메뉴 건너뛰기




Volumn 26, Issue 5, 2012, Pages 959-980

Role of Additional Novel Therapies in Myeloproliferative Neoplasms

Author keywords

HDAC inhibitor; Hsp90 inhibitors; JAK2 V617F; MEK inhibitor; Myeloproliferative neoplasms; PI3K AKT inhibitor

Indexed keywords

1 (1 CYANO 1 METHYLETHYL) 3 METHYL 8 (3 QUINOLINYL)IMIDAZO[4,5 C]QUINOLIN 2(1H,3H) ONE; 5 (2,4 DIHYDROXY 5 ISOPROPYLPHENYL) 4 (4 MORPHOLINOMETHYLPHENYL) 3 ISOXAZOLECARBOXYLIC ACID ETHYLAMIDE; 5 AZA 2' DEOXYCYTIDINE; 8 [4 (1 AMINOCYCLOBUTYL)PHENYL] 9 PHENYL 1,2,4 TRIAZOLO[3,4 F][1,6]NAPHTHYRIDIN 3(2H) ONE; AZD 1208; BC 2059; BMS 911543; EVEROLIMUS; GIVINOSTAT; HYDROXYUREA; JANUS KINASE 2; JANUS KINASE INHIBITOR; LESTAURTINIB; MOMELOTINIB; N TERT BUTYL 3 [5 METHYL 2 [4 (4 METHYL 1 PIPERAZINYL)PHENYLAMINO] 4 PYRIMIDINYLAMINO]BENZENESULFONAMIDE; N TERT BUTYL 3 [5 METHYL 2 [4 [2 (1 PYRROLIDINYL)ETHOXY]PHENYLAMINO] 4 PYRIMIDINYLAMINO]BENZENESULFONAMIDE; PANOBINOSTAT; PF 04449913; PU H71; ROMIDEPSIN; RUXOLITINIB; SELUMETINIB; SGI 1776; STAT3 PROTEIN; STAT5 PROTEIN; SUPPRESSOR OF CYTOKINE SIGNALING; TANESPIMYCIN; TRICHOSTATIN A; UNCLASSIFIED DRUG; UNINDEXED DRUG; VALINE; VORINOSTAT;

EID: 84866633871     PISSN: 08898588     EISSN: 15581977     Source Type: Journal    
DOI: 10.1016/j.hoc.2012.07.001     Document Type: Review
Times cited : (5)

References (137)
  • 1
    • 20144363192 scopus 로고    scopus 로고
    • Acquired mutation of the tyrosine kinase JAK2 in human myeloproliferative disorders
    • Baxter E.J., Scott L.M., Campbell P.J., et al. Acquired mutation of the tyrosine kinase JAK2 in human myeloproliferative disorders. Lancet 2005, 365:1054-1061.
    • (2005) Lancet , vol.365 , pp. 1054-1061
    • Baxter, E.J.1    Scott, L.M.2    Campbell, P.J.3
  • 2
    • 17844383458 scopus 로고    scopus 로고
    • A unique clonal JAK2 mutation leading to constitutive signalling causes polycythaemia vera
    • James C., Ugo V., Le Couedic J.P., et al. A unique clonal JAK2 mutation leading to constitutive signalling causes polycythaemia vera. Nature 2005, 434:1144-1148.
    • (2005) Nature , vol.434 , pp. 1144-1148
    • James, C.1    Ugo, V.2    Le Couedic, J.P.3
  • 3
    • 20244369569 scopus 로고    scopus 로고
    • Activating mutation in the tyrosine kinase JAK2 in polycythemia vera, essential thrombocythemia, and myeloid metaplasia with myelofibrosis
    • Levine R.L., Wadleigh M., Cools J., et al. Activating mutation in the tyrosine kinase JAK2 in polycythemia vera, essential thrombocythemia, and myeloid metaplasia with myelofibrosis. Cancer Cell 2005, 7:387-397.
    • (2005) Cancer Cell , vol.7 , pp. 387-397
    • Levine, R.L.1    Wadleigh, M.2    Cools, J.3
  • 4
    • 55249095625 scopus 로고    scopus 로고
    • Myeloproliferative disorders
    • Levine R.L., Gilliland D.G. Myeloproliferative disorders. Blood 2008, 112:2190-2198.
    • (2008) Blood , vol.112 , pp. 2190-2198
    • Levine, R.L.1    Gilliland, D.G.2
  • 5
    • 34548240698 scopus 로고    scopus 로고
    • Role of JAK2 in the pathogenesis and therapy of myeloproliferative disorders
    • Levine R.L., Pardanani A., Tefferi A., et al. Role of JAK2 in the pathogenesis and therapy of myeloproliferative disorders. Nat Rev Cancer 2007, 7:673-683.
    • (2007) Nat Rev Cancer , vol.7 , pp. 673-683
    • Levine, R.L.1    Pardanani, A.2    Tefferi, A.3
  • 6
    • 35348850734 scopus 로고    scopus 로고
    • Hematopoietic cytokine receptor signaling
    • Baker S.J., Rane S.G., Reddy E.P. Hematopoietic cytokine receptor signaling. Oncogene 2007, 26:6724-6737.
    • (2007) Oncogene , vol.26 , pp. 6724-6737
    • Baker, S.J.1    Rane, S.G.2    Reddy, E.P.3
  • 7
    • 77954613886 scopus 로고    scopus 로고
    • JAK2 V617F uses distinct signalling pathways to induce cell proliferation and neutrophil activation
    • Oku S., Takenaka K., Kuriyama T., et al. JAK2 V617F uses distinct signalling pathways to induce cell proliferation and neutrophil activation. Br J Haematol 2010, 150:334-344.
    • (2010) Br J Haematol , vol.150 , pp. 334-344
    • Oku, S.1    Takenaka, K.2    Kuriyama, T.3
  • 8
    • 80052492285 scopus 로고    scopus 로고
    • The pseudokinase domain of JAK2 is a dual-specificity protein kinase that negatively regulates cytokine signaling
    • Ungureanu D., Wu J., Pekkala T., et al. The pseudokinase domain of JAK2 is a dual-specificity protein kinase that negatively regulates cytokine signaling. Nat Struct Mol Biol 2011, 18:971-976.
    • (2011) Nat Struct Mol Biol , vol.18 , pp. 971-976
    • Ungureanu, D.1    Wu, J.2    Pekkala, T.3
  • 9
    • 79955136973 scopus 로고    scopus 로고
    • Analysis of Jak2 catalytic function by peptide microarrays: the role of the JH2 domain and V617F mutation
    • Sanz A., Ungureanu D., Pekkala T., et al. Analysis of Jak2 catalytic function by peptide microarrays: the role of the JH2 domain and V617F mutation. PLoS One 2011, 6:e18522.
    • (2011) PLoS One , vol.6
    • Sanz, A.1    Ungureanu, D.2    Pekkala, T.3
  • 10
    • 77956589191 scopus 로고    scopus 로고
    • Stat5a serine 725 and 779 phosphorylation is a prerequisite for hematopoietic transformation
    • Friedbichler K., Kerenyi M.A., Kovacic B., et al. Stat5a serine 725 and 779 phosphorylation is a prerequisite for hematopoietic transformation. Blood 2010, 116:1548-1558.
    • (2010) Blood , vol.116 , pp. 1548-1558
    • Friedbichler, K.1    Kerenyi, M.A.2    Kovacic, B.3
  • 11
    • 70349975711 scopus 로고    scopus 로고
    • JAK2 phosphorylates histone H3Y41 and excludes HP1alpha from chromatin
    • Dawson M.A., Bannister A.J., Gottgens B., et al. JAK2 phosphorylates histone H3Y41 and excludes HP1alpha from chromatin. Nature 2009, 461:819-822.
    • (2009) Nature , vol.461 , pp. 819-822
    • Dawson, M.A.1    Bannister, A.J.2    Gottgens, B.3
  • 12
    • 79951476020 scopus 로고    scopus 로고
    • Janus kinase 2: an epigenetic 'writer' that activates leukemogenic genes
    • He J., Zhang Y. Janus kinase 2: an epigenetic 'writer' that activates leukemogenic genes. J Mol Cell Biol 2010, 2:231-233.
    • (2010) J Mol Cell Biol , vol.2 , pp. 231-233
    • He, J.1    Zhang, Y.2
  • 13
    • 33846660947 scopus 로고    scopus 로고
    • JAK2 exon 12 mutations in polycythemia vera and idiopathic erythrocytosis
    • Scott L.M., Tong W., Levine R.L., et al. JAK2 exon 12 mutations in polycythemia vera and idiopathic erythrocytosis. N Engl J Med 2007, 356:459-468.
    • (2007) N Engl J Med , vol.356 , pp. 459-468
    • Scott, L.M.1    Tong, W.2    Levine, R.L.3
  • 14
    • 33750534561 scopus 로고    scopus 로고
    • MPL515 mutations in myeloproliferative and other myeloid disorders: a study of 1182 patients
    • Pardanani A.D., Levine R.L., Lasho T., et al. MPL515 mutations in myeloproliferative and other myeloid disorders: a study of 1182 patients. Blood 2006, 108:3472-3476.
