.280 , Issue.2 , pp. 192-200
    • Batty, N.1    Malouf, G.G.2    Issa, J.P.3
  • 62
    • 66949123770 scopus 로고    scopus 로고
    • Epigenetic modulation of radiation response in human cancer cells with activated EGFR or HER-2 signaling: potential role of histone deacetylase 6
    • doi:10.1016/j.radonc.2009.03.008
    • Kim IA, No M, Lee JM, Shin JH, Oh JS, Choi EJ, et al. Epigenetic modulation of radiation response in human cancer cells with activated EGFR or HER-2 signaling: potential role of histone deacetylase 6. Radiother Oncol (2009) 92(1):125-32. doi:10.1016/j.radonc.2009.03.008
    • (2009) Radiother Oncol , vol.92 , Issue.1 , pp. 125-132
    • Kim, I.A.1    No, M.2    Lee, J.M.3    Shin, J.H.4    Oh, J.S.5    Choi, E.J.6
  • 63
    • 68149170764 scopus 로고    scopus 로고
    • Sensitization to gamma-irradiation-induced cell cycle arrest and apoptosis by the histone deacetylase inhibitor trichostatin A in non-small cell lung cancer (NSCLC) cells.
    • doi:10.4161/cbt.8.9.8143.
    • Zhang F, Zhang T, Teng ZH, Zhang R, Wang JB, Mei QB. Sensitization to gamma-irradiation-induced cell cycle arrest and apoptosis by the histone deacetylase inhibitor trichostatin A in non-small cell lung cancer (NSCLC) cells. Cancer Biol Ther (2009) 8:9. doi:10.4161/cbt.8.9.8143
    • (2009) Cancer Biol Ther , vol.8 , pp. 9
    • Zhang, F.1    Zhang, T.2    Teng, Z.H.3    Zhang, R.4    Wang, J.B.5    Mei, Q.B.6
  • 64
    • 34250805910 scopus 로고    scopus 로고
    • Histone deacetylase inhibitor NVP-LAQ824 sensitizes human nonsmall cell lung cancer to the cytotoxic effects of ionizing radiation
    • doi:10.1097/CAD.0b013e3280b10d57
    • Cuneo KC, Fu A, Osusky K, Huamani J, Hallahan DE, Geng L. Histone deacetylase inhibitor NVP-LAQ824 sensitizes human nonsmall cell lung cancer to the cytotoxic effects of ionizing radiation. Anticancer Drugs (2007) 18(7):793-800. doi:10.1097/CAD.0b013e3280b10d57
    • (2007) Anticancer Drugs , vol.18 , Issue.7 , pp. 793-800
    • Cuneo, K.C.1    Fu, A.2    Osusky, K.3    Huamani, J.4    Hallahan, D.E.5    Geng, L.6
  • 65
    • 33845741562 scopus 로고    scopus 로고
    • Histone deacetylase (HDAC) inhibitor LBH589 increases duration of gamma-H2AX foci and confines HDAC4 to the cytoplasm in irradiated non-small cell lung cancer
    • 11298-304, doi:10.1158/0008-5472.CAN-06-0049
    • Geng L, Cuneo KC, Fu A, Tu T, Atadja PW, Hallahan DE. Histone deacetylase (HDAC) inhibitor LBH589 increases duration of gamma-H2AX foci and confines HDAC4 to the cytoplasm in irradiated non-small cell lung cancer. Cancer Res (2006) 66(23):11298-304. doi:10.1158/0008-5472.CAN-06-0049
    • (2006) Cancer Res , vol.66 , Issue.23
    • Geng, L.1    Cuneo, K.C.2    Fu, A.3    Tu, T.4    Atadja, P.W.5    Hallahan, D.E.6
  • 66
    • 66449100610 scopus 로고    scopus 로고
    • HDAC2 regulates chromatin plasticity and enhances DNA vulnerability
    • doi:10.1158/1535-7163.MCT-08-0985
    • Marchion DC, Bicaku E, Turner JG, Schmitt ML, Morelli DR, Munster PN. HDAC2 regulates chromatin plasticity and enhances DNA vulnerability. Mol Cancer Ther (2009) 8(4):794-801. doi:10.1158/1535-7163.MCT-08-0985
    • (2009) Mol Cancer Ther , vol.8 , Issue.4 , pp. 794-801
