메뉴 건너뛰기




Volumn 14, Issue 9, 2017, Pages

Oral tetrahydrouridine and decitabine for non-cytotoxic epigenetic gene regulation in sickle cell disease: A randomized phase 1 study

(24)  Molokie, Robert a,b   Lavelle, Donald a,b   Gowhari, Michel a   Pacini, Michael a   Krauz, Lani a   Hassan, Johara a   Ibanez, Vinzon b   Ruiz, Maria A b   Ng, Kwok Peng c   Woost, Philip d   Radivoyevitch, Tomas e   Pacelli, Daisy a   Fada, Sherry c   Rump, Matthew c   Hsieh, Matthew f   Tisdale, John F f   Jacobberger, James d   Phelps, Mitch g   Engel, James Douglas h   Saraf, Santhosh a   more..


Author keywords

[No Author keywords available]

Indexed keywords

BILIRUBIN; C REACTIVE PROTEIN; CYTIDINE DEAMINASE; D DIMER; DECITABINE; DNA METHYLTRANSFERASE 1; HEMOGLOBIN F; LACTATE DEHYDROGENASE; PLACEBO; TETRAHYDROURIDINE; AZACITIDINE; ENZYME INHIBITOR;

EID: 85029668221     PISSN: 15491277     EISSN: 15491676     Source Type: Journal    
DOI: 10.1371/journal.pmed.1002382     Document Type: Article
Times cited : (104)

References (119)
  • 1
    • 0028291736 scopus 로고
    • Mortality in sickle cell disease. Life expectancy and risk factors for early death
    • 799340
    • Platt OS, Brambilla DJ, Rosse WF, Milner PF, Castro O, Steinberg MH, et al. (1994) Mortality in sickle cell disease. Life expectancy and risk factors for early death. N Engl J Med330: 1639–1644. doi: 10.1056/NEJM199406093302303 7993409
    • (1994) N Engl J Med , vol.330 , pp. 1639-1644
    • Platt, O.S.1    Brambilla, D.J.2    Rosse, W.F.3    Milner, P.F.4    Castro, O.5    Steinberg, M.H.6
  • 2
    • 84876435847 scopus 로고    scopus 로고
    • Mortality rates and age at death from sickle cell disease: U.S., 1979–2005
    • 2345087
    • Lanzkron S, Carroll CP, Haywood C, Jr. (2013) Mortality rates and age at death from sickle cell disease: U.S., 1979–2005. Public Health Reports128: 110–116. doi: 10.1177/003335491312800206 23450875
    • (2013) Public Health Reports , vol.128 , pp. 110-116
    • Lanzkron, S.1    Carroll, C.P.2    Haywood, C.3
  • 3
    • 0017641722 scopus 로고
    • Participation of hemoglobins A and F in polymerization of sickle hemoglobin
    • 1690
    • Goldberg MA, Husson MA, Bunn HF, (1977) Participation of hemoglobins A and F in polymerization of sickle hemoglobin. J Biol Chem252: 3414–3421. 16902
    • (1977) J Biol Chem , vol.252 , pp. 3414-3421
    • Goldberg, M.A.1    Husson, M.A.2    Bunn, H.F.3
  • 4
    • 0006936094 scopus 로고
    • Structural bases of the inhibitory effects of hemoglobin F and hemoglobin A2 on the polymerization of hemoglobin S
    • 28439
    • Nagel RL, Bookchin RM, Johnson J, Labie D, Wajcman H, Isaac-Sodeye WA, et al. (1979) Structural bases of the inhibitory effects of hemoglobin F and hemoglobin A2 on the polymerization of hemoglobin S. Proc Natl Acad Sci U S A76: 670–672. 284392
    • (1979) Proc Natl Acad Sci U S A , vol.76 , pp. 670-672
    • Nagel, R.L.1    Bookchin, R.M.2    Johnson, J.3    Labie, D.4    Wajcman, H.5    Isaac-Sodeye, W.A.6
  • 7
    • 0021343093 scopus 로고
    • Is there a threshold level of fetal hemoglobin that ameliorates morbidity in sickle cell anemia?
    • 620016
    • Powars DR, Weiss JN, Chan LS, Schroeder WA, (1984) Is there a threshold level of fetal hemoglobin that ameliorates morbidity in sickle cell anemia?Blood63: 921–926. 6200161
    • (1984) Blood , vol.63 , pp. 921-926
    • Powars, D.R.1    Weiss, J.N.2    Chan, L.S.3    Schroeder, W.A.4
  • 8
    • 0037414164 scopus 로고    scopus 로고
    • Effect of hydroxyurea on mortality and morbidity in adult sickle cell anemia: risks and benefits up to 9 years of treatment
    • 1267273
    • Steinberg MH, Barton F, Castro O, Pegelow CH, Ballas SK, Kutlar A, et al. (2003) Effect of hydroxyurea on mortality and morbidity in adult sickle cell anemia: risks and benefits up to 9 years of treatment. JAMA289: 1645–1651. doi: 10.1001/jama.289.13.1645 12672732
    • (2003) JAMA , vol.289 , pp. 1645-1651
    • Steinberg, M.H.1    Barton, F.2    Castro, O.3    Pegelow, C.H.4    Ballas, S.K.5    Kutlar, A.6
  • 9
    • 0035094512 scopus 로고    scopus 로고
    • Pharmacologic induction of fetal hemoglobin: raising the therapeutic bar in sickle cell disease
    • 1122468
    • Atweh GF, Schechter AN, (2001) Pharmacologic induction of fetal hemoglobin: raising the therapeutic bar in sickle cell disease. Curr Opin Hematol8: 123–130. 11224687
    • (2001) Curr Opin Hematol , vol.8 , pp. 123-130
    • Atweh, G.F.1    Schechter, A.N.2
  • 10
    • 0025939624 scopus 로고
    • Chronic renal failure in sickle cell disease: risk factors, clinical course, and mortality
    • 189233
    • Powars DR, Elliott-Mills DD, Chan L, Niland J, Hiti AL, Opas LM, et al. (1991) Chronic renal failure in sickle cell disease: risk factors, clinical course, and mortality. Ann Intern Med115: 614–620. 1892333
    • (1991) Ann Intern Med , vol.115 , pp. 614-620
    • Powars, D.R.1    Elliott-Mills, D.D.2    Chan, L.3    Niland, J.4    Hiti, A.L.5    Opas, L.M.6
  • 11
    • 79955127685 scopus 로고    scopus 로고
    • Protective role of hemoglobin and fetal hemoglobin in early kidney disease for children with sickle cell anemia
    • 2152380
    • Lebensburger J, Johnson SM, Askenazi DJ, Rozario NL, Howard TH, Hilliard LM, (2011) Protective role of hemoglobin and fetal hemoglobin in early kidney disease for children with sickle cell anemia. Am J Hematol86: 430–432. doi: 10.1002/ajh.21994 21523807
    • (2011) Am J Hematol , vol.86 , pp. 430-432
    • Lebensburger, J.1    Johnson, S.M.2    Askenazi, D.J.3    Rozario, N.L.4    Howard, T.H.5    Hilliard, L.M.6
  • 12
    • 85028190226 scopus 로고    scopus 로고
    • Severe anemia early in life as a risk factor for sickle-cell kidney disease
    • 2791990
    • Aban I, Baddam S, Hilliard LM, Howard TH, Feig DI, Lebensburger JD, (2017) Severe anemia early in life as a risk factor for sickle-cell kidney disease. Blood129: 385–387. doi: 10.1182/blood-2016-09-738104 27919909
    • (2017) Blood , vol.129 , pp. 385-387
    • Aban, I.1    Baddam, S.2    Hilliard, L.M.3    Howard, T.H.4    Feig, D.I.5    Lebensburger, J.D.6
  • 13
    • 0001700270 scopus 로고
    • Hereditary persistence of fetal hemoglobin: a study of 79 affected persons in 15 Negro families in Baltimore
    • 1402258
    • Conley CL, Weatherall DJ, Richardson SN, Shepard MK, Charache S, (1963) Hereditary persistence of fetal hemoglobin: a study of 79 affected persons in 15 Negro families in Baltimore. Blood21: 261–281. 14022587
    • (1963) Blood , vol.21 , pp. 261-281
    • Conley, C.L.1    Weatherall, D.J.2    Richardson, S.N.3    Shepard, M.K.4    Charache, S.5
  • 14
    • 0017880649 scopus 로고
    • Natural history of sickle cell anemia in Saudi Arabs. A study of 270 subjects
    • 61973
    • Perrine RP, Pembrey ME, John P, Perrine S, Shoup F, (1978) Natural history of sickle cell anemia in Saudi Arabs. A study of 270 subjects. Ann Intern Med88: 1–6. 619731
    • (1978) Ann Intern Med , vol.88 , pp. 1-6
    • Perrine, R.P.1    Pembrey, M.E.2    John, P.3    Perrine, S.4    Shoup, F.5
  • 15