    • (2006) Blood , vol.108 , pp. 3472-3476
    • Pardanani, A.D.1    Levine, R.L.2    Lasho, T.3
  • 15
    • 34548138104 scopus 로고    scopus 로고
    • Role of JAK-STAT signaling in the pathogenesis of myeloproliferative disorders
    • Levine R.L., Wernig G. Role of JAK-STAT signaling in the pathogenesis of myeloproliferative disorders. Hematology Am Soc Hematol Educ Program 2006, 1:233-239.
    • (2006) Hematology Am Soc Hematol Educ Program , vol.1 , pp. 233-239
    • Levine, R.L.1    Wernig, G.2
  • 16
    • 77951031498 scopus 로고    scopus 로고
    • Efficacy of the JAK2 inhibitor INCB16562 in a murine model of MPLW515L-induced thrombocytosis and myelofibrosis
    • Koppikar P., Abdel-Wahab O., Hedvat C., et al. Efficacy of the JAK2 inhibitor INCB16562 in a murine model of MPLW515L-induced thrombocytosis and myelofibrosis. Blood 2010, 115:2919-2927.
    • (2010) Blood , vol.115 , pp. 2919-2927
    • Koppikar, P.1    Abdel-Wahab, O.2    Hedvat, C.3
  • 17
    • 74949108515 scopus 로고    scopus 로고
    • Mutations of the e3 ubiquitin ligase cbl family members constitute a novel pathogenic lesion in myeloid malignancies
    • Makishima H., Cazzolli H., Szpurka H., et al. Mutations of the e3 ubiquitin ligase cbl family members constitute a novel pathogenic lesion in myeloid malignancies. J Clin Oncol 2009, 27:6109-6116.
    • (2009) J Clin Oncol , vol.27 , pp. 6109-6116
    • Makishima, H.1    Cazzolli, H.2    Szpurka, H.3
  • 18
    • 77954581139 scopus 로고    scopus 로고
    • Novel mutations and their functional and clinical relevance in myeloproliferative neoplasms: JAK2, MPL, TET2, ASXL1, CBL, IDH and IKZF1
    • Tefferi A. Novel mutations and their functional and clinical relevance in myeloproliferative neoplasms: JAK2, MPL, TET2, ASXL1, CBL, IDH and IKZF1. Leukemia 2010, 24:1128-1138.
    • (2010) Leukemia , vol.24 , pp. 1128-1138
    • Tefferi, A.1
  • 19
    • 77950917237 scopus 로고    scopus 로고
    • Molecular aspects of myeloproliferative neoplasms
    • Delhommeau F., Jeziorowska D., Marzac C., et al. Molecular aspects of myeloproliferative neoplasms. Int J Hematol 2010, 91:165-173.
    • (2010) Int J Hematol , vol.91 , pp. 165-173
    • Delhommeau, F.1    Jeziorowska, D.2    Marzac, C.3
  • 20
    • 80051931772 scopus 로고    scopus 로고
    • New mutations and pathogenesis of myeloproliferative neoplasms
    • Vainchenker W., Delhommeau F., Constantinescu S.N., et al. New mutations and pathogenesis of myeloproliferative neoplasms. Blood 2011, 118:1723-1735.
    • (2011) Blood , vol.118 , pp. 1723-1735
    • Vainchenker, W.1    Delhommeau, F.2    Constantinescu, S.N.3
  • 21
    • 77958021645 scopus 로고    scopus 로고
    • LNK mutation studies in blast-phase myeloproliferative neoplasms, and in chronic-phase disease with TET2, IDH, JAK2 or MPL mutations
    • Pardanani A., Lasho T., Finke C., et al. LNK mutation studies in blast-phase myeloproliferative neoplasms, and in chronic-phase disease with TET2, IDH, JAK2 or MPL mutations. Leukemia 2010, 24:1713-1718.
    • (2010) Leukemia , vol.24 , pp. 1713-1718
    • Pardanani, A.1    Lasho, T.2    Finke, C.3
  • 22
    • 77953485892 scopus 로고    scopus 로고
    • Novel mutations in the inhibitory adaptor protein LNK drive JAK-STAT signaling in patients with myeloproliferative neoplasms
    • Oh S.T., Simonds E.F., Jones C., et al. Novel mutations in the inhibitory adaptor protein LNK drive JAK-STAT signaling in patients with myeloproliferative neoplasms. Blood 2010, 116:988-992.
    • (2010) Blood , vol.116 , pp. 988-992
    • Oh, S.T.1    Simonds, E.F.2    Jones, C.3
  • 23
    • 79952042287 scopus 로고    scopus 로고
    • Genetics of the myeloproliferative neoplasms
    • Abdel-Wahab O. Genetics of the myeloproliferative neoplasms. Curr Opin Hematol 2011, 18:117-123.
    • (2011) Curr Opin Hematol , vol.18 , pp. 117-123
    • Abdel-Wahab, O.1
  • 24
    • 79551575494 scopus 로고    scopus 로고
    • Janus kinase inhibitors for the treatment of myeloproliferative neoplasias and beyond
    • Quintas-Cardama A., Kantarjian H., Cortes J., et al. Janus kinase inhibitors for the treatment of myeloproliferative neoplasias and beyond. Nat Rev Drug Discov 2011, 10:127-140.
    • (2011) Nat Rev Drug Discov , vol.10 , pp. 127-140
    • Quintas-Cardama, A.1    Kantarjian, H.2    Cortes, J.3
  • 25
    • 79959712087 scopus 로고    scopus 로고
    • Ras/Raf/MEK/ERK and PI3K/PTEN/Akt/mTOR inhibitors: rationale and importance to inhibiting these pathways in human health
    • Chappell W.H., Steelman L.S., Long J.M., et al. Ras/Raf/MEK/ERK and PI3K/PTEN/Akt/mTOR inhibitors: rationale and importance to inhibiting these pathways in human health. Oncotarget 2011, 2:135-164.
    • (2011) Oncotarget , vol.2 , pp. 135-164
    • Chappell, W.H.1    Steelman, L.S.2    Long, J.M.3
  • 26
    • 79960235543 scopus 로고    scopus 로고
    • Roles of the Ras/Raf/MEK/ERK pathway in leukemia therapy
    • Steelman L.S., Franklin R.A., Abrams S.L., et al. Roles of the Ras/Raf/MEK/ERK pathway in leukemia therapy. Leukemia 2011, 25:1080-1094.
    • (2011) Leukemia , vol.25 , pp. 1080-1094
    • Steelman, L.S.1    Franklin, R.A.2    Abrams, S.L.3
  • 27
    • 82555170606 scopus 로고    scopus 로고
    • Constitutive MAP kinase activation in hematopoietic stem cells induces a myeloproliferative disorder
    • Chung E., Hsu C.L., Kondo M. Constitutive MAP kinase activation in hematopoietic stem cells induces a myeloproliferative disorder. PLoS One 2011, 6:e28350.
    • (2011) PLoS One , vol.6
    • Chung, E.1    Hsu, C.L.2    Kondo, M.3
  • 28
    • 67749122122 scopus 로고    scopus 로고
    • Targeting PI3K signalling in cancer: opportunities, challenges and limitations
    • Engelman J.A. Targeting PI3K signalling in cancer: opportunities, challenges and limitations. Nat Rev Cancer 2009, 9:550-562.
    • (2009) Nat Rev Cancer , vol.9 , pp. 550-562
    • Engelman, J.A.1
  • 29
    • 43549109620 scopus 로고    scopus 로고
    • The Jak2V617F oncogene associated with myeloproliferative diseases requires a functional FERM domain for transformation and for expression of the Myc and Pim proto-oncogenes
    • Wernig G., Gonneville J.R., Crowley B.J., et al. The Jak2V617F oncogene associated with myeloproliferative diseases requires a functional FERM domain for transformation and for expression of the Myc and Pim proto-oncogenes. Blood 2008, 111:3751-3759.
    • (2008) Blood , vol.111 , pp. 3751-3759
    • Wernig, G.1    Gonneville, J.R.2    Crowley, B.J.3
  • 30
    • 53249137796 scopus 로고    scopus 로고
    • Potential roles for the PIM1 kinase in human cancer- a molecular and therapeutic appraisal
    • Shah N., Pang B., Yeoh K.G., et al. Potential roles for the PIM1 kinase in human cancer- a molecular and therapeutic appraisal. Eur J Cancer 2008, 44:2144-2151.
    • (2008) Eur J Cancer , vol.44 , pp. 2144-2151
    • Shah, N.1    Pang, B.2    Yeoh, K.G.3
  • 31
    • 79960227474 scopus 로고    scopus 로고
    • Concomitant analysis of EZH2 and ASXL1 mutations in myelofibrosis, chronic myelomonocytic leukemia and blast-phase myeloproliferative neoplasms
    • Abdel-Wahab O., Pardanani A., Patel J., et al. Concomitant analysis of EZH2 and ASXL1 mutations in myelofibrosis, chronic myelomonocytic leukemia and blast-phase myeloproliferative neoplasms. Leukemia 2011, 25:1200-1202.
    • (2011) Leukemia , vol.25 , pp. 1200-1202
    • Abdel-Wahab, O.1    Pardanani, A.2    Patel, J.3
  • 32
    • 70450239681 scopus 로고    scopus 로고
    • Mutations of ASXL1 gene in myeloproliferative neoplasms
    • Carbuccia N., Murati A., Trouplin V., et al. Mutations of ASXL1 gene in myeloproliferative neoplasms. Leukemia 2009, 23:2183-2186.