    • Marchion, D.C.1    Bicaku, E.2    Turner, J.G.3    Schmitt, M.L.4    Morelli, D.R.5    Munster, P.N.6
  • 67
    • 38949146354 scopus 로고    scopus 로고
    • Histone deacetylase inhibitors enhance the chemosensitivity of tumor cells with cross-resistance to a wide range of DNA-damaging drugs
    • doi:10.1111/j.1349-7006.2007.00669.x
    • Ozaki K, Kishikawa F, Tanaka M, Sakamoto T, Tanimura S, Kohno M. Histone deacetylase inhibitors enhance the chemosensitivity of tumor cells with cross-resistance to a wide range of DNA-damaging drugs. Cancer Sci (2008) 99(2):376-84. doi:10.1111/j.1349-7006.2007.00669.x
    • (2008) Cancer Sci , vol.99 , Issue.2 , pp. 376-384
    • Ozaki, K.1    Kishikawa, F.2    Tanaka, M.3    Sakamoto, T.4    Tanimura, S.5    Kohno, M.6
  • 68
    • 0038544192 scopus 로고    scopus 로고
    • Histone deacetylase in carcinogenesis and its inhibitors as anti-cancer agents
    • doi:10.5483/BMBRep.2003.36.1.110
    • Kim DH, Kim M, Kwon HJ. Histone deacetylase in carcinogenesis and its inhibitors as anti-cancer agents. J Biochem Mol Biol (2003) 36(1):110-9. doi:10.5483/BMBRep.2003.36.1.110
    • (2003) J Biochem Mol Biol , vol.36 , Issue.1 , pp. 110-119
    • Kim, D.H.1    Kim, M.2    Kwon, H.J.3
  • 69
    • 2442713811 scopus 로고    scopus 로고
    • Cisplatin-induced post-translational modification of histones H3 and H4
    • 20622-5, doi:10.1074/jbc.M402547200
    • Wang D, Lippard SJ. Cisplatin-induced post-translational modification of histones H3 and H4. J Biol Chem (2004) 279(20):20622-5. doi:10.1074/jbc.M402547200
    • (2004) J Biol Chem , vol.279 , Issue.20
    • Wang, D.1    Lippard, S.J.2
  • 70
    • 0035224059 scopus 로고    scopus 로고
    • Cisplatin: from DNA damage to cancer chemotherapy.
    • doi:10.1016/S0079-6603(01)67026-0.
    • Cohen SM, Lippard SJ. Cisplatin: from DNA damage to cancer chemotherapy. Prog Nucleic Acid Res Mol Biol (2001) 67:93-130. doi:10.1016/S0079-6603(01)67026-0
    • (2001) Prog Nucleic Acid Res Mol Biol , vol.67 , pp. 93-130
    • Cohen, S.M.1    Lippard, S.J.2
  • 71
    • 61749093379 scopus 로고    scopus 로고
    • Combined inhibition of DNA methylation and histone acetylation enhances gene re-expression and drug sensitivity in vivo
    • doi:10.1038/sj.bjc.6604932
    • Steele N, Finn P, Brown R, Plumb JA. Combined inhibition of DNA methylation and histone acetylation enhances gene re-expression and drug sensitivity in vivo. Br J Cancer (2009) 100(5):758-63. doi:10.1038/sj.bjc.6604932
    • (2009) Br J Cancer , vol.100 , Issue.5 , pp. 758-763
    • Steele, N.1    Finn, P.2    Brown, R.3    Plumb, J.A.4
  • 72
    • 41949120723 scopus 로고    scopus 로고
    • Liver-specific deletion of histone deacetylase 3 disrupts metabolic transcriptional networks
    • doi:10.1038/emboj.2008.51
    • Knutson SK, Chyla BJ, Amann JM, Bhaskara S, Huppert SS, Hiebert SW. Liver-specific deletion of histone deacetylase 3 disrupts metabolic transcriptional networks. EMBO J (2008) 27(7):1017-28. doi:10.1038/emboj.2008.51
    • (2008) EMBO J. , vol.27 , Issue.7 , pp. 1017-1028
    • Knutson, S.K.1    Chyla, B.J.2    Amann, J.M.3    Bhaskara, S.4    Huppert, S.S.5    Hiebert, S.W.6
  • 73