    • 84155186479 scopus 로고    scopus 로고
    • Fetal haemoglobin levels and haematological characteristics of compound heterozygotes for haemoglobin S and deletional hereditary persistence of fetal haemoglobin
    • 2201764
    • Ngo DA, Aygun B, Akinsheye I, Hankins JS, Bhan I, Luo HY, et al. (2012) Fetal haemoglobin levels and haematological characteristics of compound heterozygotes for haemoglobin S and deletional hereditary persistence of fetal haemoglobin. Br J Haematol156: 259–264. doi: 10.1111/j.1365-2141.2011.08916.x 22017641
    • (2012) Br J Haematol , vol.156 , pp. 259-264
    • Ngo, D.A.1    Aygun, B.2    Akinsheye, I.3    Hankins, J.S.4    Bhan, I.5    Luo, H.Y.6
  • 16
    • 0018098588 scopus 로고
    • Stimulation of fetal hemoglobin synthesis in baboons by hemolysis and hypoxia
    • 9644
    • DeSimone J, Biel SI, Heller P, (1978) Stimulation of fetal hemoglobin synthesis in baboons by hemolysis and hypoxia. Proc Natl Acad Sci U S A75: 2937–2940. 96444
    • (1978) Proc Natl Acad Sci U S A , vol.75 , pp. 2937-2940
    • DeSimone, J.1    Biel, S.I.2    Heller, P.3
  • 17
    • 0018638697 scopus 로고
    • Hemopoietic stress and fetal hemoglobin synthesis: comparative studies in vivo and in vitro
    • 11551
    • DeSimone J, Heller P, Adams JG, (1979) Hemopoietic stress and fetal hemoglobin synthesis: comparative studies in vivo and in vitro. Blood54: 1176–1181. 115514
    • (1979) Blood , vol.54 , pp. 1176-1181
    • DeSimone, J.1    Heller, P.2    Adams, J.G.3
  • 18
    • 0018921139 scopus 로고
    • Magnitude of the fetal hemoglobin response to acute hemolytic anemia in baboons is controlled by genetic factors
    • 676595
    • DeSimone J, Heller P, Amsel J, Usman M, (1980) Magnitude of the fetal hemoglobin response to acute hemolytic anemia in baboons is controlled by genetic factors. J Clin Invest65: 224–226. doi: 10.1172/JCI109654 6765958
    • (1980) J Clin Invest , vol.65 , pp. 224-226
    • DeSimone, J.1    Heller, P.2    Amsel, J.3    Usman, M.4
  • 19
    • 0019512161 scopus 로고
    • Genetic relationship between fetal Hb levels in normal and erythropoietically stressed baboons
    • 617030
    • DeSimone J, Heller P, Biel M, Zwiers D, (1981) Genetic relationship between fetal Hb levels in normal and erythropoietically stressed baboons. Br J Haematol49: 175–183. 6170305
    • (1981) Br J Haematol , vol.49 , pp. 175-183
    • DeSimone, J.1    Heller, P.2    Biel, M.3    Zwiers, D.4
  • 20
    • 0019960147 scopus 로고
    • Maintenance of fetal hemoglobin (HbF) elevations in the baboon by prolonged erythropoietic stress
    • 617845
    • DeSimone J, Biel M, Heller P, (1982) Maintenance of fetal hemoglobin (HbF) elevations in the baboon by prolonged erythropoietic stress. Blood60: 519–523. 6178456
    • (1982) Blood , vol.60 , pp. 519-523
    • DeSimone, J.1    Biel, M.2    Heller, P.3
  • 21
    • 0022619760 scopus 로고
    • On the mechanism of Hb F elevations in the baboon by erythropoietic stress and pharmacologic manipulation
    • 242039
    • Lavelle D, DeSimone J, Heller P, Zwiers D, Hall L, (1986) On the mechanism of Hb F elevations in the baboon by erythropoietic stress and pharmacologic manipulation. Blood67: 1083–1089. 2420395
    • (1986) Blood , vol.67 , pp. 1083-1089
    • Lavelle, D.1    DeSimone, J.2    Heller, P.3    Zwiers, D.4    Hall, L.5
  • 22
    • 0029025475 scopus 로고
    • Effect of hydroxyurea on the frequency of painful crises in sickle cell anemia. Investigators of the Multicenter Study of Hydroxyurea in Sickle Cell Anemia
    • 771563
    • Charache S, Terrin ML, Moore RD, Dover GJ, Barton FB, Eckert SV, et al. (1995) Effect of hydroxyurea on the frequency of painful crises in sickle cell anemia. Investigators of the Multicenter Study of Hydroxyurea in Sickle Cell Anemia. N Engl J Med332: 1317–1322. doi: 10.1056/NEJM199505183322001 7715639
    • (1995) N Engl J Med , vol.332 , pp. 1317-1322
    • Charache, S.1    Terrin, M.L.2    Moore, R.D.3    Dover, G.J.4    Barton, F.B.5    Eckert, S.V.6
  • 23
    • 0030893396 scopus 로고    scopus 로고
    • Fetal hemoglobin in sickle cell anemia: determinants of response to hydroxyurea. Multicenter Study of Hydroxyurea
    • 902834
    • Steinberg MH, Lu ZH, Barton FB, Terrin ML, Charache S, Dover GJ, (1997) Fetal hemoglobin in sickle cell anemia: determinants of response to hydroxyurea. Multicenter Study of Hydroxyurea. Blood89: 1078–1088. 9028341
    • (1997) Blood , vol.89 , pp. 1078-1088
    • Steinberg, M.H.1    Lu, Z.H.2    Barton, F.B.3    Terrin, M.L.4    Charache, S.5    Dover, G.J.6
  • 24
    • 0032946891 scopus 로고    scopus 로고
    • Erythropoietic activity in patients with sickle cell anaemia before and after treatment with hydroxyurea
    • 1023342
    • Ballas SK, Marcolina MJ, Dover GJ, Barton FB, (1999) Erythropoietic activity in patients with sickle cell anaemia before and after treatment with hydroxyurea. Br J Haematol105: 491–496. 10233426
    • (1999) Br J Haematol , vol.105 , pp. 491-496
    • Ballas, S.K.1    Marcolina, M.J.2    Dover, G.J.3    Barton, F.B.4
  • 25
    • 0026631318 scopus 로고
    • Hydroxyurea: effects on hemoglobin F production in patients with sickle cell anemia
    • 137510
    • Charache S, Dover GJ, Moore RD, Eckert S, Ballas SK, Koshy M, et al. (1992) Hydroxyurea: effects on hemoglobin F production in patients with sickle cell anemia. Blood79: 2555–2565. 1375104
    • (1992) Blood , vol.79 , pp. 2555-2565
    • Charache, S.1    Dover, G.J.2    Moore, R.D.3    Eckert, S.4    Ballas, S.K.5    Koshy, M.6
  • 28
    • 33746040474 scopus 로고    scopus 로고
    • N-terminal pro-brain natriuretic peptide levels and risk of death in sickle cell disease
    • 1684966
    • Machado RF, Anthi A, Steinberg MH, Bonds D, Sachdev V, Kato GJ, et al. (2006) N-terminal pro-brain natriuretic peptide levels and risk of death in sickle cell disease. JAMA296: 310–318. doi: 10.1001/jama.296.3.310 16849664
    • (2006) JAMA , vol.296 , pp. 310-318
    • Machado, R.F.1    Anthi, A.2    Steinberg, M.H.3    Bonds, D.4    Sachdev, V.5    Kato, G.J.6
  • 29
    • 0036096085 scopus 로고    scopus 로고
    • Predictors of fetal hemoglobin response in children with sickle cell anemia receiving hydroxyurea therapy
    • 1175614
    • Ware RE, Eggleston B, Redding-Lallinger R, Wang WC, Smith-Whitley K, Daeschner C, et al. (2002) Predictors of fetal hemoglobin response in children with sickle cell anemia receiving hydroxyurea therapy. Blood99: 10–14. 11756146
    • (2002) Blood , vol.99 , pp. 10-14
    • Ware, R.E.1    Eggleston, B.2    Redding-Lallinger, R.3    Wang, W.C.4    Smith-Whitley, K.5    Daeschner, C.6
  • 30
    • 84992029060 scopus 로고    scopus 로고
    • Decreased fetal hemoglobin over time among youth with sickle cell disease on hydroxyurea is associated with higher urgent hospital use
    • Green NS, Manwani D, Qureshi M, Ireland K, Sinha A, Smaldone AM, (2016) Decreased fetal hemoglobin over time among youth with sickle cell disease on hydroxyurea is associated with higher urgent hospital use. Pediatric Blood & Cancer63: 2146–2153.