    • (2009) Leukemia , vol.23 , pp. 2183-2186
    • Carbuccia, N.1    Murati, A.2    Trouplin, V.3
  • 33
    • 79952075257 scopus 로고    scopus 로고
    • Myeloproliferative neoplasms: molecular pathophysiology, essential clinical understanding, and treatment strategies
    • Tefferi A., Vainchenker W. Myeloproliferative neoplasms: molecular pathophysiology, essential clinical understanding, and treatment strategies. J Clin Oncol 2011, 29:573-582.
    • (2011) J Clin Oncol , vol.29 , pp. 573-582
    • Tefferi, A.1    Vainchenker, W.2
  • 34
    • 79955513100 scopus 로고    scopus 로고
    • Aberrations of EZH2 in cancer
    • Chase A., Cross N.C. Aberrations of EZH2 in cancer. Clin Cancer Res 2011, 17:2613-2618.
    • (2011) Clin Cancer Res , vol.17 , pp. 2613-2618
    • Chase, A.1    Cross, N.C.2
  • 35
    • 78650495421 scopus 로고    scopus 로고
    • Polycomb proteins in hematologic malignancies
    • Martin-Perez D., Piris M.A., Sanchez-Beato M. Polycomb proteins in hematologic malignancies. Blood 2010, 116:5465-5475.
    • (2010) Blood , vol.116 , pp. 5465-5475
    • Martin-Perez, D.1    Piris, M.A.2    Sanchez-Beato, M.3
  • 36
    • 77955085750 scopus 로고    scopus 로고
    • Inactivating mutations of the histone methyltransferase gene EZH2 in myeloid disorders
    • Ernst T., Chase A.J., Score J., et al. Inactivating mutations of the histone methyltransferase gene EZH2 in myeloid disorders. Nat Genet 2010, 42:722-726.
    • (2010) Nat Genet , vol.42 , pp. 722-726
    • Ernst, T.1    Chase, A.J.2    Score, J.3
  • 37
    • 81055126771 scopus 로고    scopus 로고
    • EZH2 mutational status predicts poor survival in myelofibrosis
    • Guglielmelli P., Biamonte F., Score J., et al. EZH2 mutational status predicts poor survival in myelofibrosis. Blood 2011, 118:5227-5234.
    • (2011) Blood , vol.118 , pp. 5227-5234
    • Guglielmelli, P.1    Biamonte, F.2    Score, J.3
  • 38
    • 84855787692 scopus 로고    scopus 로고
    • ASXL1 mutations in primary and secondary myelofibrosis
    • Ricci C., Spinelli O., Salmoiraghi S., et al. ASXL1 mutations in primary and secondary myelofibrosis. Br J Haematol 2012, 156:404-407.
    • (2012) Br J Haematol , vol.156 , pp. 404-407
    • Ricci, C.1    Spinelli, O.2    Salmoiraghi, S.3
  • 41
    • 80053629571 scopus 로고    scopus 로고
    • Disruption of the ASXL1 gene is frequent in primary, post-essential thrombocytosis and post-polycythemia vera myelofibrosis, but not essential thrombocytosis or polycythemia vera: analysis of molecular genetics and clinical phenotypes
    • Stein B.L., Williams D.M., O'Keefe C., et al. Disruption of the ASXL1 gene is frequent in primary, post-essential thrombocytosis and post-polycythemia vera myelofibrosis, but not essential thrombocytosis or polycythemia vera: analysis of molecular genetics and clinical phenotypes. Haematologica 2011, 96:1462-1469.
    • (2011) Haematologica , vol.96 , pp. 1462-1469
    • Stein, B.L.1    Williams, D.M.2    O'Keefe, C.3
  • 42
    • 78650175023 scopus 로고    scopus 로고
    • Impaired hydroxylation of 5-methylcytosine in myeloid cancers with mutant TET2
    • Ko M., Huang Y., Jankowska A.M., et al. Impaired hydroxylation of 5-methylcytosine in myeloid cancers with mutant TET2. Nature 2010, 468:839-843.
    • (2010) Nature , vol.468 , pp. 839-843
    • Ko, M.1    Huang, Y.2    Jankowska, A.M.3
  • 43
    • 67349124376 scopus 로고    scopus 로고
    • TET2 mutations and their clinical correlates in polycythemia vera, essential thrombocythemia and myelofibrosis
    • Tefferi A., Pardanani A., Lim K.H., et al. TET2 mutations and their clinical correlates in polycythemia vera, essential thrombocythemia and myelofibrosis. Leukemia 2009, 23:905-911.
    • (2009) Leukemia , vol.23 , pp. 905-911
    • Tefferi, A.1    Pardanani, A.2    Lim, K.H.3
  • 44
    • 84860824343 scopus 로고    scopus 로고
    • TET2, ASXL1, IDH1, IDH2, and c-CBL genes in JAK2- and MPL-negative myeloproliferative neoplasms
    • Martinez-Aviles L., Besses C., Alvarez-Larran A., et al. TET2, ASXL1, IDH1, IDH2, and c-CBL genes in JAK2- and MPL-negative myeloproliferative neoplasms. Ann Hematol 2012, 91(4):533-541.
    • (2012) Ann Hematol , vol.91 , Issue.4 , pp. 533-541
    • Martinez-Aviles, L.1    Besses, C.2    Alvarez-Larran, A.3
  • 45
    • 77149134353 scopus 로고    scopus 로고
    • Cancer-associated metabolite 2-hydroxyglutarate accumulates in acute myelogenous leukemia with isocitrate dehydrogenase 1 and 2 mutations
    • Gross S., Cairns R.A., Minden M.D., et al. Cancer-associated metabolite 2-hydroxyglutarate accumulates in acute myelogenous leukemia with isocitrate dehydrogenase 1 and 2 mutations. J Exp Med 2010, 207:339-344.
    • (2010) J Exp Med , vol.207 , pp. 339-344
    • Gross, S.1    Cairns, R.A.2    Minden, M.D.3
  • 46
    • 77649305610 scopus 로고    scopus 로고
    • The common feature of leukemia-associated IDH1 and IDH2 mutations is a neomorphic enzyme activity converting alpha-ketoglutarate to 2-hydroxyglutarate
    • Ward P.S., Patel J., Wise D.R., et al. The common feature of leukemia-associated IDH1 and IDH2 mutations is a neomorphic enzyme activity converting alpha-ketoglutarate to 2-hydroxyglutarate. Cancer Cell 2010, 17:225-234.
    • (2010) Cancer Cell , vol.17 , pp. 225-234
    • Ward, P.S.1    Patel, J.2    Wise, D.R.3
  • 47
    • 78650019179 scopus 로고    scopus 로고
    • Leukemic IDH1 and IDH2 mutations result in a hypermethylation phenotype, disrupt TET2 function, and impair hematopoietic differentiation
    • Figueroa M.E., Abdel-Wahab O., Lu C., et al. Leukemic IDH1 and IDH2 mutations result in a hypermethylation phenotype, disrupt TET2 function, and impair hematopoietic differentiation. Cancer Cell 2010, 18:553-567.
    • (2010) Cancer Cell , vol.18 , pp. 553-567
    • Figueroa, M.E.1    Abdel-Wahab, O.2    Lu, C.3
  • 48
    • 78651463452 scopus 로고    scopus 로고
    • Oncometabolite 2-hydroxyglutarate is a competitive inhibitor of alpha-ketoglutarate-dependent dioxygenases
    • Xu W., Yang H., Liu Y., et al. Oncometabolite 2-hydroxyglutarate is a competitive inhibitor of alpha-ketoglutarate-dependent dioxygenases. Cancer Cell 2011, 19:17-30.
    • (2011) Cancer Cell , vol.19 , pp. 17-30
    • Xu, W.1    Yang, H.2    Liu, Y.3
  • 49
    • 79955547561 scopus 로고    scopus 로고
    • The oncometabolite 2-hydroxyglutarate inhibits histone lysine demethylases
    • Chowdhury R., Yeoh K.K., Tian Y.M., et al. The oncometabolite 2-hydroxyglutarate inhibits histone lysine demethylases. EMBO Rep 2011, 12:463-469.
    • (2011) EMBO Rep , vol.12 , pp. 463-469
    • Chowdhury, R.1    Yeoh, K.K.2    Tian, Y.M.3
  • 50
    • 84858796262 scopus 로고    scopus 로고
    • IDH mutation impairs histone demethylation and results in a block to cell differentiation
    • Lu C., Ward P.S., Kapoor G.S., et al. IDH mutation impairs histone demethylation and results in a block to cell differentiation. Nature 2012, 483:474-478.
    • (2012) Nature , vol.483 , pp. 474-478
    • Lu, C.1    Ward, P.S.2    Kapoor, G.S.3
  • 51
    • 84875006605 scopus 로고    scopus 로고
    • IDH mutations in primary myelofibrosis predict leukemic transformation and shortened survival: clinical evidence for leukemogenic collaboration with JAK2V617F
    • Sulai N., Jimma T., Lasho T.L., et al. IDH mutations in primary myelofibrosis predict leukemic transformation and shortened survival: clinical evidence for leukemogenic collaboration with JAK2V617F. ASH Annual Meeting Abstracts 2011, 118:1751.