    • 84868517231 scopus 로고    scopus 로고
    • Romidepsin (FK228) combined with cisplatin stimulates DNA damage-induced cell death in ovarian cancer
    • doi:10.1016/j.ygyno.2012.09.016
    • Wilson AJ, Lalani AS, Wass E, Saskowski J, Khabele D. Romidepsin (FK228) combined with cisplatin stimulates DNA damage-induced cell death in ovarian cancer. Gynecol Oncol (2012) 127(3):579-86. doi:10.1016/j.ygyno.2012.09.016
    • (2012) Gynecol Oncol , vol.127 , Issue.3 , pp. 579-586
    • Wilson, A.J.1    Lalani, A.S.2    Wass, E.3    Saskowski, J.4    Khabele, D.5
  • 74
    • 54349100901 scopus 로고    scopus 로고
    • HDAC4 promotes growth of colon cancer cells via repression of p21
    • doi:10.1091/mbc.E08-02-0139
    • Wilson AJ, Byun DS, Nasser S, Murray LB, Ayyanar K, Arango D, et al. HDAC4 promotes growth of colon cancer cells via repression of p21. Mol Biol Cell (2008) 19(10):4062-75. doi:10.1091/mbc.E08-02-0139
    • (2008) Mol Biol Cell , vol.19 , Issue.10 , pp. 4062-4075
    • Wilson, A.J.1    Byun, D.S.2    Nasser, S.3    Murray, L.B.4    Ayyanar, K.5    Arango, D.6
  • 75
    • 28144434441 scopus 로고    scopus 로고
    • Histone deacetylase 3 represses p15(INK4b) and p21(WAF1/cip1) transcription by interacting with Sp1
    • doi:10.1016/j.bbrc.2005.11.010
    • Huang W, Tan D, Wang X, Han S, Tan J, Zhao Y, et al. Histone deacetylase 3 represses p15(INK4b) and p21(WAF1/cip1) transcription by interacting with Sp1. Biochem Biophys Res Commun (2006) 339(1):165-71. doi:10.1016/j.bbrc.2005.11.010
    • (2006) Biochem Biophys Res Commun , vol.339 , Issue.1 , pp. 165-171
    • Huang, W.1    Tan, D.2    Wang, X.3    Han, S.4    Tan, J.5    Zhao, Y.6
  • 76
    • 0031723957 scopus 로고    scopus 로고
    • Aberrant recruitment of the nuclear receptor corepressor-histone deacetylase complex by the acute myeloid leukemia fusion partner ETO
    • Gelmetti V, Zhang J, Fanelli M, Minucci S, Pelicci PG, Lazar MA. Aberrant recruitment of the nuclear receptor corepressor-histone deacetylase complex by the acute myeloid leukemia fusion partner ETO. Mol Cell Biol (1998) 18(12):7185-91.
    • (1998) Mol Cell Biol , vol.18 , Issue.12 , pp. 7185-7191
    • Gelmetti, V.1    Zhang, J.2    Fanelli, M.3    Minucci, S.4    Pelicci, P.G.5    Lazar, M.A.6
  • 77
    • 17144458786 scopus 로고    scopus 로고
    • Fusion proteins of the retinoic acid receptor-alpha recruit histone deacetylase in promyelocytic leukaemia
    • doi:10.1038/35901
    • Grignani F, De Matteis S, Nervi C, Tomassoni L, Gelmetti V, Cioce M, et al. Fusion proteins of the retinoic acid receptor-alpha recruit histone deacetylase in promyelocytic leukaemia. Nature (1998) 391(6669):815-8. doi:10.1038/35901
    • (1998) Nature , vol.391 , Issue.6669 , pp. 815-818
    • Grignani, F.1    De Matteis, S.2    Nervi, C.3    Tomassoni, L.4    Gelmetti, V.5    Cioce, M.6
  • 78
    • 33744522801 scopus 로고    scopus 로고
    • Histone deacetylase 3 (HDAC3) is recruited to target promoters by PML-RARalpha as a component of the N-CoR co-repressor complex to repress transcription in vivo
    • doi:10.1016/j.bbrc.2006.05.047