    • (2016) Pediatric Blood & Cancer , vol.63 , pp. 2146-2153
    • Green, N.S.1    Manwani, D.2    Qureshi, M.3    Ireland, K.4    Sinha, A.5    Smaldone, A.M.6
  • 31
    • 0026767491 scopus 로고
    • Laboratory profile of sickle cell disease: a cross-sectional analysis. The Cooperative Study of Sickle Cell Disease
    • 162497
    • West MS, Wethers D, Smith J, Steinberg M, (1992) Laboratory profile of sickle cell disease: a cross-sectional analysis. The Cooperative Study of Sickle Cell Disease. J Clin Epidemiol45: 893–909. 1624972
    • (1992) J Clin Epidemiol , vol.45 , pp. 893-909
    • West, M.S.1    Wethers, D.2    Smith, J.3    Steinberg, M.4
  • 32
    • 79954419129 scopus 로고    scopus 로고
    • Standard clinical practice underestimates the role and significance of erythropoietin deficiency in sickle cell disease
    • 2141817
    • Saraf S, Farooqui M, Infusino G, Oza B, Sidhwani S, Gowhari M, et al. (2011) Standard clinical practice underestimates the role and significance of erythropoietin deficiency in sickle cell disease. Br J Haematol153: 386–392. doi: 10.1111/j.1365-2141.2010.08479.x 21418176
    • (2011) Br J Haematol , vol.153 , pp. 386-392
    • Saraf, S.1    Farooqui, M.2    Infusino, G.3    Oza, B.4    Sidhwani, S.5    Gowhari, M.6
  • 33
    • 0018905582 scopus 로고
    • DNA methylation in the human gamma delta beta-globin locus in erythroid and nonerythroid tissues
    • 624707
    • van der Ploeg LH, Flavell RA, (1980) DNA methylation in the human gamma delta beta-globin locus in erythroid and nonerythroid tissues. Cell19: 947–958. 6247075
    • (1980) Cell , vol.19 , pp. 947-958
    • van der Ploeg, L.H.1    Flavell, R.A.2
  • 34
    • 0021034956 scopus 로고
    • Molecular mechanisms of human hemoglobin switching: selective undermethylation and expression of globin genes in embryonic, fetal, and adult erythroblasts
    • 631633
    • Mavilio F, Giampaolo A, Care A, Migliaccio G, Calandrini M, Russo G, et al. (1983) Molecular mechanisms of human hemoglobin switching: selective undermethylation and expression of globin genes in embryonic, fetal, and adult erythroblasts. Proc Natl Acad Sci U S A80: 6907–6911. 6316333
    • (1983) Proc Natl Acad Sci U S A , vol.80 , pp. 6907-6911
    • Mavilio, F.1    Giampaolo, A.2    Care, A.3    Migliaccio, G.4    Calandrini, M.5    Russo, G.6
  • 35
    • 0023821768 scopus 로고
    • Embryonic epsilon and gamma globin genes of a prosimian primate (Galago crassicaudatus). Nucleotide and amino acid sequences, developmental regulation and phylogenetic footprints
    • 319944
    • Tagle DA, Koop BF, Goodman M, Slightom JL, Hess DL, Jones RT, (1988) Embryonic epsilon and gamma globin genes of a prosimian primate (Galago crassicaudatus). Nucleotide and amino acid sequences, developmental regulation and phylogenetic footprints. J Mol Biol203: 439–455. 3199442
    • (1988) J Mol Biol , vol.203 , pp. 439-455
    • Tagle, D.A.1    Koop, B.F.2    Goodman, M.3    Slightom, J.L.4    Hess, D.L.5    Jones, R.T.6
  • 36
    • 0000206049 scopus 로고
    • 5-Azacytidine stimulates fetal hemoglobin synthesis in anemic baboons
    • 618150
    • DeSimone J, Heller P, Hall L, Zwiers D, (1982) 5-Azacytidine stimulates fetal hemoglobin synthesis in anemic baboons. Proc Natl Acad Sci U S A79: 4428–4431. 6181507
    • (1982) Proc Natl Acad Sci U S A , vol.79 , pp. 4428-4431
    • DeSimone, J.1    Heller, P.2    Hall, L.3    Zwiers, D.4
  • 37
    • 0020466344 scopus 로고
    • 5-azacytidine selectively increases gamma-globin synthesis in a patient with beta+ thalassemia
    • 618358
    • Ley TJ, DeSimone J, Anagnou NP, Keller GH, Humphries RK, Turner PH, et al. (1982) 5-azacytidine selectively increases gamma-globin synthesis in a patient with beta+ thalassemia. N Engl J Med307: 1469–1475. doi: 10.1056/NEJM198212093072401 6183586
    • (1982) N Engl J Med , vol.307 , pp. 1469-1475
    • Ley, T.J.1    DeSimone, J.2    Anagnou, N.P.3    Keller, G.H.4    Humphries, R.K.5    Turner, P.H.6
  • 38
    • 0020563258 scopus 로고
    • 5-Azacytidine increases gamma-globin synthesis and reduces the proportion of dense cells in patients with sickle cell anemia
    • 619179
    • Ley TJ, DeSimone J, Noguchi CT, Turner PH, Schechter AN, Heller P, et al. (1983) 5-Azacytidine increases gamma-globin synthesis and reduces the proportion of dense cells in patients with sickle cell anemia. Blood62: 370–380. 6191799
    • (1983) Blood , vol.62 , pp. 370-380
    • Ley, T.J.1    DeSimone, J.2    Noguchi, C.T.3    Turner, P.H.4    Schechter, A.N.5    Heller, P.6
  • 39
    • 0004959517 scopus 로고
    • Treatment of sickle cell anemia with 5-azacytidine results in increased fetal hemoglobin production and is associated with nonrandom hypomethylation of DNA around the gamma-delta-beta-globin gene complex
    • 619244
    • Charache S, Dover G, Smith K, Talbot CC, Jr.Moyer M, Boyer S, (1983) Treatment of sickle cell anemia with 5-azacytidine results in increased fetal hemoglobin production and is associated with nonrandom hypomethylation of DNA around the gamma-delta-beta-globin gene complex. Proc Natl Acad Sci U S A80: 4842–4846. 6192443
    • (1983) Proc Natl Acad Sci U S A , vol.80 , pp. 4842-4846
    • Charache, S.1    Dover, G.2    Smith, K.3    Talbot, C.C.4    Moyer, M.5    Boyer, S.6
  • 40
    • 0021927659 scopus 로고
    • 5-Azacytidine increases HbF production and reduces anemia in sickle cell disease: dose-response analysis of subcutaneous and oral dosage regimens
    • 241131
    • Dover GJ, Charache S, Boyer SH, Vogelsang G, Moyer M, (1985) 5-Azacytidine increases HbF production and reduces anemia in sickle cell disease: dose-response analysis of subcutaneous and oral dosage regimens. Blood66: 527–532. 2411310
    • (1985) Blood , vol.66 , pp. 527-532
    • Dover, G.J.1    Charache, S.2    Boyer, S.H.3    Vogelsang, G.4    Moyer, M.5
  • 41
    • 0027274649 scopus 로고
    • Brief report: treatment with azacitidine of patients with end-stage beta-thalassemia
    • 768917
    • Lowrey CH, Nienhuis AW, (1993) Brief report: treatment with azacitidine of patients with end-stage beta-thalassemia. N Engl J Med329: 845–848. doi: 10.1056/NEJM199309163291205 7689171
    • (1993) N Engl J Med , vol.329 , pp. 845-848
    • Lowrey, C.H.1    Nienhuis, A.W.2
  • 42
    • 0005809081 scopus 로고    scopus 로고
    • Augmentation of fetal hemoglobin (HbF) levels by low-dose short-duration 5 '-aza-2-deoxycytidine (decitabine) administration in sickle cell anemia patients who had no HbF elevation following hydroxyurea therapy
    • Koshy M, DeSimone J, Molokie R, Dorn L, van der Galien T, Bressler L, (1998) Augmentation of fetal hemoglobin (HbF) levels by low-dose short-duration 5 '-aza-2-deoxycytidine (decitabine) administration in sickle cell anemia patients who had no HbF elevation following hydroxyurea therapy. Blood92: 30b–30b.