    • (2011) ASH Annual Meeting Abstracts , vol.118 , pp. 1751
    • Sulai, N.1    Jimma, T.2    Lasho, T.L.3
  • 52
    • 84857997263 scopus 로고    scopus 로고
    • IDH mutations in primary myelofibrosis predict leukemic transformation and shortened survival: clinical evidence for leukemogenic collaboration with JAK2V617F
    • Tefferi A., Jimma T., Sulai N.H., et al. IDH mutations in primary myelofibrosis predict leukemic transformation and shortened survival: clinical evidence for leukemogenic collaboration with JAK2V617F. Leukemia 2012, 26:475-480.
    • (2012) Leukemia , vol.26 , pp. 475-480
    • Tefferi, A.1    Jimma, T.2    Sulai, N.H.3
  • 53
    • 79960210747 scopus 로고    scopus 로고
    • DNMT3A mutations in myeloproliferative neoplasms
    • Stegelmann F., Bullinger L., Schlenk R.F., et al. DNMT3A mutations in myeloproliferative neoplasms. Leukemia 2011, 25:1217-1219.
    • (2011) Leukemia , vol.25 , pp. 1217-1219
    • Stegelmann, F.1    Bullinger, L.2    Schlenk, R.F.3
  • 54
    • 84555207349 scopus 로고    scopus 로고
    • Dnmt3a is essential for hematopoietic stem cell differentiation
    • Challen G.A., Sun D., Jeong M., et al. Dnmt3a is essential for hematopoietic stem cell differentiation. Nat Genet 2012, 44:23-31.
    • (2012) Nat Genet , vol.44 , pp. 23-31
    • Challen, G.A.1    Sun, D.2    Jeong, M.3
  • 55
    • 84861702964 scopus 로고    scopus 로고
    • Mutation analysis of ASXL1, CBL, DNMT3A, IDH1, IDH2, JAK2, MPL, NF1, SF3B1, SUZ12, and TET2 in myeloproliferative neoplasms
    • Brecqueville M., Rey J., Bertucci F., et al. Mutation analysis of ASXL1, CBL, DNMT3A, IDH1, IDH2, JAK2, MPL, NF1, SF3B1, SUZ12, and TET2 in myeloproliferative neoplasms. Genes Chromosomes Cancer 2012, 51(8):743-755.
    • (2012) Genes Chromosomes Cancer , vol.51 , Issue.8 , pp. 743-755
    • Brecqueville, M.1    Rey, J.2    Bertucci, F.3
  • 56
    • 79960248721 scopus 로고    scopus 로고
    • DNMT3A mutational analysis in primary myelofibrosis, chronic myelomonocytic leukemia and advanced phases of myeloproliferative neoplasms
    • Abdel-Wahab O., Pardanani A., Rampal R., et al. DNMT3A mutational analysis in primary myelofibrosis, chronic myelomonocytic leukemia and advanced phases of myeloproliferative neoplasms. Leukemia 2011, 25:1219-1220.
    • (2011) Leukemia , vol.25 , pp. 1219-1220
    • Abdel-Wahab, O.1    Pardanani, A.2    Rampal, R.3
  • 57
    • 79960255863 scopus 로고    scopus 로고
    • Recurrent DNMT3A mutations in patients with myelodysplastic syndromes
    • Walter M.J., Ding L., Shen D., et al. Recurrent DNMT3A mutations in patients with myelodysplastic syndromes. Leukemia 2011, 25:1153-1158.
    • (2011) Leukemia , vol.25 , pp. 1153-1158
    • Walter, M.J.1    Ding, L.2    Shen, D.3
  • 58
    • 84860772157 scopus 로고    scopus 로고
    • DNMT3A mutations and response to the hypomethylating agent decitabine in acute myeloid leukemia
    • Metzeler K.H., Walker A., Geyer S., et al. DNMT3A mutations and response to the hypomethylating agent decitabine in acute myeloid leukemia. Leukemia 2012, 26(5):1106-1107.
    • (2012) Leukemia , vol.26 , Issue.5 , pp. 1106-1107
    • Metzeler, K.H.1    Walker, A.2    Geyer, S.3
  • 59
    • 84857048507 scopus 로고    scopus 로고
    • New generation small-molecule inhibitors in myeloproliferative neoplasms
    • Passamonti F., Maffioli M., Caramazza D. New generation small-molecule inhibitors in myeloproliferative neoplasms. Curr Opin Hematol 2012, 19:117-123.
    • (2012) Curr Opin Hematol , vol.19 , pp. 117-123
    • Passamonti, F.1    Maffioli, M.2    Caramazza, D.3
  • 60
    • 77956696835 scopus 로고    scopus 로고
    • Safety and efficacy of INCB018424, a JAK1 and JAK2 inhibitor, in myelofibrosis
    • Verstovsek S., Kantarjian H., Mesa R.A., et al. Safety and efficacy of INCB018424, a JAK1 and JAK2 inhibitor, in myelofibrosis. N Engl J Med 2010, 363:1117-1127.
    • (2010) N Engl J Med , vol.363 , pp. 1117-1127
    • Verstovsek, S.1    Kantarjian, H.2    Mesa, R.A.3
  • 61
    • 84861780038 scopus 로고    scopus 로고
    • Ruxolitinib: the first FDA approved therapy for the treatment of myelofibrosis
    • Mascarenhas J., Hoffman R. Ruxolitinib: the first FDA approved therapy for the treatment of myelofibrosis. Clin Cancer Res 2012, 18(11):3008-3014.
    • (2012) Clin Cancer Res , vol.18 , Issue.11 , pp. 3008-3014
    • Mascarenhas, J.1    Hoffman, R.2
  • 62
    • 84858855487 scopus 로고    scopus 로고
    • JAK inhibitors for myeloproliferative neoplasms; clarifying facts from myths
    • Tefferi A. JAK inhibitors for myeloproliferative neoplasms; clarifying facts from myths. Blood 2012, 119:2721-2730.
    • (2012) Blood , vol.119 , pp. 2721-2730
    • Tefferi, A.1
  • 63
    • 79952333359 scopus 로고    scopus 로고
    • Safety and efficacy of TG101348, a selective JAK2 inhibitor, in myelofibrosis
    • Pardanani A., Gotlib J.R., Jamieson C., et al. Safety and efficacy of TG101348, a selective JAK2 inhibitor, in myelofibrosis. J Clin Oncol 2011, 29:789-796.
    • (2011) J Clin Oncol , vol.29 , pp. 789-796
    • Pardanani, A.1    Gotlib, J.R.2    Jamieson, C.3
  • 64
    • 84857828112 scopus 로고    scopus 로고
    • Challenges facing JAK inhibitor therapy for myeloproliferative neoplasms
    • Tefferi A. Challenges facing JAK inhibitor therapy for myeloproliferative neoplasms. N Engl J Med 2012, 366:844-846.
    • (2012) N Engl J Med , vol.366 , pp. 844-846
    • Tefferi, A.1
  • 65
    • 84875006650 scopus 로고    scopus 로고
    • Kinase domain mutations in JAK2V617F confer resistance to the novel JAK2 inhibitor ruxolitinib
    • Deshpande A., Reddy M.M., Schade G., et al. Kinase domain mutations in JAK2V617F confer resistance to the novel JAK2 inhibitor ruxolitinib. ASH Annual Meeting Abstracts 2011, 118:125.
    • (2011) ASH Annual Meeting Abstracts , vol.118 , pp. 125
    • Deshpande, A.1    Reddy, M.M.2    Schade, G.3
  • 67
    • 79751532774 scopus 로고    scopus 로고
    • JAK inhibitor therapy for myelofibrosis: critical assessment of value and limitations
    • Pardanani A., Vannucchi A.M., Passamonti F., et al. JAK inhibitor therapy for myelofibrosis: critical assessment of value and limitations. Leukemia 2011, 25:218-225.
    • (2011) Leukemia , vol.25 , pp. 218-225
    • Pardanani, A.1    Vannucchi, A.M.2    Passamonti, F.3
  • 68
    • 79953711716 scopus 로고    scopus 로고
    • How I treat myelofibrosis
    • Tefferi A. How I treat myelofibrosis. Blood 2011, 117:3494-3504.
    • (2011) Blood , vol.117 , pp. 3494-3504
    • Tefferi, A.1
  • 69
    • 0842309105 scopus 로고    scopus 로고
    • Multiple signaling pathways are involved in erythropoietin-independent differentiation of erythroid progenitors in polycythemia vera
    • Ugo V., Marzac C., Teyssandier I., et al. Multiple signaling pathways are involved in erythropoietin-independent differentiation of erythroid progenitors in polycythemia vera. Exp Hematol 2004, 32:179-187.
    • (2004) Exp Hematol , vol.32 , pp. 179-187
    • Ugo, V.1    Marzac, C.2    Teyssandier, I.3
  • 70
    • 77950916292 scopus 로고    scopus 로고
    • The mTOR inhibitor, RAD001, inhibits the growth of cells from patients with myeloproliferative neoplasms
    • Vannucchi A.M., Bogani C., Bartalucci N., et al. The mTOR inhibitor, RAD001, inhibits the growth of cells from patients with myeloproliferative neoplasms. ASH Annual Meeting Abstracts 2009, 114:2914.