    • Atsumi A, Tomita A, Kiyoi H, Naoe T. Histone deacetylase 3 (HDAC3) is recruited to target promoters by PML-RARalpha as a component of the N-CoR co-repressor complex to repress transcription in vivo. Biochem Biophys Res Commun (2006) 345(4):1471-80. doi:10.1016/j.bbrc.2006.05.047
    • (2006) Biochem Biophys Res Commun , vol.345 , Issue.4 , pp. 1471-1480
    • Atsumi, A.1    Tomita, A.2    Kiyoi, H.3    Naoe, T.4
  • 79
    • 84879621358 scopus 로고    scopus 로고
    • HDAC3 is essential for DNA replication in hematopoietic progenitor cells
    • doi:10.1172/JCI60806
    • Summers AR, Fischer MA, Stengel KR, Zhao Y, Kaiser JF, Wells CE, et al. HDAC3 is essential for DNA replication in hematopoietic progenitor cells. J Clin Invest (2013) 123(7):3112-23. doi:10.1172/JCI60806
    • (2013) J Clin Invest , vol.123 , Issue.7 , pp. 3112-3123
    • Summers, A.R.1    Fischer, M.A.2    Stengel, K.R.3    Zhao, Y.4    Kaiser, J.F.5    Wells, C.E.6
  • 80
    • 84880655164 scopus 로고    scopus 로고
    • Inhibition of histone deacetylase 3 causes replication stress in cutaneous T cell lymphoma
    • doi:10.1371/journal.pone.0068915
    • Wells CE, Bhaskara S, Stengel KR, Zhao Y, Sirbu B, Chagot B, et al. Inhibition of histone deacetylase 3 causes replication stress in cutaneous T cell lymphoma. PLoS One (2013) 8(7):e68915. doi:10.1371/journal.pone.0068915
    • (2013) PLoS One , vol.8 , Issue.7
    • Wells, C.E.1    Bhaskara, S.2    Stengel, K.R.3    Zhao, Y.4    Sirbu, B.5    Chagot, B.6
  • 81
    • 84875978060 scopus 로고    scopus 로고
    • HDAC inhibitors induce tumor-cell-selective pro-apoptotic transcriptional responses.
    • doi:10.1038/cddis.2013.9.
    • Bolden JE, Shi W, Jankowski K, Kan CY, Cluse L, Martin BP, et al. HDAC inhibitors induce tumor-cell-selective pro-apoptotic transcriptional responses. Cell Death Dis (2013) 4:e519. doi:10.1038/cddis.2013.9
    • (2013) Cell Death Dis. , vol.4
    • Bolden, J.E.1    Shi, W.2    Jankowski, K.3    Kan, C.Y.4    Cluse, L.5    Martin, B.P.6
  • 82
    • 84856381695 scopus 로고    scopus 로고
    • Distinct subpopulations of epithelial ovarian cancer cells can differentially induce macrophages and T regulatory cells toward a pro-tumor phenotype
    • doi:10.1111/j.1600-0897.2011.01068.x
    • Alvero AB, Montagna MK, Craveiro V, Liu L, Mor G. Distinct subpopulations of epithelial ovarian cancer cells can differentially induce macrophages and T regulatory cells toward a pro-tumor phenotype. Am J Reprod Immunol (2012) 67(3):256-65. doi:10.1111/j.1600-0897.2011.01068.x
    • (2012) Am J Reprod Immunol , vol.67 , Issue.3 , pp. 256-265
    • Alvero, A.B.1    Montagna, M.K.2    Craveiro, V.3    Liu, L.4    Mor, G.5
  • 83
    • 79959549102 scopus 로고    scopus 로고
    • Tumor-infiltrating programmed death receptor-1+ dendritic cells mediate immune suppression in ovarian cancer
    • doi:10.4049/jimmunol.1100274
    • Krempski J, Karyampudi L, Behrens MD, Erskine CL, Hartmann L, Dong H, et al. Tumor-infiltrating programmed death receptor-1+ dendritic cells mediate immune suppression in ovarian cancer. J Immunol (2011) 186(12):6905-13. doi:10.4049/jimmunol.1100274
    • (2011) J Immunol , vol.186 , Issue.12 , pp. 6905-6913
    • Krempski, J.1    Karyampudi, L.2    Behrens, M.D.3    Erskine, C.L.4    Hartmann, L.5    Dong, H.6