    • (1998) Blood , vol.92 , pp. 30b
    • Koshy, M.1    DeSimone, J.2    Molokie, R.3    Dorn, L.4    van der Galien, T.5    Bressler, L.6
  • 43
    • 10744231450 scopus 로고    scopus 로고
    • Effects of 5-aza-2 '-deoxycytidine on fetal hemoglobin levels, red cell adhesion, and hematopoietic differentiation in patients with sickle cell disease
    • 1290744
    • Saunthararajah Y, Hillery CA, Lavelle D, Molokie R, Dorn L, Bressler L, et al. (2003) Effects of 5-aza-2 '-deoxycytidine on fetal hemoglobin levels, red cell adhesion, and hematopoietic differentiation in patients with sickle cell disease. Blood102: 3865–3870. doi: 10.1182/blood-2003-05-1738 12907443
    • (2003) Blood , vol.102 , pp. 3865-3870
    • Saunthararajah, Y.1    Hillery, C.A.2    Lavelle, D.3    Molokie, R.4    Dorn, L.5    Bressler, L.6
  • 44
    • 84876239967 scopus 로고    scopus 로고
    • Corepressor-dependent silencing of fetal hemoglobin expression by BCL11A
    • 2357675
    • Xu J, Bauer DE, Kerenyi MA, Vo TD, Hou S, Hsu YJ, et al. (2013) Corepressor-dependent silencing of fetal hemoglobin expression by BCL11A. Proc Natl Acad Sci U S A110: 6518–6523. doi: 10.1073/pnas.1303976110 23576758
    • (2013) Proc Natl Acad Sci U S A , vol.110 , pp. 6518-6523
    • Xu, J.1    Bauer, D.E.2    Kerenyi, M.A.3    Vo, T.D.4    Hou, S.5    Hsu, Y.J.6
  • 45
    • 79961145533 scopus 로고    scopus 로고
    • Nuclear receptors TR2 and TR4 recruit multiple epigenetic transcriptional corepressors that associate specifically with the embryonic beta-type globin promoters in differentiated adult erythroid cells
    • 2167014
    • Cui S, Kolodziej KE, Obara N, Amaral-Psarris A, Demmers J, Shi L, et al. (2011) Nuclear receptors TR2 and TR4 recruit multiple epigenetic transcriptional corepressors that associate specifically with the embryonic beta-type globin promoters in differentiated adult erythroid cells. Mol Cell Biol31: 3298–3311. doi: 10.1128/MCB.05310-11 21670149
    • (2011) Mol Cell Biol , vol.31 , pp. 3298-3311
    • Cui, S.1    Kolodziej, K.E.2    Obara, N.3    Amaral-Psarris, A.4    Demmers, J.5    Shi, L.6
  • 46
    • 0020582853 scopus 로고
    • On the mechanism of inhibition of DNA-cytosine methyltransferases by cytosine analogs
    • 620576
    • Santi DV, Garrett CE, Barr PJ, (1983) On the mechanism of inhibition of DNA-cytosine methyltransferases by cytosine analogs. Cell33: 9–10. 6205762
    • (1983) Cell , vol.33 , pp. 9-10
    • Santi, D.V.1    Garrett, C.E.2    Barr, P.J.3
  • 47
    • 0023035969 scopus 로고
    • Differences in DNA damage produced by incorporation of 5-aza-2'-deoxycytidine or 5,6-dihydro-5-azacytidine into DNA of mammalian cells
    • 242847
    • Covey JM, D'Incalci M, Tilchen EJ, Zaharko DS, Kohn KW, (1986) Differences in DNA damage produced by incorporation of 5-aza-2'-deoxycytidine or 5,6-dihydro-5-azacytidine into DNA of mammalian cells. Cancer Res46: 5511–5517. 2428479
    • (1986) Cancer Res , vol.46 , pp. 5511-5517
    • Covey, J.M.1    D'Incalci, M.2    Tilchen, E.J.3    Zaharko, D.S.4    Kohn, K.W.5
  • 48
    • 34547842515 scopus 로고    scopus 로고
    • Dynamics of Dnmt1 interaction with the replication machinery and its role in postreplicative maintenance of DNA methylation
    • 1757669
    • Schermelleh L, Haemmer A, Spada F, Rosing N, Meilinger D, Rothbauer U, et al. (2007) Dynamics of Dnmt1 interaction with the replication machinery and its role in postreplicative maintenance of DNA methylation. Nucleic Acids Res35: 4301–4312. doi: 10.1093/nar/gkm432 17576694
    • (2007) Nucleic Acids Res , vol.35 , pp. 4301-4312
    • Schermelleh, L.1    Haemmer, A.2    Spada, F.3    Rosing, N.4    Meilinger, D.5    Rothbauer, U.6
  • 49
    • 84938809941 scopus 로고    scopus 로고
    • CDA directs metabolism of epigenetic nucleosides revealing a therapeutic window in cancer
    • 2620033
    • Zauri M, Berridge G, Thezenas ML, Pugh KM, Goldin R, Kessler BM, et al. (2015) CDA directs metabolism of epigenetic nucleosides revealing a therapeutic window in cancer. Nature524: 114–118. doi: 10.1038/nature14948 26200337
    • (2015) Nature , vol.524 , pp. 114-118
    • Zauri, M.1    Berridge, G.2    Thezenas, M.L.3    Pugh, K.M.4    Goldin, R.5    Kessler, B.M.6
  • 50
    • 84962212092 scopus 로고    scopus 로고
    • CETSA screening identifies known and novel thymidylate synthase inhibitors and slow intracellular activation of 5-fluorouracil
    • 2701051
    • Almqvist H, Axelsson H, Jafari R, Dan C, Mateus A, Haraldsson M, et al. (2016) CETSA screening identifies known and novel thymidylate synthase inhibitors and slow intracellular activation of 5-fluorouracil. Nat Commun7: 11040. doi: 10.1038/ncomms11040 27010513
    • (2016) Nat Commun , vol.7 , pp. 11040
    • Almqvist, H.1    Axelsson, H.2    Jafari, R.3    Dan, C.4    Mateus, A.5    Haraldsson, M.6
  • 51
  • 52
    • 84924030193 scopus 로고    scopus 로고
    • Evaluation of noncytotoxic DNMT1-depleting therapy in patients with myelodysplastic syndromes
    • 2562149
    • Saunthararajah Y, Sekeres M, Advani A, Mahfouz R, Durkin L, Radivoyevitch T, et al. (2015) Evaluation of noncytotoxic DNMT1-depleting therapy in patients with myelodysplastic syndromes. J Clin Invest125: 1043–1055. doi: 10.1172/JCI78789 25621498
    • (2015) J Clin Invest , vol.125 , pp. 1043-1055
    • Saunthararajah, Y.1    Sekeres, M.2    Advani, A.3    Mahfouz, R.4    Durkin, L.5    Radivoyevitch, T.6
  • 53
    • 2542448184 scopus 로고    scopus 로고
    • Modification of hematopoietic stem cell fate by 5aza 2 ' deoxycytidine and trichostatin A
    • 1497603
    • Milhem M, Mahmud N, Lavelle D, Araki H, DeSimone J, Saunthararajah Y, et al. (2004) Modification of hematopoietic stem cell fate by 5aza 2 ' deoxycytidine and trichostatin A. Blood103: 4102–4110. doi: 10.1182/blood-2003-07-2431 14976039
    • (2004) Blood , vol.103 , pp. 4102-4110
    • Milhem, M.1    Mahmud, N.2    Lavelle, D.3    Araki, H.4    DeSimone, J.5    Saunthararajah, Y.6
  • 54
    • 77953365022 scopus 로고    scopus 로고
    • Decitabine maintains hematopoietic precursor self-renewal by preventing repression of stem cell genes by a differentiation-inducing stimulus
    • 2050180
    • Hu Z, Negrotto S, Gu X, Mahfouz R, Ng KP, Ebrahem Q, et al. (2010) Decitabine maintains hematopoietic precursor self-renewal by preventing repression of stem cell genes by a differentiation-inducing stimulus. Mol Cancer Ther9: 1536–1543. doi: 10.1158/1535-7163.MCT-10-0191 20501800
    • (2010) Mol Cancer Ther , vol.9 , pp. 1536-1543
    • Hu, Z.1    Negrotto, S.2    Gu, X.3    Mahfouz, R.4    Ng, K.P.5    Ebrahem, Q.6
  • 55
    • 0036625004 scopus 로고    scopus 로고
    • Maintenance of elevated fetal hemoglobin levels by decitabine during dose interval treatment of sickle cell anemia
    • 1201078
    • DeSimone J, Koshy M, Dorn L, Lavelle D, Bressier L, Molokie R, et al. (2002) Maintenance of elevated fetal hemoglobin levels by decitabine during dose interval treatment of sickle cell anemia. Blood99: 3905–3908. 12010787
    • (2002) Blood , vol.99 , pp. 3905-3908
    • DeSimone, J.1    Koshy, M.2    Dorn, L.3    Lavelle, D.4    Bressier, L.5    Molokie, R.6
  • 56
    • 33645521995 scopus 로고    scopus 로고
    • Characterization of decomposition products and preclinical and low dose clinical pharmacokinetics of decitabine (5-aza-2'-deoxycytidine) by a new liquid chromatography/tandem mass spectrometry quantification method
    • 1652352
    • Liu Z, Marcucci G, Byrd JC, Grever M, Xiao J, Chan KK, (2006) Characterization of decomposition products and preclinical and low dose clinical pharmacokinetics of decitabine (5-aza-2'-deoxycytidine) by a new liquid chromatography/tandem mass spectrometry quantification method. Rapid Commun Mass Spectrom20: 1117–1126. doi: 10.1002/rcm.2423 16523529
    • (2006) Rapid Commun Mass Spectrom , vol.20 , pp. 1117-1126
    • Liu, Z.1    Marcucci, G.2    Byrd, J.C.3    Grever, M.4    Xiao, J.5    Chan, K.K.6
  • 57
    • 34247094617 scopus 로고    scopus 로고
    • Characterization of in vitro and in vivo hypomethylating effects of decitabine in acute myeloid leukemia by a rapid, specific and sensitive LC-MS/MS method
    • 1726412
    • Liu Z, Liu S, Xie Z, Blum W, Perrotti D, Paschka P, et al. (2007) Characterization of in vitro and in vivo hypomethylating effects of decitabine in acute myeloid leukemia by a rapid, specific and sensitive LC-MS/MS method. Nucleic Acids Res35: e31. doi: 10.1093/nar/gkl1156 17264127
    • (2007) Nucleic Acids Res , vol.35 , pp. e31
    • Liu, Z.1    Liu, S.2    Xie, Z.3    Blum, W.4    Perrotti, D.5    Paschka, P.6
  • 58
    • 0013805757 scopus 로고
    • Studies of the enzymatic deamination of cytosine arabinoside. I. Enzyme distribution and species specificity
    • 495602
    • Camiener GW, Smith CG, (1965) Studies of the enzymatic deamination of cytosine arabinoside. I. Enzyme distribution and species specificity. Biochem Pharmacol14: 1405–1416. 4956026
    • (1965) Biochem Pharmacol , vol.14 , pp. 1405-1416
    • Camiener, G.W.1    Smith, C.G.2
  • 59
    • 0016696005 scopus 로고
    • Enhancement by tetrahydrouridine (NSC-112907) of the oral activity of 5-azacytidine (NSC-102816) in L1210 leukemic mice
    • 5421