    • (2009) ASH Annual Meeting Abstracts , vol.114 , pp. 2914
    • Vannucchi, A.M.1    Bogani, C.2    Bartalucci, N.3
  • 71
    • 77950917641 scopus 로고    scopus 로고
    • RAD001, an inhibitor of mTOR, shows clinical activity in a phase I/II study in patients with primary myelofibrosis (PMF) and post polycythemia vera/essential thrombocythemia myelofibrosis (PPV/PET MF)
    • Vannucchi A.M., Guglielmelli P., Gattoni E., et al. RAD001, an inhibitor of mTOR, shows clinical activity in a phase I/II study in patients with primary myelofibrosis (PMF) and post polycythemia vera/essential thrombocythemia myelofibrosis (PPV/PET MF). ASH Annual Meeting Abstracts 2009, 114:307.
    • (2009) ASH Annual Meeting Abstracts , vol.114 , pp. 307
    • Vannucchi, A.M.1    Guglielmelli, P.2    Gattoni, E.3
  • 72
    • 84859778293 scopus 로고    scopus 로고
    • MTOR signaling in growth control and disease
    • Laplante M., Sabatini D.M. mTOR signaling in growth control and disease. Cell 2012, 149:274-293.
    • (2012) Cell , vol.149 , pp. 274-293
    • Laplante, M.1    Sabatini, D.M.2
  • 73
    • 80052184481 scopus 로고    scopus 로고
    • Safety and efficacy of everolimus, a mTOR inhibitor, as single agent in a phase 1/2 study in patients with myelofibrosis
    • Guglielmelli P., Barosi G., Rambaldi A., et al. Safety and efficacy of everolimus, a mTOR inhibitor, as single agent in a phase 1/2 study in patients with myelofibrosis. Blood 2011, 118:2069-2076.
    • (2011) Blood , vol.118 , pp. 2069-2076
    • Guglielmelli, P.1    Barosi, G.2    Rambaldi, A.3
  • 74
    • 84864036647 scopus 로고    scopus 로고
    • Synergistic activity of combinations of JAK2 kinase inhibitor with PI3K/mTOR, MEK or PIM kinase inhibitor against human myeloproliferative neoplasm cells expressing JAK2V617F
    • Fiskus W., Manepalli R.R., Balusu R., et al. Synergistic activity of combinations of JAK2 kinase inhibitor with PI3K/mTOR, MEK or PIM kinase inhibitor against human myeloproliferative neoplasm cells expressing JAK2V617F. ASH Annual Meeting Abstracts 2010, 116:798.
    • (2010) ASH Annual Meeting Abstracts , vol.116 , pp. 798
    • Fiskus, W.1    Manepalli, R.R.2    Balusu, R.3
  • 75
    • 84875007306 scopus 로고    scopus 로고
    • Inhibition of AKT signaling potently inhibits the growth of JAK and MPL-mutant cells in the myeloproliferative neoplasms in vitro and in vivo
    • Khan I., Huang Z., Wen Q.J., et al. Inhibition of AKT signaling potently inhibits the growth of JAK and MPL-mutant cells in the myeloproliferative neoplasms in vitro and in vivo. ASH Annual Meeting Abstracts 2011, 118:3862.
    • (2011) ASH Annual Meeting Abstracts , vol.118 , pp. 3862
    • Khan, I.1    Huang, Z.2    Wen, Q.J.3
  • 76
    • 79952704794 scopus 로고    scopus 로고
    • Role of Ras/Raf/MEK/ERK signaling in physiological hematopoiesis and leukemia development
    • Chung E., Kondo M. Role of Ras/Raf/MEK/ERK signaling in physiological hematopoiesis and leukemia development. Immunol Res 2011, 49:248-268.
    • (2011) Immunol Res , vol.49 , pp. 248-268
    • Chung, E.1    Kondo, M.2
  • 77
    • 77953237268 scopus 로고    scopus 로고
    • Phase II study of the oral MEK inhibitor AZD6244 in advanced acute myeloid leukemia (AML)
    • Odenike O., Curran E., Iyengar N., et al. Phase II study of the oral MEK inhibitor AZD6244 in advanced acute myeloid leukemia (AML). ASH Annual Meeting Abstracts 2009, 114:2081.
    • (2009) ASH Annual Meeting Abstracts , vol.114 , pp. 2081
    • Odenike, O.1    Curran, E.2    Iyengar, N.3
  • 78
    • 29244464759 scopus 로고    scopus 로고
    • The oncogenic serine/threonine kinase Pim-1 directly phosphorylates and activates the G2/M specific phosphatase Cdc25C
    • Bachmann M., Kosan C., Xing P.X., et al. The oncogenic serine/threonine kinase Pim-1 directly phosphorylates and activates the G2/M specific phosphatase Cdc25C. Int J Biochem Cell Biol 2006, 38:430-443.
    • (2006) Int J Biochem Cell Biol , vol.38 , pp. 430-443
    • Bachmann, M.1    Kosan, C.2    Xing, P.X.3
  • 80
    • 80052025089 scopus 로고    scopus 로고
    • The oncogenic PIM kinase family regulates drug resistance through multiple mechanisms
    • Isaac M., Siu A., Jongstra J. The oncogenic PIM kinase family regulates drug resistance through multiple mechanisms. Drug Resist Updat 2011, 14:203-211.
    • (2011) Drug Resist Updat , vol.14 , pp. 203-211
    • Isaac, M.1    Siu, A.2    Jongstra, J.3
  • 81
    • 78650606450 scopus 로고    scopus 로고
    • For better or for worse: the role of Pim oncogenes in tumorigenesis
    • Nawijn M.C., Alendar A., Berns A. For better or for worse: the role of Pim oncogenes in tumorigenesis. Nat Rev Cancer 2011, 11:23-34.
    • (2011) Nat Rev Cancer , vol.11 , pp. 23-34
    • Nawijn, M.C.1    Alendar, A.2    Berns, A.3
  • 82
    • 68149158207 scopus 로고    scopus 로고
    • PIM1 protein kinase regulates PRAS40 phosphorylation and mTOR activity in FDCP1 cells
    • Zhang F., Beharry Z.M., Harris T.E., et al. PIM1 protein kinase regulates PRAS40 phosphorylation and mTOR activity in FDCP1 cells. Cancer Biol Ther 2009, 8:846-853.
    • (2009) Cancer Biol Ther , vol.8 , pp. 846-853
    • Zhang, F.1    Beharry, Z.M.2    Harris, T.E.3
  • 83
    • 78649641859 scopus 로고    scopus 로고
    • Synergistic activity of co-treatment with pim1 kinase inhibitor SGI-1776 and histone deacetylase inhibitor panobinostat or heat shock protein 90 inhibitor AUY922 against human CML and myeloproliferative neoplasm (MPN) cells
    • [abstract: 2651]
    • Fiskus W.C., Buckley K.M., Rao R., et al. Synergistic activity of co-treatment with pim1 kinase inhibitor SGI-1776 and histone deacetylase inhibitor panobinostat or heat shock protein 90 inhibitor AUY922 against human CML and myeloproliferative neoplasm (MPN) cells. ASH Annual Meeting Abstracts 2009, 114. [abstract: 2651].
    • (2009) ASH Annual Meeting Abstracts , vol.114
    • Fiskus, W.C.1    Buckley, K.M.2    Rao, R.3
  • 84
    • 84875003825 scopus 로고    scopus 로고
    • AZD1208, a novel, potent and selective pan PIM kinase inhibitor, demonstrates efficacy in models of acute myeloid leukemia
    • [abstract: 1540]
    • Keeton E., Palakurthi S., Alimzhanov M., et al. AZD1208, a novel, potent and selective pan PIM kinase inhibitor, demonstrates efficacy in models of acute myeloid leukemia. ASH Annual Meeting Abstracts 2011, 118. [abstract: 1540].
    • (2011) ASH Annual Meeting Abstracts , vol.118
    • Keeton, E.1    Palakurthi, S.2    Alimzhanov, M.3
  • 85
    • 57449114002 scopus 로고    scopus 로고
    • Enhanced histone deacetylase enzyme activity in primary myelofibrosis
    • Wang J.C., Chen C., Dumlao T., et al. Enhanced histone deacetylase enzyme activity in primary myelofibrosis. Leuk Lymphoma 2008, 49:2321-2327.
    • (2008) Leuk Lymphoma , vol.49 , pp. 2321-2327
    • Wang, J.C.1    Chen, C.2    Dumlao, T.3
  • 86
    • 78651358562 scopus 로고    scopus 로고
    • Deactylase inhibition in myeloproliferative neoplasms
    • Mithraprabhu S., Grigoriadis G., Khong T., et al. Deactylase inhibition in myeloproliferative neoplasms. Invest New Drugs 2010, 28(Suppl 1):S50-S57.