  • 84
    • 45149134605 scopus 로고    scopus 로고
    • "Re-educating" tumor-associated macrophages by targeting NF-kappaB
    • doi:10.1084/jem.20080108
    • Hagemann T, Lawrence T, McNeish I, Charles KA, Kulbe H, Thompson RG, et al. "Re-educating" tumor-associated macrophages by targeting NF-kappaB. J Exp Med (2008) 205(6):1261-8. doi:10.1084/jem.20080108
    • (2008) J Exp Med , vol.205 , Issue.6 , pp. 1261-1268
    • Hagemann, T.1    Lawrence, T.2    McNeish, I.3    Charles, K.A.4    Kulbe, H.5    Thompson, R.G.6
  • 85
    • 34447319652 scopus 로고    scopus 로고
    • Ovarian cancer cell-derived migration inhibitory factor enhances tumor growth, progression, and angiogenesis
    • doi:10.1158/1535-7163.MCT-07-0118
    • Hagemann T, Robinson SC, Thompson RG, Charles K, Kulbe H, Balkwill FR. Ovarian cancer cell-derived migration inhibitory factor enhances tumor growth, progression, and angiogenesis. Mol Cancer Ther (2007) 6(7):1993-2002. doi:10.1158/1535-7163.MCT-07-0118
    • (2007) Mol Cancer Ther , vol.6 , Issue.7 , pp. 1993-2002
    • Hagemann, T.1    Robinson, S.C.2    Thompson, R.G.3    Charles, K.4    Kulbe, H.5    Balkwill, F.R.6
  • 86
    • 34250821793 scopus 로고    scopus 로고
    • Macrophages mediate inflammation-enhanced metastasis of ovarian tumors in mice
    • doi:10.1158/0008-5472.CAN-06-4375
    • Robinson-Smith TM, Isaacsohn I, Mercer CA, Zhou M, Van Rooijen N, Husseinzadeh N, et al. Macrophages mediate inflammation-enhanced metastasis of ovarian tumors in mice. Cancer Res (2007) 67(12):5708-16. doi:10.1158/0008-5472.CAN-06-4375
    • (2007) Cancer Res , vol.67 , Issue.12 , pp. 5708-5716
    • Robinson-Smith, T.M.1    Isaacsohn, I.2    Mercer, C.A.3    Zhou, M.4    Van Rooijen, N.5    Husseinzadeh, N.6
  • 87
    • 84872445265 scopus 로고    scopus 로고
    • Neutralizing tumor-promoting chronic inflammation: a magic bullet?
    • doi:10.1126/science.1232227.
    • Coussens LM, Zitvogel L, Palucka AK. Neutralizing tumor-promoting chronic inflammation: a magic bullet? Science (2013) 339(6117):286-91. doi:10.1126/science.1232227
    • (2013) Science , vol.339 , Issue.6117 , pp. 286-291
    • Coussens, L.M.1    Zitvogel, L.2    Palucka, A.K.3
  • 88
    • 23244451640 scopus 로고    scopus 로고
    • Macrophages induce invasiveness of epithelial cancer cells via NF-kappa B and JNK
    • doi:10.4049/jimmunol.175.2.1197
    • Hagemann T, Wilson J, Kulbe H, Li NF, Leinster DA, Charles K, et al. Macrophages induce invasiveness of epithelial cancer cells via NF-kappa B and JNK. J Immunol (2005) 175(2):1197-205. doi:10.4049/jimmunol.175.2.1197
    • (2005) J Immunol , vol.175 , Issue.2 , pp. 1197-1205
    • Hagemann, T.1    Wilson, J.2    Kulbe, H.3    Li, N.F.4    Leinster, D.A.5    Charles, K.6
  • 89
    • 84855393623 scopus 로고    scopus 로고
    • A dynamic inflammatory cytokine network in the human ovarian cancer microenvironment
    • doi:10.1158/0008-5472.CAN-11-2178
    • Kulbe H, Chakravarty P, Leinster DA, Charles KA, Kwong J, Thompson RG, et al. A dynamic inflammatory cytokine network in the human ovarian cancer microenvironment. Cancer Res (2012) 72(1):66-75. doi:10.1158/0008-5472.CAN-11-2178