    • Neil GL, Moxley TE, Kuentzel SL, Manak RC, Hanka LJ, (1975) Enhancement by tetrahydrouridine (NSC-112907) of the oral activity of 5-azacytidine (NSC-102816) in L1210 leukemic mice. Cancer Chemother Rep59: 459–465. 54211
    • (1975) Cancer Chemother Rep , vol.59 , pp. 459-465
    • Neil, G.L.1    Moxley, T.E.2    Kuentzel, S.L.3    Manak, R.C.4    Hanka, L.J.5
  • 60
  • 61
    • 50349090153 scopus 로고    scopus 로고
    • Modulation of gemcitabine (2',2'-difluoro-2'-deoxycytidine) pharmacokinetics, metabolism, and bioavailability in mice by 3,4,5,6-tetrahydrouridine
    • 1851978
    • Beumer JH, Eiseman JL, Parise RA, Joseph E, Covey JM, Egorin MJ, (2008) Modulation of gemcitabine (2',2'-difluoro-2'-deoxycytidine) pharmacokinetics, metabolism, and bioavailability in mice by 3,4,5,6-tetrahydrouridine. Clin Cancer Res14: 3529–3535. doi: 10.1158/1078-0432.CCR-07-4885 18519786
    • (2008) Clin Cancer Res , vol.14 , pp. 3529-3535
    • Beumer, J.H.1    Eiseman, J.L.2    Parise, R.A.3    Joseph, E.4    Covey, J.M.5    Egorin, M.J.6
  • 62
    • 84872796193 scopus 로고    scopus 로고
    • High cytidine deaminase expression in the liver provides sanctuary for cancer cells from decitabine treatment effects
    • 2308715
    • Ebrahem Q, Mahfouz R, Ng KP, Saunthararajah Y, (2012) High cytidine deaminase expression in the liver provides sanctuary for cancer cells from decitabine treatment effects. Oncotarget3: 1137–1145. doi: 10.18632/oncotarget.597 23087155
    • (2012) Oncotarget , vol.3 , pp. 1137-1145
    • Ebrahem, Q.1    Mahfouz, R.2    Ng, K.P.3    Saunthararajah, Y.4
  • 63
    • 0020053056 scopus 로고
    • Influence of tetrahydrouridine on the pharmacokinetics of intrathecally administered 1-beta-D-arabinofuranosylcytosine
    • 689601
    • Riccardi R, Chabner B, Glaubiger DL, Wood J, Poplack DG, (1982) Influence of tetrahydrouridine on the pharmacokinetics of intrathecally administered 1-beta-D-arabinofuranosylcytosine. Cancer Res42: 1736–1739. 6896013
    • (1982) Cancer Res , vol.42 , pp. 1736-1739
    • Riccardi, R.1    Chabner, B.2    Glaubiger, D.L.3    Wood, J.4    Poplack, D.G.5
  • 64
    • 0026409338 scopus 로고
    • Therapy of refractory/relapsed acute leukemia with cytosine arabinoside plus tetrahydrouridine (an inhibitor of cytidine deaminase)—a pilot study
    • 196104
    • Kreis W, Budman DR, Chan K, Allen SL, Schulman P, Lichtman S, et al. (1991) Therapy of refractory/relapsed acute leukemia with cytosine arabinoside plus tetrahydrouridine (an inhibitor of cytidine deaminase)—a pilot study. Leukemia5: 991–998. 1961042
    • (1991) Leukemia , vol.5 , pp. 991-998
    • Kreis, W.1    Budman, D.R.2    Chan, K.3    Allen, S.L.4    Schulman, P.5    Lichtman, S.6
  • 67
    • 0017729835 scopus 로고
    • Tetrahydrouridine: Physiologic disposition and effect upon deamination of cytosine arabinoside in man
    • 58960
    • Kreis W, Woodcock TM, Gordon CS, Krakoff IH, (1977) Tetrahydrouridine: Physiologic disposition and effect upon deamination of cytosine arabinoside in man. Cancer Treat Rep61: 1347–1353. 589600
    • (1977) Cancer Treat Rep , vol.61 , pp. 1347-1353
    • Kreis, W.1    Woodcock, T.M.2    Gordon, C.S.3    Krakoff, I.H.4
  • 68
    • 0027449966 scopus 로고
    • Therapy of refractory/relapsed acute myeloid leukemia and blast crisis of chronic myeloid leukemia with the combination of cytosine arabinoside, tetrahydrouridine, and carboplatin
    • 845368
    • Marsh JH, Kreis W, Barile B, Akerman S, Schulman P, Allen SL, et al. (1993) Therapy of refractory/relapsed acute myeloid leukemia and blast crisis of chronic myeloid leukemia with the combination of cytosine arabinoside, tetrahydrouridine, and carboplatin. Cancer Chemother Pharmacol31: 481–484. 8453688
    • (1993) Cancer Chemother Pharmacol , vol.31 , pp. 481-484
    • Marsh, J.H.1    Kreis, W.2    Barile, B.3    Akerman, S.4    Schulman, P.5    Allen, S.L.6
  • 69
    • 0016175119 scopus 로고
    • Preclinical toxicologic evaluation of tetrahydrouridine (NSC-112907) in beagle dogs and rhesus monkeys
    • Goldenthal EI, Cookson KM, Geil RG, Wazeter FX, (1974) Preclinical toxicologic evaluation of tetrahydrouridine (NSC-112907) in beagle dogs and rhesus monkeys. Cancer Chemother Rep35: 15–16.
    • (1974) Cancer Chemother Rep3 , vol.5 , pp. 15-16
    • Goldenthal, E.I.1    Cookson, K.M.2    Geil, R.G.3    Wazeter, F.X.4
  • 70
    • 84856606869 scopus 로고    scopus 로고
    • Effects of tetrahydrouridine on pharmacokinetics and pharmacodynamics of oral decitabine
    • 2216038
    • Lavelle D, Vaitkus K, Ling Y, Ruiz MA, Mahfouz R, Ng KP, et al. (2012) Effects of tetrahydrouridine on pharmacokinetics and pharmacodynamics of oral decitabine. Blood119: 1240–1247. doi: 10.1182/blood-2011-08-371690 22160381
    • (2012) Blood , vol.119 , pp. 1240-1247
    • Lavelle, D.1    Vaitkus, K.2    Ling, Y.3    Ruiz, M.A.4    Mahfouz, R.5    Ng, K.P.6
  • 71
    • 66449125866 scopus 로고    scopus 로고
    • LINE-1 methylation in plasma DNA as a biomarker of activity of DNA methylation inhibitors in patients with solid tumors
    • 1942100
    • Aparicio A, North B, Barske L, Wang X, Bollati V, Weisenberger D, et al. (2009) LINE-1 methylation in plasma DNA as a biomarker of activity of DNA methylation inhibitors in patients with solid tumors. Epigenetics4: 176–184. 19421002
    • (2009) Epigenetics , vol.4 , pp. 176-184
    • Aparicio, A.1    North, B.2    Barske, L.3    Wang, X.4    Bollati, V.5    Weisenberger, D.6
  • 72
    • 84924030193 scopus 로고    scopus 로고
    • Evaluation of noncytotoxic DNMT1-depleting therapy in patients with myelodysplastic syndromes
    • 2562149
    • Saunthararajah Y, Sekeres M, Advani A, Mahfouz R, Durkin L, Radivoyevitch T, et al. (2015) Evaluation of noncytotoxic DNMT1-depleting therapy in patients with myelodysplastic syndromes. J Clin Invest. 2;125(3):1043–55. doi: 10.1172/JCI78789 25621498
    • (2015) J Clin Invest , vol.125 , Issue.3 , pp. 1043-1055
    • Saunthararajah, Y.1    Sekeres, M.2    Advani, A.3    Mahfouz, R.4    Durkin, L.5    Radivoyevitch, T.6
  • 73
    • 0000418476 scopus 로고
    • Covalent bond formation between a DNA-cytosine methyltransferase and DNA containing 5-azacytosine
    • 620971
    • Santi DV, Norment A, Garrett CE, (1984) Covalent bond formation between a DNA-cytosine methyltransferase and DNA containing 5-azacytosine. Proc Natl Acad Sci U S A81: 6993–6997. 6209710
    • (1984) Proc Natl Acad Sci U S A , vol.81 , pp. 6993-6997
    • Santi, D.V.1    Norment, A.2    Garrett, C.E.3
  • 74
    • 77955238361 scopus 로고    scopus 로고
    • Targeting of 5-aza-2'-deoxycytidine residues by chromatin-associated DNMT1 induces proteasomal degradation of the free enzyme
    • 2034813
    • Patel K, Dickson J, Din S, Macleod K, Jodrell D, Ramsahoye B, (2010) Targeting of 5-aza-2'-deoxycytidine residues by chromatin-associated DNMT1 induces proteasomal degradation of the free enzyme. Nucleic Acids Res38: 4313–4324. doi: 10.1093/nar/gkq187 20348135
    • (2010) Nucleic Acids Res , vol.38 , pp. 4313-4324
    • Patel, K.1    Dickson, J.2    Din, S.3    Macleod, K.4    Jodrell, D.5    Ramsahoye, B.6
  • 75
    • 0020032366 scopus 로고
    • Inhibition of DNA methyltransferase and induction of Friend erythroleukemia cell differentiation by 5-azacytidine and 5-aza-2'-deoxycytidine
    • 617338
    • Creusot F, Acs G, Christman JK, (1982) Inhibition of DNA methyltransferase and induction of Friend erythroleukemia cell differentiation by 5-azacytidine and 5-aza-2'-deoxycytidine. J Biol Chem257: 2041–2048. 6173384
    • (1982) J Biol Chem , vol.257 , pp. 2041-2048
    • Creusot, F.1    Acs, G.2    Christman, J.K.3
  • 76
    • 84899960103 scopus 로고    scopus 로고
    • Key clinical observations after 5-azacytidine and decitabine treatment of myelodysplastic syndromes suggest practical solutions for better outcomes
    • 2431922
    • Saunthararajah Y, (2013) Key clinical observations after 5-azacytidine and decitabine treatment of myelodysplastic syndromes suggest practical solutions for better outcomes. Hematology Am Soc Hematol Educ Program2013: 511–521. doi: 10.1182/asheducation-2013.1.511 24319226
    • (2013) Hematology Am Soc Hematol Educ Program , vol.2013 , pp. 511-521
    • Saunthararajah, Y.1
  • 77
    • 33750326078 scopus 로고    scopus 로고
    • Phase I study of decitabine-mediated gene expression in patients with cancers involving the lungs, esophagus, or pleura
    • 1702098
    • Schrump DS, Fischette MR, Nguyen DM, Zhao M, Li X, Kunst TF, et al. (2006) Phase I study of decitabine-mediated gene expression in patients with cancers involving the lungs, esophagus, or pleura. Clin Cancer Res12: 5777–5785. doi: 10.1158/1078-0432.CCR-06-0669 17020984
    • (2006) Clin Cancer Res , vol.12 , pp. 5777-5785
    • Schrump, D.S.1    Fischette, M.R.2    Nguyen, D.M.3    Zhao, M.4    Li, X.5    Kunst, T.F.6
  • 78
    • 84863802864 scopus 로고    scopus 로고
    • Decitabine triphosphate levels in peripheral blood mononuclear cells from patients receiving prolonged low-dose decitabine administration: a pilot study
    • Jansen RS, Rosing H, Wijermans PW, Keizer RJ, Schellens JH, Beijnen JH, (2012) Decitabine triphosphate levels in peripheral blood mononuclear cells from patients receiving prolonged low-dose decitabine administration: a pilot study. Cancer Chemotherapy and Pharmacol69: 1457–1466.