    • (2010) Invest New Drugs , vol.28 , Issue.SUPPL 1
    • Mithraprabhu, S.1    Grigoriadis, G.2    Khong, T.3
  • 87
    • 30344477367 scopus 로고    scopus 로고
    • Histone deacetylase inhibitors and the promise of epigenetic (and more) treatments for cancer
    • Minucci S., Pelicci P.G. Histone deacetylase inhibitors and the promise of epigenetic (and more) treatments for cancer. Nat Rev Cancer 2006, 6:38-51.
    • (2006) Nat Rev Cancer , vol.6 , pp. 38-51
    • Minucci, S.1    Pelicci, P.G.2
  • 88
    • 84860371953 scopus 로고    scopus 로고
    • Epigenetic protein families: a new frontier for drug discovery
    • Arrowsmith C.H., Bountra C., Fish P.V., et al. Epigenetic protein families: a new frontier for drug discovery. Nat Rev Drug Discov 2012, 11:384-400.
    • (2012) Nat Rev Drug Discov , vol.11 , pp. 384-400
    • Arrowsmith, C.H.1    Bountra, C.2    Fish, P.V.3
  • 89
    • 28044471827 scopus 로고    scopus 로고
    • Acetylation and deacetylation of non-histone proteins
    • Glozak M.A., Sengupta N., Zhang X., et al. Acetylation and deacetylation of non-histone proteins. Gene 2005, 363:15-23.
    • (2005) Gene , vol.363 , pp. 15-23
    • Glozak, M.A.1    Sengupta, N.2    Zhang, X.3
  • 90
    • 68949212379 scopus 로고    scopus 로고
    • Lysine acetylation targets protein complexes and co-regulates major cellular functions
    • Choudhary C., Kumar C., Gnad F., et al. Lysine acetylation targets protein complexes and co-regulates major cellular functions. Science 2009, 325:834-840.
    • (2009) Science , vol.325 , pp. 834-840
    • Choudhary, C.1    Kumar, C.2    Gnad, F.3
  • 91
    • 84863012417 scopus 로고    scopus 로고
    • Histone deacetylases mediate the silencing of miR-15a, miR-16, and miR-29b in chronic lymphocytic leukemia
    • Sampath D., Liu C., Vasan K., et al. Histone deacetylases mediate the silencing of miR-15a, miR-16, and miR-29b in chronic lymphocytic leukemia. Blood 2012, 119:1162-1172.
    • (2012) Blood , vol.119 , pp. 1162-1172
    • Sampath, D.1    Liu, C.2    Vasan, K.3
  • 92
    • 73949128107 scopus 로고    scopus 로고
    • Histone deacetylase inhibitors in cancer therapy
    • Lane A.A., Chabner B.A. Histone deacetylase inhibitors in cancer therapy. J Clin Oncol 2009, 27:5459-5468.
    • (2009) J Clin Oncol , vol.27 , pp. 5459-5468
    • Lane, A.A.1    Chabner, B.A.2
  • 93
    • 36048958965 scopus 로고    scopus 로고
    • Histone deacetylase inhibitors: overview and perspectives
    • Dokmanovic M., Clarke C., Marks P.A. Histone deacetylase inhibitors: overview and perspectives. Mol Cancer Res 2007, 5:981-989.
    • (2007) Mol Cancer Res , vol.5 , pp. 981-989
    • Dokmanovic, M.1    Clarke, C.2    Marks, P.A.3
  • 94
    • 78651352243 scopus 로고    scopus 로고
    • Histone deacetylase inhibitors: potential targets responsible for their anti-cancer effect
    • Dickinson M., Johnstone R.W., Prince H.M. Histone deacetylase inhibitors: potential targets responsible for their anti-cancer effect. Invest New Drugs 2010, 28:3-20.
    • (2010) Invest New Drugs , vol.28 , pp. 3-20
    • Dickinson, M.1    Johnstone, R.W.2    Prince, H.M.3
  • 95
    • 33646495294 scopus 로고    scopus 로고
    • The histone deacetylase inhibitor ITF2357 reduces production of pro-inflammatory cytokines in vitro and systemic inflammation in vivo
    • Leoni F., Fossati G., Lewis E.C., et al. The histone deacetylase inhibitor ITF2357 reduces production of pro-inflammatory cytokines in vitro and systemic inflammation in vivo. Mol Med 2005, 11:1-15.
    • (2005) Mol Med , vol.11 , pp. 1-15
    • Leoni, F.1    Fossati, G.2    Lewis, E.C.3
  • 96
    • 22844432021 scopus 로고    scopus 로고
    • Inhibition of histone deacetylase 6 acetylates and disrupts the chaperone function of heat shock protein 90: a novel basis for antileukemia activity of histone deacetylase inhibitors
    • Bali P., Pranpat M., Bradner J., et al. Inhibition of histone deacetylase 6 acetylates and disrupts the chaperone function of heat shock protein 90: a novel basis for antileukemia activity of histone deacetylase inhibitors. J Biol Chem 2005, 280:26729-26734.
    • (2005) J Biol Chem , vol.280 , pp. 26729-26734
    • Bali, P.1    Pranpat, M.2    Bradner, J.3
  • 97
    • 33846225576 scopus 로고    scopus 로고
    • Histone deacetylase inhibitors deplete enhancer of zeste 2 and associated polycomb repressive complex 2 proteins in human acute leukemia cells
    • Fiskus W., Pranpat M., Balasis M., et al. Histone deacetylase inhibitors deplete enhancer of zeste 2 and associated polycomb repressive complex 2 proteins in human acute leukemia cells. Mol Cancer Ther 2006, 5:3096-3104.
    • (2006) Mol Cancer Ther , vol.5 , pp. 3096-3104
    • Fiskus, W.1    Pranpat, M.2    Balasis, M.3
  • 98
    • 33745888156 scopus 로고    scopus 로고
    • Combined effects of novel tyrosine kinase inhibitor AMN107 and histone deacetylase inhibitor LBH589 against Bcr-Abl-expressing human leukemia cells
    • Fiskus W., Pranpat M., Bali P., et al. Combined effects of novel tyrosine kinase inhibitor AMN107 and histone deacetylase inhibitor LBH589 against Bcr-Abl-expressing human leukemia cells. Blood 2006, 108:645-652.
    • (2006) Blood , vol.108 , pp. 645-652
    • Fiskus, W.1    Pranpat, M.2    Bali, P.3
  • 99
    • 20844444898 scopus 로고    scopus 로고
    • Combination of the histone deacetylase inhibitor LBH589 and the hsp90 inhibitor 17-AAG is highly active against human CML-BC cells and AML cells with activating mutation of FLT-3
    • George P., Bali P., Annavarapu S., et al. Combination of the histone deacetylase inhibitor LBH589 and the hsp90 inhibitor 17-AAG is highly active against human CML-BC cells and AML cells with activating mutation of FLT-3. Blood 2005, 105:1768-1776.
    • (2005) Blood , vol.105 , pp. 1768-1776
    • George, P.1    Bali, P.2    Annavarapu, S.3
  • 100
    • 73949136283 scopus 로고    scopus 로고
    • Cotreatment with panobinostat and JAK2 inhibitor TG101209 attenuates JAK2V617F levels and signaling and exerts synergistic cytotoxic effects against human myeloproliferative neoplastic cells
    • Wang Y., Fiskus W., Chong D.G., et al. Cotreatment with panobinostat and JAK2 inhibitor TG101209 attenuates JAK2V617F levels and signaling and exerts synergistic cytotoxic effects against human myeloproliferative neoplastic cells. Blood 2009, 114:5024-5033.
    • (2009) Blood , vol.114 , pp. 5024-5033
    • Wang, Y.1    Fiskus, W.2    Chong, D.G.3
  • 101
    • 68049144931 scopus 로고    scopus 로고
    • Panobinostat treatment depletes EZH2 and DNMT1 levels and enhances decitabine mediated de-repression of JunB and loss of survival of human acute leukemia cells
    • Fiskus W., Buckley K., Rao R., et al. Panobinostat treatment depletes EZH2 and DNMT1 levels and enhances decitabine mediated de-repression of JunB and loss of survival of human acute leukemia cells. Cancer Biol Ther 2009, 8:939-950.
    • (2009) Cancer Biol Ther , vol.8 , pp. 939-950
    • Fiskus, W.1    Buckley, K.2    Rao, R.3
  • 102
    • 77957284593 scopus 로고    scopus 로고
    • Activity of oral panobinostat (LBH589) in patients with myelofibrosis
    • [abstract: 2898]
    • DeAngelo D.J., Spencer A., Fischer T., et al. Activity of oral panobinostat (LBH589) in patients with myelofibrosis. ASH Annual Meeting Abstracts 2009, 114. [abstract: 2898].
    • (2009) ASH Annual Meeting Abstracts , vol.114
    • DeAngelo, D.J.1    Spencer, A.2    Fischer, T.3
  • 103
    • 84867496036 scopus 로고    scopus 로고
    • Prolonged low dose therapy with a pan-deacetylase inhibitor, panobinostat (LBH589), in patients with myelofibrosis
    • [abstract: 794]
    • Mascarenhas J., Mercado A., Rodriguez A., et al. Prolonged low dose therapy with a pan-deacetylase inhibitor, panobinostat (LBH589), in patients with myelofibrosis. ASH Annual Meeting Abstracts 2011, 118. [abstract: 794].