    • (2012) Cancer Res , vol.72 , Issue.1 , pp. 66-75
    • Kulbe, H.1    Chakravarty, P.2    Leinster, D.A.3    Charles, K.A.4    Kwong, J.5    Thompson, R.G.6
  • 90
    • 33645760428 scopus 로고    scopus 로고
    • Ovarian cancer cells polarize macrophages toward a tumor-associated phenotype
    • doi:10.4049/jimmunol.176.8.5023
    • Hagemann T, Wilson J, Burke F, Kulbe H, Li NF, Pluddemann A, et al. Ovarian cancer cells polarize macrophages toward a tumor-associated phenotype. J Immunol (2006) 176(8):5023-32. doi:10.4049/jimmunol.176.8.5023
    • (2006) J Immunol , vol.176 , Issue.8 , pp. 5023-5032
    • Hagemann, T.1    Wilson, J.2    Burke, F.3    Kulbe, H.4    Li, N.F.5    Pluddemann, A.6
  • 91
    • 82955247088 scopus 로고    scopus 로고
    • Histone deacetylase 3 is an epigenomic brake in macrophage alternative activation
    • doi:10.1101/gad.175950.111
    • Mullican SE, Gaddis CA, Alenghat T, Nair MG, Giacomin PR, Everett LJ, et al. Histone deacetylase 3 is an epigenomic brake in macrophage alternative activation. Genes Dev (2011) 25(23):2480-8. doi:10.1101/gad.175950.111
    • (2011) Genes Dev , vol.25 , Issue.23 , pp. 2480-2488
    • Mullican, S.E.1    Gaddis, C.A.2    Alenghat, T.3    Nair, M.G.4    Giacomin, P.R.5    Everett, L.J.6
  • 92
    • 84867652835 scopus 로고    scopus 로고
    • Requirement for the histone deacetylase Hdac3 for the inflammatory gene expression program in macrophages
    • doi:10.1073/pnas.1121131109
    • Chen X, Barozzi I, Termanini A, Prosperini E, Recchiuti A, Dalli J, et al. Requirement for the histone deacetylase Hdac3 for the inflammatory gene expression program in macrophages. Proc Natl Acad Sci U S A (2012) 109(42):E2865-74. doi:10.1073/pnas.1121131109
    • (2012) Proc Natl Acad Sci U S A. , vol.109 , Issue.42
    • Chen, X.1    Barozzi, I.2    Termanini, A.3    Prosperini, E.4    Recchiuti, A.5    Dalli, J.6
  • 93
    • 0035979737 scopus 로고    scopus 로고
    • Duration of nuclear NF-kappaB action regulated by reversible acetylation
    • doi:10.1126/science.1062374
    • Chen L, Fischle W, Verdin E, Greene WC. Duration of nuclear NF-kappaB action regulated by reversible acetylation. Science (2001) 293(5535):1653-7. doi:10.1126/science.1062374
    • (2001) Science , vol.293 , Issue.5535 , pp. 1653-1657
    • Chen, L.1    Fischle, W.2    Verdin, E.3    Greene, W.C.4
  • 94
    • 84871745733 scopus 로고    scopus 로고
    • The coactivator role of histone deacetylase 3 in IL-1-signaling involves deacetylation of p65 NF-kappaB
    • doi:10.1093/nar/gks916
    • Ziesche E, Kettner-Buhrow D, Weber A, Wittwer T, Jurida L, Soelch J, et al. The coactivator role of histone deacetylase 3 in IL-1-signaling involves deacetylation of p65 NF-kappaB. Nucleic Acids Res (2013) 41(1):90-109. doi:10.1093/nar/gks916
    • (2013) Nucleic Acids Res , vol.41 , Issue.1 , pp. 90-109
    • Ziesche, E.1    Kettner-Buhrow, D.2    Weber, A.3    Wittwer, T.4    Jurida, L.5    Soelch, J.6
  • 95
    • 84865547244 scopus 로고    scopus 로고
    • Nuclear factor-kappaB binding motifs specify toll-like receptor-induced gene repression through an inducible repressosome
    • 14140-5, doi:10.1073/pnas.1119842109