    • (2012) Cancer Chemotherapy and Pharmacol , vol.69 , pp. 1457-1466
    • Jansen, R.S.1    Rosing, H.2    Wijermans, P.W.3    Keizer, R.J.4    Schellens, J.H.5    Beijnen, J.H.6
  • 79
    • 79959318635 scopus 로고    scopus 로고
    • Phase I study of oral azacitidine in myelodysplastic syndromes, chronic myelomonocytic leukemia, and acute myeloid leukemia
    • 2157664
    • Garcia-Manero G, Gore SD, Cogle C, Ward R, Shi T, Macbeth KJ, et al. (2011) Phase I study of oral azacitidine in myelodysplastic syndromes, chronic myelomonocytic leukemia, and acute myeloid leukemia. J Clin Oncol29: 2521–2527. doi: 10.1200/JCO.2010.34.4226 21576646
    • (2011) J Clin Oncol , vol.29 , pp. 2521-2527
    • Garcia-Manero, G.1    Gore, S.D.2    Cogle, C.3    Ward, R.4    Shi, T.5    Macbeth, K.J.6
  • 80
    • 66649105473 scopus 로고    scopus 로고
    • Decitabine effect on tumor global DNA methylation and other parameters in a phase I trial in refractory solid tumors and lymphomas
    • 1947073
    • Stewart DJ, Issa JP, Kurzrock R, Nunez MI, Jelinek J, Hong D, et al. (2009) Decitabine effect on tumor global DNA methylation and other parameters in a phase I trial in refractory solid tumors and lymphomas. Clin Cancer Res15: 3881–3888. doi: 10.1158/1078-0432.CCR-08-2196 19470736
    • (2009) Clin Cancer Res , vol.15 , pp. 3881-3888
    • Stewart, D.J.1    Issa, J.P.2    Kurzrock, R.3    Nunez, M.I.4    Jelinek, J.5    Hong, D.6
  • 81
    • 77954121574 scopus 로고    scopus 로고
    • A phase 1 and pharmacodynamic study of decitabine in combination with carboplatin in patients with recurrent, platinum-resistant, epithelial ovarian cancer
    • 2056412
    • Fang F, Balch C, Schilder J, Breen T, Zhang S, Shen C, et al. (2010) A phase 1 and pharmacodynamic study of decitabine in combination with carboplatin in patients with recurrent, platinum-resistant, epithelial ovarian cancer. Cancer116: 4043–4053. doi: 10.1002/cncr.25204 20564122
    • (2010) Cancer , vol.116 , pp. 4043-4053
    • Fang, F.1    Balch, C.2    Schilder, J.3    Breen, T.4    Zhang, S.5    Shen, C.6
  • 82
    • 55549100633 scopus 로고    scopus 로고
    • Increased platelet adhesion under flow conditions is induced by both thalassemic platelets and red blood cells
    • 1898953
    • Goldschmidt N, Spectre G, Brill A, Zelig O, Goldfarb A, Rachmilewitz E, et al. (2008) Increased platelet adhesion under flow conditions is induced by both thalassemic platelets and red blood cells. Thrombosis and Haemostasis100: 864–870. 18989531
    • (2008) Thrombosis and Haemostasis , vol.100 , pp. 864-870
    • Goldschmidt, N.1    Spectre, G.2    Brill, A.3    Zelig, O.4    Goldfarb, A.5    Rachmilewitz, E.6
  • 83
    • 84959423289 scopus 로고    scopus 로고
    • Neutrophils, platelets, and inflammatory pathways at the nexus of sickle cell disease pathophysiology
    • 2675891
    • Zhang D, Xu C, Manwani D, Frenette PS, (2016) Neutrophils, platelets, and inflammatory pathways at the nexus of sickle cell disease pathophysiology. Blood127: 801–809. doi: 10.1182/blood-2015-09-618538 26758915
    • (2016) Blood , vol.127 , pp. 801-809
    • Zhang, D.1    Xu, C.2    Manwani, D.3    Frenette, P.S.4
  • 84
    • 44949126303 scopus 로고    scopus 로고
    • Elevated levels of circulating procoagulant microparticles in patients with beta-thalassemia intermedia
    • 1846064
    • Habib A, Kunzelmann C, Shamseddeen W, Zobairi F, Freyssinet JM, Taher A, (2008) Elevated levels of circulating procoagulant microparticles in patients with beta-thalassemia intermedia. Haematologica93: 941–942. doi: 10.3324/haematol.12460 18460647
    • (2008) Haematologica , vol.93 , pp. 941-942
    • Habib, A.1    Kunzelmann, C.2    Shamseddeen, W.3    Zobairi, F.4    Freyssinet, J.M.5    Taher, A.6
  • 85
    • 38549117898 scopus 로고    scopus 로고
    • Hemolysis-associated hypercoagulability in sickle cell disease: the plot (and blood) thickens!