    • (2011) ASH Annual Meeting Abstracts , vol.118
    • Mascarenhas, J.1    Mercado, A.2    Rodriguez, A.3
  • 104
    • 84866629631 scopus 로고    scopus 로고
    • Improved efficacy upon combined JAK1/2 and pan-deacetylase inhibition using ruxolitinib (INC424) and panobinostat (LBH589) in preclinical mouse models of JAK2V617F-driven disease
    • Baffert F., Evrot E., Ebel N., et al. Improved efficacy upon combined JAK1/2 and pan-deacetylase inhibition using ruxolitinib (INC424) and panobinostat (LBH589) in preclinical mouse models of JAK2V617F-driven disease. ASH Annual Meeting Abstracts 2011, 118:798.
    • (2011) ASH Annual Meeting Abstracts , vol.118 , pp. 798
    • Baffert, F.1    Evrot, E.2    Ebel, N.3
  • 105
    • 42349087790 scopus 로고    scopus 로고
    • The histone deacetylase inhibitor ITF2357 selectively targets cells bearing mutated JAK2(V617F)
    • Guerini V., Barbui V., Spinelli O., et al. The histone deacetylase inhibitor ITF2357 selectively targets cells bearing mutated JAK2(V617F). Leukemia 2008, 22:740-747.
    • (2008) Leukemia , vol.22 , pp. 740-747
    • Guerini, V.1    Barbui, V.2    Spinelli, O.3
  • 106
    • 77955168425 scopus 로고    scopus 로고
    • A pilot study of the Histone-Deacetylase inhibitor Givinostat in patients with JAK2V617F positive chronic myeloproliferative neoplasms
    • Rambaldi A., Dellacasa C.M., Finazzi G., et al. A pilot study of the Histone-Deacetylase inhibitor Givinostat in patients with JAK2V617F positive chronic myeloproliferative neoplasms. Br J Haematol 2010, 150:446-455.
    • (2010) Br J Haematol , vol.150 , pp. 446-455
    • Rambaldi, A.1    Dellacasa, C.M.2    Finazzi, G.3
  • 107
    • 84866602529 scopus 로고    scopus 로고
    • A phase II study of the HDAC inhibitor givinostat in combination with hydroxyurea in patients with polycythemia vera resistant to hydroxyurea monotherapy
    • Rambaldi A., Finazzi G., Vannucchi A.M., et al. A phase II study of the HDAC inhibitor givinostat in combination with hydroxyurea in patients with polycythemia vera resistant to hydroxyurea monotherapy. ASH Annual Meeting Abstracts 2011, 118:1748.
    • (2011) ASH Annual Meeting Abstracts , vol.118 , pp. 1748
    • Rambaldi, A.1    Finazzi, G.2    Vannucchi, A.M.3
  • 108
    • 78650670563 scopus 로고    scopus 로고
    • Sequential treatment of CD34+ cells from patients with primary myelofibrosis with chromatin-modifying agents eliminate JAK2V617F-positive NOD/SCID marrow repopulating cells
    • Wang X., Zhang W., Tripodi J., et al. Sequential treatment of CD34+ cells from patients with primary myelofibrosis with chromatin-modifying agents eliminate JAK2V617F-positive NOD/SCID marrow repopulating cells. Blood 2010, 116:5972-5982.
    • (2010) Blood , vol.116 , pp. 5972-5982
    • Wang, X.1    Zhang, W.2    Tripodi, J.3
  • 109
    • 34447118173 scopus 로고    scopus 로고
    • Effects of chromatin-modifying agents on CD34+ cells from patients with idiopathic myelofibrosis
    • Shi J., Zhao Y., Ishii T., et al. Effects of chromatin-modifying agents on CD34+ cells from patients with idiopathic myelofibrosis. Cancer Res 2007, 67:6417-6424.
    • (2007) Cancer Res , vol.67 , pp. 6417-6424
    • Shi, J.1    Zhao, Y.2    Ishii, T.3
  • 110
    • 78649658732 scopus 로고    scopus 로고
    • Myeloproliferative neoplasms: new translational therapies
    • Mascarenhas J., Hoffman R. Myeloproliferative neoplasms: new translational therapies. Mt Sinai J Med 2010, 77:667-683.
    • (2010) Mt Sinai J Med , vol.77 , pp. 667-683
    • Mascarenhas, J.1    Hoffman, R.2
  • 111
    • 84866336515 scopus 로고    scopus 로고
    • A novel selective EZH2 inhibitor exhibits anti-tumor activity in lymphoma with EZH2 activating mutations
    • March 31-April 4. Chicago (IL), Philadelphia: AACR; 2012. Abstract 4700.
    • Creasy CL, McCabe MT, Korenchuk S, et al. A novel selective EZH2 inhibitor exhibits anti-tumor activity in lymphoma with EZH2 activating mutations. Proceedings of the 103rd Annual Meeting of the American Association for Cancer Research; 2012 March 31-April 4. Chicago (IL), Philadelphia: AACR; 2012. Abstract 4700.
    • (2012) Proceedings of the 103rd Annual Meeting of the American Association for Cancer Research
    • Creasy, C.L.1    McCabe, M.T.2    Korenchuk, S.3
  • 112
    • 0036810352 scopus 로고    scopus 로고
    • Heat-shock protein 90, a chaperone for folding and regulation
    • Picard D. Heat-shock protein 90, a chaperone for folding and regulation. Cell Mol Life Sci 2002, 59:1640-1648.
    • (2002) Cell Mol Life Sci , vol.59 , pp. 1640-1648
    • Picard, D.1
  • 113
    • 77954945333 scopus 로고    scopus 로고
    • Targeting the dynamic HSP90 complex in cancer
    • Trepel J., Mollapour M., Giaccone G., et al. Targeting the dynamic HSP90 complex in cancer. Nat Rev Cancer 2010, 10:537-549.
    • (2010) Nat Rev Cancer , vol.10 , pp. 537-549
    • Trepel, J.1    Mollapour, M.2    Giaccone, G.3
  • 114
    • 77953916528 scopus 로고    scopus 로고
    • HSP90 at the hub of protein homeostasis: emerging mechanistic insights
    • Taipale M., Jarosz D.F., Lindquist S. HSP90 at the hub of protein homeostasis: emerging mechanistic insights. Nat Rev Mol Cell Biol 2010, 11:515-528.
    • (2010) Nat Rev Mol Cell Biol , vol.11 , pp. 515-528
    • Taipale, M.1    Jarosz, D.F.2    Lindquist, S.3
  • 115
    • 82555173115 scopus 로고    scopus 로고
    • Heat shock protein 90 inhibitor is synergistic with JAK2 inhibitor and overcomes resistance to JAK2-TKI in human myeloproliferative neoplasm cells
    • Fiskus W., Verstovsek S., Manshouri T., et al. Heat shock protein 90 inhibitor is synergistic with JAK2 inhibitor and overcomes resistance to JAK2-TKI in human myeloproliferative neoplasm cells. Clin Cancer Res 2011, 17:7347-7358.
    • (2011) Clin Cancer Res , vol.17 , pp. 7347-7358
    • Fiskus, W.1    Verstovsek, S.2    Manshouri, T.3
  • 116
    • 2442695516 scopus 로고    scopus 로고
    • Cotreatment with 17-allylamino-demethoxygeldanamycin and FLT-3 kinase inhibitor PKC412 is highly effective against human acute myelogenous leukemia cells with mutant FLT-3
    • George P., Bali P., Cohen P., et al. Cotreatment with 17-allylamino-demethoxygeldanamycin and FLT-3 kinase inhibitor PKC412 is highly effective against human acute myelogenous leukemia cells with mutant FLT-3. Cancer Res 2004, 64:3645-3652.
    • (2004) Cancer Res , vol.64 , pp. 3645-3652
    • George, P.1    Bali, P.2    Cohen, P.3
  • 117
    • 41149111451 scopus 로고    scopus 로고
    • The Hsp90 molecular chaperone: an open and shut case for treatment
    • Pearl L.H., Prodromou C., Workman P. The Hsp90 molecular chaperone: an open and shut case for treatment. Biochem J 2008, 410:439-453.
    • (2008) Biochem J , vol.410 , pp. 439-453
    • Pearl, L.H.1    Prodromou, C.2    Workman, P.3
  • 118
    • 25844519550 scopus 로고    scopus 로고
    • HSP90 and the chaperoning of cancer
    • Whitesell L., Lindquist S.L. HSP90 and the chaperoning of cancer. Nat Rev Cancer 2005, 5:761-772.
    • (2005) Nat Rev Cancer , vol.5 , pp. 761-772
    • Whitesell, L.1    Lindquist, S.L.2
  • 119
    • 17144410021 scopus 로고    scopus 로고
    • Pim-1 kinase stability is regulated by heat shock proteins and the ubiquitin-proteasome pathway
    • Shay K.P., Wang Z., Xing P.X., et al. Pim-1 kinase stability is regulated by heat shock proteins and the ubiquitin-proteasome pathway. Mol Cancer Res 2005, 3:170-181.