    • Yan Q, Carmody RJ, Qu Z, Ruan Q, Jager J, Mullican SE, et al. Nuclear factor-kappaB binding motifs specify toll-like receptor-induced gene repression through an inducible repressosome. Proc Natl Acad Sci U S A (2012) 109(35):14140-5. doi:10.1073/pnas.1119842109
    • (2012) Proc Natl Acad Sci U S A. , vol.109 , Issue.35
    • Yan, Q.1    Carmody, R.J.2    Qu, Z.3    Ruan, Q.4    Jager, J.5    Mullican, S.E.6
  • 96
    • 84871209715 scopus 로고    scopus 로고
    • Targeting class I histone deacetylases in cancer therapy
    • doi:10.1517/14728222.2013.729042
    • Delcuve GP, Khan DH, Davie JR. Targeting class I histone deacetylases in cancer therapy. Expert Opin Ther Targets (2013) 17(1):29-41. doi:10.1517/14728222.2013.729042
    • (2013) Expert Opin Ther Targets , vol.17 , Issue.1 , pp. 29-41
    • Delcuve, G.P.1    Khan, D.H.2    Davie, J.R.3
  • 97
    • 84885802146 scopus 로고    scopus 로고
    • Enhanced cytotoxic effects of combined valproic acid and the aurora kinase inhibitor VE465 on gynecologic cancer cells.
    • doi:10.3389/fonc.2013.00058.
    • Li Y, Liu T, Ivan C, Huang J, Shen DY, Kavanagh JJ, et al. Enhanced cytotoxic effects of combined valproic acid and the aurora kinase inhibitor VE465 on gynecologic cancer cells. Front Oncol (2013) 3:58. doi:10.3389/fonc.2013.00058
    • (2013) Front Oncol , vol.3 , pp. 58
    • Li, Y.1    Liu, T.2    Ivan, C.3    Huang, J.4    Shen, D.Y.5    Kavanagh, J.J.6
  • 98
    • 80053162877 scopus 로고    scopus 로고
    • Drug targeting to monocytes and macrophages using esterase-sensitive chemical motifs
    • doi:10.1124/jpet.111.183640
    • Needham LA, Davidson AH, Bawden LJ, Belfield A, Bone EA, Brotherton DH, et al. Drug targeting to monocytes and macrophages using esterase-sensitive chemical motifs. J Pharmacol Exp Ther (2011) 339(1):132-42. doi:10.1124/jpet.111.183640
    • (2011) J Pharmacol Exp Ther , vol.339 , Issue.1 , pp. 132-142
    • Needham, L.A.1    Davidson, A.H.2    Bawden, L.J.3    Belfield, A.4    Bone, E.A.5    Brotherton, D.H.6
  • 99
    • 84879341188 scopus 로고    scopus 로고
    • A dual role for Hdac1: oncosuppressor in tumorigenesis, oncogene in tumor maintenance
    • doi:10.1182/blood-2012-10-461988
    • Santoro F, Botrugno OA, Dal Zuffo R, Pallavicini I, Matthews GM, Cluse L, et al. A dual role for Hdac1: oncosuppressor in tumorigenesis, oncogene in tumor maintenance. Blood (2013) 121(17):3459-68. doi:10.1182/blood-2012-10-461988
    • (2013) Blood , vol.121 , Issue.17 , pp. 3459-3468
    • Santoro, F.1    Botrugno, O.A.2    Dal Zuffo, R.3    Pallavicini, I.4    Matthews, G.M.5    Cluse, L.6
  • 100
    • 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
    • doi:10.1182/blood-2012-08-450916
    • Heideman MR, Wilting RH, Yanover E, Velds A, de Jong J, Kerkhoven RM, et al. Dosage-dependent tumor suppression by histone deacetylases 1 and 2 through regulation of c-Myc collaborating genes and p53 function. Blood (2013) 121(11):2038-50. doi:10.1182/blood-2012-08-450916
    • (2013) Blood , vol.121 , Issue.11 , pp. 2038-2050
    • Heideman, M.R.1    Wilting, R.H.2    Yanover, E.3    Velds, A.4    de Jong, J.5    Kerkhoven, R.M.6


* 이 정보는 Elsevier사의 SCOPUS DB에서 KISTI가 분석하여 추출한 것입니다.