    • 1816677
    • Gladwin MT, Kato GJ, (2008) Hemolysis-associated hypercoagulability in sickle cell disease: the plot (and blood) thickens!Haematologica93: 1–3. doi: 10.3324/haematol.12318 18166776
    • (2008) Haematologica , vol.93 , pp. 1-3
    • Gladwin, M.T.1    Kato, G.J.2
  • 86
    • 38549131853 scopus 로고    scopus 로고
    • Coagulation activation and inflammation in sickle cell disease-associated pulmonary hypertension
    • 1816678
    • Ataga KI, Moore CG, Hillery CA, Jones S, Whinna HC, Strayhorn D, et al. (2008) Coagulation activation and inflammation in sickle cell disease-associated pulmonary hypertension. Haematologica93: 20–26. doi: 10.3324/haematol.11763 18166781
    • (2008) Haematologica , vol.93 , pp. 20-26
    • Ataga, K.I.1    Moore, C.G.2    Hillery, C.A.3    Jones, S.4    Whinna, H.C.5    Strayhorn, D.6
  • 87
    • 84868526330 scopus 로고    scopus 로고
    • Prothrombin activation in blood coagulation: the erythrocyte contribution to thrombin generation
    • 2296846
    • Whelihan MF, Zachary V, Orfeo T, Mann KG, (2012) Prothrombin activation in blood coagulation: the erythrocyte contribution to thrombin generation. Blood120: 3837–3845. doi: 10.1182/blood-2012-05-427856 22968460
    • (2012) Blood , vol.120 , pp. 3837-3845
    • Whelihan, M.F.1    Zachary, V.2    Orfeo, T.3    Mann, K.G.4
  • 88
    • 0030751208 scopus 로고    scopus 로고
    • In-vivo platelet activation correlates with red cell anionic phospholipid exposure in patients with beta-thalassaemia major
    • 923356
    • Ruf A, Pick M, Deutsch V, Patscheke H, Goldfarb A, Rachmilewitz EA, et al. (1997) In-vivo platelet activation correlates with red cell anionic phospholipid exposure in patients with beta-thalassaemia major. Br J Haematol98: 51–56. 9233563
    • (1997) Br J Haematol , vol.98 , pp. 51-56
    • Ruf, A.1    Pick, M.2    Deutsch, V.3    Patscheke, H.4    Goldfarb, A.5    Rachmilewitz, E.A.6
  • 89
    • 70449359661 scopus 로고    scopus 로고
    • Hypercoagulability in splenectomized thalassemic patients detected by whole-blood thromboelastometry, but not by thrombin generation in platelet-poor plasma
    • 1964816
    • Tripodi A, Cappellini MD, Chantarangkul V, Padovan L, Fasulo MR, Marcon A, et al. (2009) Hypercoagulability in splenectomized thalassemic patients detected by whole-blood thromboelastometry, but not by thrombin generation in platelet-poor plasma. Haematologica94: 1520–1527. doi: 10.3324/haematol.2009.010546 19648162
    • (2009) Haematologica , vol.94 , pp. 1520-1527
    • Tripodi, A.1    Cappellini, M.D.2    Chantarangkul, V.3    Padovan, L.4    Fasulo, M.R.5    Marcon, A.6
  • 90
    • 84889648423 scopus 로고    scopus 로고
    • The contribution of red blood cells to thrombin generation in sickle cell disease: meizothrombin generation on sickled red blood cells
    • 2411916
    • Whelihan MF, Mooberry MJ, Zachary V, Bradford RL, Ataga KI, Mann KG, et al. (2013) The contribution of red blood cells to thrombin generation in sickle cell disease: meizothrombin generation on sickled red blood cells. J Thromb Haemost11: 2187–2189. doi: 10.1111/jth.12423 24119168
    • (2013) J Thromb Haemost , vol.11 , pp. 2187-2189
    • Whelihan, M.F.1    Mooberry, M.J.2    Zachary, V.3    Bradford, R.L.4    Ataga, K.I.5    Mann, K.G.6
  • 93
    • 0034254238 scopus 로고    scopus 로고
    • Fetal hemoglobin in sickle cell disease: relationship to erythrocyte phosphatidylserine exposure and coagulation activation
    • 1091093
    • Setty BN, Kulkarni S, Rao AK, Stuart MJ, (2000) Fetal hemoglobin in sickle cell disease: relationship to erythrocyte phosphatidylserine exposure and coagulation activation. Blood96: 1119–1124. 10910931
    • (2000) Blood , vol.96 , pp. 1119-1124
    • Setty, B.N.1    Kulkarni, S.2    Rao, A.K.3    Stuart, M.J.4
  • 94
    • 0035353182 scopus 로고    scopus 로고
    • Fetal hemoglobin in sickle cell anemia: relationship to erythrocyte adhesion markers and adhesion
    • 1131324
    • Setty BN, Kulkarni S, Dampier CD, Stuart MJ, (2001) Fetal hemoglobin in sickle cell anemia: relationship to erythrocyte adhesion markers and adhesion. Blood97: 2568–2573. 11313243
    • (2001) Blood , vol.97 , pp. 2568-2573
    • Setty, B.N.1    Kulkarni, S.2    Dampier, C.D.3    Stuart, M.J.4
  • 95
    • 85047686332 scopus 로고    scopus 로고
    • Platelet count in essential thrombocythemia: the more the better?
    • 1884072
    • Tefferi A, (2008) Platelet count in essential thrombocythemia: the more the better?Blood112: 3526–3527. doi: 10.1182/blood-2008-07-168807 18840727
    • (2008) Blood , vol.112 , pp. 3526-3527
    • Tefferi, A.1
  • 97
    • 34848893773 scopus 로고    scopus 로고
    • Red cell exchange in sickle cell disease
    • 1712403
    • Swerdlow PS, (2006) Red cell exchange in sickle cell disease. Hematology Am Soc Hematol Educ Program: 48–53. doi: 10.1182/asheducation-2006.1.48 17124039
    • (2006) Hematology Am Soc Hematol Educ Program , pp. 48-53
    • Swerdlow, P.S.1
  • 98
    • 0031028973 scopus 로고    scopus 로고
    • Mouse strain-dependent changes in frequency and proliferation of hematopoietic stem cells during aging: correlation between lifespan and cycling activity
    • 905763
    • de Haan G, Nijhof W, Van Zant G, (1997) Mouse strain-dependent changes in frequency and proliferation of hematopoietic stem cells during aging: correlation between lifespan and cycling activity. Blood89: 1543–1550. 9057635
    • (1997) Blood , vol.89 , pp. 1543-1550
    • de Haan, G.1    Nijhof, W.2    Van Zant, G.3
  • 99
    • 84863411353 scopus 로고    scopus 로고
    • p53-Independent, normal stem cell sparing epigenetic differentiation therapy for myeloid and other malignancies
    • 2228949
    • Saunthararajah Y, Triozzi P, Rini B, Singh A, Radivoyevitch T, Sekeres M, et al. (2012) p53-Independent, normal stem cell sparing epigenetic differentiation therapy for myeloid and other malignancies. Semin Oncol39: 97–108. doi: 10.1053/j.seminoncol.2011.11.011 22289496
    • (2012) Semin Oncol , vol.39 , pp. 97-108
    • Saunthararajah, Y.1    Triozzi, P.2    Rini, B.3    Singh, A.4    Radivoyevitch, T.5    Sekeres, M.6
  • 100
    • 80755140039 scopus 로고    scopus 로고
    • p53 independent epigenetic-differentiation treatment in xenotransplant models of acute myeloid leukemia
    • 2170149
    • Ng KP, Ebrahem Q, Negrotto S, Mahfouz RZ, Link KA, Hu Z, et al. (2011) p53 independent epigenetic-differentiation treatment in xenotransplant models of acute myeloid leukemia. Leukemia25: 1739–1750. doi: 10.1038/leu.2011.159 21701495
    • (2011) Leukemia , vol.25 , pp. 1739-1750
    • Ng, K.P.1    Ebrahem, Q.2    Negrotto, S.3    Mahfouz, R.Z.4    Link, K.A.5    Hu, Z.6
  • 101
    • 84856720426 scopus 로고    scopus 로고
    • CpG methylation patterns and decitabine treatment response in acute myeloid leukemia cells and normal hematopoietic precursors
    • 2183661
    • Negrotto S, Ng KP, Jankowska AM, Bodo J, Gopalan B, Guinta K, et al. (2012) CpG methylation patterns and decitabine treatment response in acute myeloid leukemia cells and normal hematopoietic precursors. Leukemia26: 244–254. doi: 10.1038/leu.2011.207 21836612
    • (2012) Leukemia , vol.26 , pp. 244-254
    • Negrotto, S.1    Ng, K.P.2    Jankowska, A.M.3    Bodo, J.4    Gopalan, B.5    Guinta, K.6
  • 102
    • 84863337757 scopus 로고    scopus 로고
    • Transient Low Doses of DNA-Demethylating Agents Exert Durable Antitumor Effects on Hematological and Epithelial Tumor Cells
    • 2243993
    • Tsai HC, Li H, Van Neste L, Cai Y, Robert C, Rassool FV, et al. (2012) Transient Low Doses of DNA-Demethylating Agents Exert Durable Antitumor Effects on Hematological and Epithelial Tumor Cells. Cancer Cell21: 430–446. doi: 10.1016/j.ccr.2011.12.029 22439938
    • (2012) Cancer Cell , vol.21 , pp. 430-446
    • Tsai, H.C.1    Li, H.2    Van Neste, L.3    Cai, Y.4    Robert, C.5    Rassool, F.V.6
  • 103
    • 84879788695 scopus 로고    scopus 로고
    • Epigenetic action of decitabine (5-aza-2'-deoxycytidine) is more effective against acute myeloid leukemia than cytotoxic action of cytarabine (ARA-C)
    • 2366038
    • Momparler RL, Cote S, Momparler LF, (2013) Epigenetic action of decitabine (5-aza-2'-deoxycytidine) is more effective against acute myeloid leukemia than cytotoxic action of cytarabine (ARA-C). Leuk Res. 37: 980–984. doi: 10.1016/j.leukres.2013.04.019 23660386
    • (2013) Leuk Res , vol.37 , pp. 980-984
    • Momparler, R.L.1    Cote, S.2    Momparler, L.F.3
  • 104
    • 85028928385 scopus 로고    scopus 로고
    • Higher-Level Pathway Objectives of Epigenetic Therapy: A Solution to the p53 Problem in Cancer
    • 2856165
    • Velcheti V, Radivoyevitch T, Saunthararajah Y, (2017) Higher-Level Pathway Objectives of Epigenetic Therapy: A Solution to the p53 Problem in Cancer. Am Soc Clin Oncol Educ Book37: 812–824. doi: 10.14694/EDBK_174175 28561650
    • (2017) Am Soc Clin Oncol Educ Book , vol.37 , pp. 812-824
    • Velcheti, V.1    Radivoyevitch, T.2    Saunthararajah, Y.3
  • 105
    • 79951846889 scopus 로고    scopus 로고
    • Noncytotoxic differentiation treatment of renal cell cancer
    • 2130398
    • Negrotto S, Hu Z, Alcazar O, Ng KP, Triozzi P, Lindner D, et al. (2011) Noncytotoxic differentiation treatment of renal cell cancer. Cancer Res71: 1431–1441. doi: 10.1158/0008-5472.CAN-10-2422 21303982
    • (2011) Cancer Res , vol.71 , pp. 1431-1441
    • Negrotto, S.1    Hu, Z.2    Alcazar, O.3    Ng, K.P.4    Triozzi, P.5    Lindner, D.6
  • 106
    • 84856377986 scopus 로고    scopus 로고
    • Epigenetic regulation by decitabine of melanoma differentiation in vitro and in vivo
    • 2179662
    • Alcazar O, Achberger S, Aldrich W, Hu Z, Negrotto S, Saunthararajah Y, et al. (2012) Epigenetic regulation by decitabine of melanoma differentiation in vitro and in vivo. Int J Cancer131: 18–29. doi: 10.1002/ijc.26320 21796622
    • (2012) Int J Cancer , vol.131 , pp. 18-29
    • Alcazar, O.1    Achberger, S.2    Aldrich, W.3    Hu, Z.4    Negrotto, S.5    Saunthararajah, Y.6
  • 107
    • 84898953170 scopus 로고    scopus 로고
    • Subchronic oral toxicity study of decitabine in combination with tetrahydrouridine in CD-1 mice
    • Terse P, Engelke K, Chan K, Ling Y, Sharpnack D, Saunthararajah Y, et al. (2014) Subchronic oral toxicity study of decitabine in combination with tetrahydrouridine in CD-1 mice. Int J of Toxicol33: 75–85.