    • (2005) Mol Cancer Res , vol.3 , pp. 170-181
    • Shay, K.P.1    Wang, Z.2    Xing, P.X.3
  • 120
    • 77950929054 scopus 로고    scopus 로고
    • Phase I study of the heat shock protein 90 inhibitor alvespimycin (KOS-1022, 17-DMAG) administered intravenously twice weekly to patients with acute myeloid leukemia
    • Lancet J.E., Gojo I., Burton M., et al. Phase I study of the heat shock protein 90 inhibitor alvespimycin (KOS-1022, 17-DMAG) administered intravenously twice weekly to patients with acute myeloid leukemia. Leukemia 2010, 24:699-705.
    • (2010) Leukemia , vol.24 , pp. 699-705
    • Lancet, J.E.1    Gojo, I.2    Burton, M.3
  • 121
    • 79960983822 scopus 로고    scopus 로고
    • AUY922, a novel HSP90 inhibitor: final results of a first-in-human study in patients with advanced solid malignancies
    • [abstract: 2528]
    • Samuel T.A., Sessa C., Britten C., et al. AUY922, a novel HSP90 inhibitor: final results of a first-in-human study in patients with advanced solid malignancies. J Clin Oncol 2010, 28(Suppl):15s. [abstract: 2528].
    • (2010) J Clin Oncol , vol.28 , Issue.SUPPL
    • Samuel, T.A.1    Sessa, C.2    Britten, C.3
  • 122
    • 79955044159 scopus 로고    scopus 로고
    • Multifaceted intervention by the Hsp90 inhibitor ganetespib (STA-9090) in cancer cells with activated JAK/STAT signaling
    • Proia D.A., Foley K.P., Korbut T., et al. Multifaceted intervention by the Hsp90 inhibitor ganetespib (STA-9090) in cancer cells with activated JAK/STAT signaling. PLoS One 2011, 6:e18552.
    • (2011) PLoS One , vol.6
    • Proia, D.A.1    Foley, K.P.2    Korbut, T.3
  • 123
    • 77957854088 scopus 로고    scopus 로고
    • HSP90 is a therapeutic target in JAK2-dependent myeloproliferative neoplasms in mice and humans
    • Marubayashi S., Koppikar P., Taldone T., et al. HSP90 is a therapeutic target in JAK2-dependent myeloproliferative neoplasms in mice and humans. J Clin Invest 2010, 120:3578-3593.
    • (2010) J Clin Invest , vol.120 , pp. 3578-3593
    • Marubayashi, S.1    Koppikar, P.2    Taldone, T.3
  • 124
    • 84856932936 scopus 로고    scopus 로고
    • Genetic resistance to JAK2 enzymatic inhibitors is overcome by HSP90 inhibition
    • Weigert O., Lane A.A., Bird L., et al. Genetic resistance to JAK2 enzymatic inhibitors is overcome by HSP90 inhibition. J Exp Med 2012, 209:259-273.
    • (2012) J Exp Med , vol.209 , pp. 259-273
    • Weigert, O.1    Lane, A.A.2    Bird, L.3
  • 125
    • 84861352086 scopus 로고    scopus 로고
    • Hedgehog pathway as a drug target: smoothened inhibitors in development
    • Lin T.L., Matsui W. Hedgehog pathway as a drug target: smoothened inhibitors in development. OncoTargets Ther 2012, 5:47-58.
    • (2012) OncoTargets Ther , vol.5 , pp. 47-58
    • Lin, T.L.1    Matsui, W.2
  • 126
    • 80052869447 scopus 로고    scopus 로고
    • Hedgehog signaling: networking to nurture a promalignant tumor microenvironment
    • Harris L.G., Samant R.S., Shevde L.A. Hedgehog signaling: networking to nurture a promalignant tumor microenvironment. Mol Cancer Res 2011, 9:1165-1174.
    • (2011) Mol Cancer Res , vol.9 , pp. 1165-1174
    • Harris, L.G.1    Samant, R.S.2    Shevde, L.A.3
  • 127
    • 84858009739 scopus 로고    scopus 로고
    • Targeting hedgehog in hematologic malignancy
    • Irvine D.A., Copland M. Targeting hedgehog in hematologic malignancy. Blood 2012, 119:2196-2204.
    • (2012) Blood , vol.119 , pp. 2196-2204
    • Irvine, D.A.1    Copland, M.2
  • 128
    • 84875004153 scopus 로고    scopus 로고
    • Phase I dose-escalation study of PF-04449913, an oral Hedgehog (Hh) inhibitor, in patients with select hematologic malignancies
    • [abstract: 424]
    • Jamieson C., Cortes J.E., Oehler V., et al. Phase I dose-escalation study of PF-04449913, an oral Hedgehog (Hh) inhibitor, in patients with select hematologic malignancies. Blood (ASH Annual Meeting Abstracts) 2011, 118. [abstract: 424].
    • (2011) Blood (ASH Annual Meeting Abstracts) , vol.118
    • Jamieson, C.1    Cortes, J.E.2    Oehler, V.3
  • 129
    • 62849106337 scopus 로고    scopus 로고
    • Beta-catenin hits chromatin: regulation of Wnt target gene activation
    • Mosimann C., Hausmann G., Basler K. Beta-catenin hits chromatin: regulation of Wnt target gene activation. Nat Rev Mol Cell Biol 2009, 10:276-286.
    • (2009) Nat Rev Mol Cell Biol , vol.10 , pp. 276-286
    • Mosimann, C.1    Hausmann, G.2    Basler, K.3
  • 130
    • 70350023790 scopus 로고    scopus 로고
    • Targeting the WNT/beta-catenin/TCF/LEF1 axis in solid and haematological cancers: multiplicity of therapeutic options
    • Gehrke I., Gandhirajan R.K., Kreuzer K.A. Targeting the WNT/beta-catenin/TCF/LEF1 axis in solid and haematological cancers: multiplicity of therapeutic options. Eur J Cancer 2009, 45:2759-2767.
    • (2009) Eur J Cancer , vol.45 , pp. 2759-2767
    • Gehrke, I.1    Gandhirajan, R.K.2    Kreuzer, K.A.3
  • 131
    • 79957989939 scopus 로고    scopus 로고
    • Drug discovery approaches to target Wnt signaling in cancer stem cells
    • Curtin J.C., Lorenzi M.V. Drug discovery approaches to target Wnt signaling in cancer stem cells. Oncotarget 2010, 1:552-566.
    • (2010) Oncotarget , vol.1 , pp. 552-566
    • Curtin, J.C.1    Lorenzi, M.V.2
  • 132
    • 77957252252 scopus 로고    scopus 로고
    • Blockade of JAK2 activity suppressed accumulation of β-catenin in leukemic cells
    • Liu Y.C., Lai W.C., Chuang K.A., et al. Blockade of JAK2 activity suppressed accumulation of β-catenin in leukemic cells. J Cell Biochem 2010, 111:402-411.
    • (2010) J Cell Biochem , vol.111 , pp. 402-411
    • Liu, Y.C.1    Lai, W.C.2    Chuang, K.A.3
  • 133
    • 84875006156 scopus 로고    scopus 로고
    • Treatment with β-catenin antagonist BC2059 exerts single agent efficacy and exerts improved activity with tyrosine kinase inhibitor (TKI) or pan-histone deacetylase (HDAC) inhibitor against human CML and myeloproliferative neoplasm (MPN) progenitor cells
    • [abstract: 65]
    • Fiskus W., Smith J., Mudunuru U., et al. Treatment with β-catenin antagonist BC2059 exerts single agent efficacy and exerts improved activity with tyrosine kinase inhibitor (TKI) or pan-histone deacetylase (HDAC) inhibitor against human CML and myeloproliferative neoplasm (MPN) progenitor cells. Blood (ASH Annual Meeting Abstracts) 2011, 118. [abstract: 65].
    • (2011) Blood (ASH Annual Meeting Abstracts) , vol.118
    • Fiskus, W.1    Smith, J.2    Mudunuru, U.3
  • 134
    • 84859986297 scopus 로고    scopus 로고
    • Allogeneic stem cell transplantation for myelofibrosis in 2012
    • McLornan D.P., Mead A.J., Jackson G., et al. Allogeneic stem cell transplantation for myelofibrosis in 2012. Br J Haematol 2012, 157:413-425.
    • (2012) Br J Haematol , vol.157 , pp. 413-425
    • McLornan, D.P.1    Mead, A.J.2    Jackson, G.3
  • 135
    • 81355147194 scopus 로고    scopus 로고
    • Primary myelofibrosis: 2012 update on diagnosis, risk stratification, and management
    • Tefferi A. Primary myelofibrosis: 2012 update on diagnosis, risk stratification, and management. Am J Hematol 2011, 86:1017-1026.
    • (2011) Am J Hematol , vol.86 , pp. 1017-1026
    • Tefferi, A.1
  • 137
    • 77956302639 scopus 로고    scopus 로고
    • Impact of JAK2V617F mutation status, allele burden, and clearance after allogeneic stem cell transplantation for myelofibrosis
    • Alchalby H., Badbaran A., Zabelina T., et al. Impact of JAK2V617F mutation status, allele burden, and clearance after allogeneic stem cell transplantation for myelofibrosis. Blood 2010, 116:3572-3581.
    • (2010) Blood , vol.116 , pp. 3572-3581
    • Alchalby, H.1    Badbaran, A.2    Zabelina, T.3


* 이 정보는 Elsevier사의 SCOPUS DB에서 KISTI가 분석하여 추출한 것입니다.