    • (2014) Int J of Toxicol , vol.33 , pp. 75-85
    • Terse, P.1    Engelke, K.2    Chan, K.3    Ling, Y.4    Sharpnack, D.5    Saunthararajah, Y.6
  • 108
    • 0024369444 scopus 로고
    • Modulation of deoxynucleotide metabolism by the deoxycytidylate deaminase inhibitor 3,4,5,6-tetrahydrodeoxyuridine
    • 268865
    • Heinemann V, Plunkett W, (1989) Modulation of deoxynucleotide metabolism by the deoxycytidylate deaminase inhibitor 3,4,5,6-tetrahydrodeoxyuridine. Biochem Pharmacol38: 4115–4121. 2688654
    • (1989) Biochem Pharmacol , vol.38 , pp. 4115-4121
    • Heinemann, V.1    Plunkett, W.2
  • 109
    • 0015896830 scopus 로고
    • Pharmacokinetics of 1-beta-D-arabinofuranosylcytosine (ARA-C) deamination in several species
    • 420088
    • Dedrick RL, Forrester DD, Cannon JN, el-Dareer SM, Mellett LB, (1973) Pharmacokinetics of 1-beta-D-arabinofuranosylcytosine (ARA-C) deamination in several species. Biochem Pharmacol22: 2405–2417. 4200888
    • (1973) Biochem Pharmacol , vol.22 , pp. 2405-2417
    • Dedrick, R.L.1    Forrester, D.D.2    Cannon, J.N.3    el-Dareer, S.M.4    Mellett, L.B.5
  • 110
    • 44149110031 scopus 로고    scopus 로고
    • Fine tuning of globin gene expression by DNA methylation
    • 1718367
    • Goren A, Simchen G, Fibach E, Szabo PE, Tanimoto K, Chakalova L, et al. (2006) Fine tuning of globin gene expression by DNA methylation. PLoS ONE1(1): e46. doi: 10.1371/journal.pone.0000046 17183675
    • (2006) PLoS ONE , vol.1 , Issue.1 , pp. e46
    • Goren, A.1    Simchen, G.2    Fibach, E.3    Szabo, P.E.4    Tanimoto, K.5    Chakalova, L.6
  • 111
    • 0021286875 scopus 로고
    • Hydroxyurea enhances fetal hemoglobin production in sickle cell anemia
    • 620502
    • Platt OS, Orkin SH, Dover G, Beardsley GP, Miller B, Nathan DG, (1984) Hydroxyurea enhances fetal hemoglobin production in sickle cell anemia. J Clin Invest74: 652–656. doi: 10.1172/JCI111464 6205021
    • (1984) J Clin Invest , vol.74 , pp. 652-656
    • Platt, O.S.1    Orkin, S.H.2    Dover, G.3    Beardsley, G.P.4    Miller, B.5    Nathan, D.G.6
  • 112
    • 4544364980 scopus 로고    scopus 로고
    • DNA hypo-methylating agents and sickle cell disease
    • Saunthararajah Y, Lavelle D, DeSimone J, (2004) DNA hypo-methylating agents and sickle cell disease. Brit J Haematol126: 629–636.
    • (2004) Brit J Haematol , vol.126 , pp. 629-636
    • Saunthararajah, Y.1    Lavelle, D.2    DeSimone, J.3
  • 113
    • 46749139575 scopus 로고    scopus 로고
    • The novel histone deacetylase inhibitor, LBH589, induces expression of DNA damage response genes and apoptosis in Ph- acute lymphoblastic leukemia cells
    • 1834932
    • Scuto A, Kirschbaum M, Kowolik C, Kretzner L, Juhasz A, Atadja P, et al. (2008) The novel histone deacetylase inhibitor, LBH589, induces expression of DNA damage response genes and apoptosis in Ph- acute lymphoblastic leukemia cells. Blood111: 5093–5100. doi: 10.1182/blood-2007-10-117762 18349321
    • (2008) Blood , vol.111 , pp. 5093-5100
    • Scuto, A.1    Kirschbaum, M.2    Kowolik, C.3    Kretzner, L.4    Juhasz, A.5    Atadja, P.6
  • 114
    • 77957091318 scopus 로고    scopus 로고
    • Histone deacetylase inhibitor induces DNA damage, which normal but not transformed cells can repair
    • 2067923
    • Lee JH, Choy ML, Ngo L, Foster SS, Marks PA, (2010) Histone deacetylase inhibitor induces DNA damage, which normal but not transformed cells can repair. Proc Natl Acad Sci U S A107: 14639–14644. doi: 10.1073/pnas.1008522107 20679231
    • (2010) Proc Natl Acad Sci U S A , vol.107 , pp. 14639-14644
    • Lee, J.H.1    Choy, M.L.2    Ngo, L.3    Foster, S.S.4    Marks, P.A.5
  • 115
    • 77953170728 scopus 로고    scopus 로고
    • Inhibition of histone deacetylase in cancer cells slows down replication forks, activates dormant origins, and induces DNA damage
    • 2046051
    • Conti C, Leo E, Eichler GS, Sordet O, Martin MM, Fan A, et al. (2010) Inhibition of histone deacetylase in cancer cells slows down replication forks, activates dormant origins, and induces DNA damage. Cancer Res70: 4470–4480. doi: 10.1158/0008-5472.CAN-09-3028 20460513
    • (2010) Cancer Res , vol.70 , pp. 4470-4480
    • Conti, C.1    Leo, E.2    Eichler, G.S.3    Sordet, O.4    Martin, M.M.5    Fan, A.6
  • 116
    • 33748427812 scopus 로고    scopus 로고
    • Histone deacetylase inhibitors (HDI) cause DNA damage in leukemia cells: a mechanism for leukemia-specific HDI-dependent apoptosis?
    • 1687770
    • Gaymes TJ, Padua RA, Pla M, Orr S, Omidvar N, Chomienne C, et al. (2006) Histone deacetylase inhibitors (HDI) cause DNA damage in leukemia cells: a mechanism for leukemia-specific HDI-dependent apoptosis?Mol Cancer Res4: 563–573. doi: 10.1158/1541-7786.MCR-06-0111 16877702
    • (2006) Mol Cancer Res , vol.4 , pp. 563-573
    • Gaymes, T.J.1    Padua, R.A.2    Pla, M.3    Orr, S.4    Omidvar, N.5    Chomienne, C.6
  • 117
    • 30344477367 scopus 로고    scopus 로고
    • Histone deacetylase inhibitors and the promise of epigenetic (and more) treatments for cancer
    • 1639752
    • Minucci S, Pelicci PG, (2006) Histone deacetylase inhibitors and the promise of epigenetic (and more) treatments for cancer. Nat Rev Cancer6: 38–51. doi: 10.1038/nrc1779 16397526
    • (2006) Nat Rev Cancer , vol.6 , pp. 38-51
    • Minucci, S.1    Pelicci, P.G.2
  • 118
    • 84861568350 scopus 로고    scopus 로고
    • DNMT1 modulates gene expression without its catalytic activity partially through its interactions with histone-modifying enzymes
    • 2227888
    • Clements EG, Mohammad HP, Leadem BR, Easwaran H, Cai Y, Van Neste L, et al. (2012) DNMT1 modulates gene expression without its catalytic activity partially through its interactions with histone-modifying enzymes. Nucleic Acids Res40: 4334–4346. doi: 10.1093/nar/gks031 22278882
    • (2012) Nucleic Acids Res , vol.40 , pp. 4334-4346
    • Clements, E.G.1    Mohammad, H.P.2    Leadem, B.R.3    Easwaran, H.4    Cai, Y.5    Van Neste, L.6
  • 119
    • 84991381413 scopus 로고    scopus 로고
    • The interplay between the lysine demethylase KDM1A and DNA methyltransferases in cancer cells is cell cycle dependent
    • 2744928
    • Brenner C, Luciani J, Bizet M, Ndlovu M, Josseaux E, Dedeurwaerder S, et al. (2016) The interplay between the lysine demethylase KDM1A and DNA methyltransferases in cancer cells is cell cycle dependent. Oncotarget7: 58939–58952. doi: 10.18632/oncotarget.10624 27449289
    • (2016) Oncotarget , vol.7 , pp. 58939-58952
    • Brenner, C.1    Luciani, J.2    Bizet, M.3    Ndlovu, M.4    Josseaux, E.5    Dedeurwaerder, S.6


* 이 정보는 Elsevier사의 SCOPUS DB에서 KISTI가 분석하여 추출한 것입니다.