#PAGE_PARAMS# #ADS_HEAD_SCRIPTS# #MICRODATA#

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


In a clinical trial, Yogen Saunthararajah and colleagues target DNA methyltransferase with small molecules to reactivate fetal hemoglobin which inhibits polymerisation of mutated sickle cell hemoglobin.


Vyšlo v časopise: Oral tetrahydrouridine and decitabine for non-cytotoxic epigenetic gene regulation in sickle cell disease: A randomized phase 1 study. PLoS Med 14(9): e32767. doi:10.1371/journal.pmed.1002382
Kategorie: Research Article
prolekare.web.journal.doi_sk: https://doi.org/10.1371/journal.pmed.1002382

Souhrn

In a clinical trial, Yogen Saunthararajah and colleagues target DNA methyltransferase with small molecules to reactivate fetal hemoglobin which inhibits polymerisation of mutated sickle cell hemoglobin.


Zdroje

1. 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 Med 330: 1639–1644. doi: 10.1056/NEJM199406093302303 7993409

2. Lanzkron S, Carroll CP, Haywood C Jr. (2013) Mortality rates and age at death from sickle cell disease: U.S., 1979–2005. Public Health Reports 128: 110–116. doi: 10.1177/003335491312800206 23450875

3. Goldberg MA, Husson MA, Bunn HF (1977) Participation of hemoglobins A and F in polymerization of sickle hemoglobin. J Biol Chem 252: 3414–3421. 16902

4. 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 A 76: 670–672. 284392

5. Akinsheye I, Alsultan A, Solovieff N, Ngo D, Baldwin CT, Sebastiani P, et al. (2011) Fetal hemoglobin in sickle cell anemia. Blood 118: 19–27. doi: 10.1182/blood-2011-03-325258 21490337

6. Platt OS, Thorington BD, Brambilla DJ, Milner PF, Rosse WF, Vichinsky E, et al. (1991) Pain in sickle cell disease. Rates and risk factors. N Engl J Med 325: 11–16. doi: 10.1056/NEJM199107043250103 1710777

7. Powars DR, Weiss JN, Chan LS, Schroeder WA (1984) Is there a threshold level of fetal hemoglobin that ameliorates morbidity in sickle cell anemia? Blood 63: 921–926. 6200161

8. 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. JAMA 289: 1645–1651. doi: 10.1001/jama.289.13.1645 12672732

9. Atweh GF, Schechter AN (2001) Pharmacologic induction of fetal hemoglobin: raising the therapeutic bar in sickle cell disease. Curr Opin Hematol 8: 123–130. 11224687

10. 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 Med 115: 614–620. 1892333

11. 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 Hematol 86: 430–432. doi: 10.1002/ajh.21994 21523807

12. 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. Blood 129: 385–387. doi: 10.1182/blood-2016-09-738104 27919909

13. 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. Blood 21: 261–281. 14022587

14. 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 Med 88: 1–6. 619731

15. 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 Haematol 156: 259–264. doi: 10.1111/j.1365-2141.2011.08916.x 22017641

16. DeSimone J, Biel SI, Heller P (1978) Stimulation of fetal hemoglobin synthesis in baboons by hemolysis and hypoxia. Proc Natl Acad Sci U S A 75: 2937–2940. 96444

17. DeSimone J, Heller P, Adams JG (1979) Hemopoietic stress and fetal hemoglobin synthesis: comparative studies in vivo and in vitro. Blood 54: 1176–1181. 115514

18. 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 Invest 65: 224–226. doi: 10.1172/JCI109654 6765958

19. DeSimone J, Heller P, Biel M, Zwiers D (1981) Genetic relationship between fetal Hb levels in normal and erythropoietically stressed baboons. Br J Haematol 49: 175–183. 6170305

20. DeSimone J, Biel M, Heller P (1982) Maintenance of fetal hemoglobin (HbF) elevations in the baboon by prolonged erythropoietic stress. Blood 60: 519–523. 6178456

21. 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. Blood 67: 1083–1089. 2420395

22. 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 Med 332: 1317–1322. doi: 10.1056/NEJM199505183322001 7715639

23. 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. Blood 89: 1078–1088. 9028341

24. 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 Haematol 105: 491–496. 10233426

25. 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. Blood 79: 2555–2565. 1375104

26. Ballas SK, Barton FB, Waclawiw MA, Swerdlow P, Eckman JR, Pegelow CH, et al. (2006) Hydroxyurea and sickle cell anemia: effect on quality of life. Health and Quality of Life Outcomes 4: 59. doi: 10.1186/1477-7525-4-59 16942629

27. Segal JB, Strouse JJ, Beach MC, Haywood C, Witkop C, Park H, et al. (2008) Hydroxyurea for the treatment of sickle cell disease. Evidence Report/technology Assessment: 1–95.

28. 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. JAMA 296: 310–318. doi: 10.1001/jama.296.3.310 16849664

29. 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. Blood 99: 10–14. 11756146

30. 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 & Cancer 63: 2146–2153.

31. 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 Epidemiol 45: 893–909. 1624972

32. 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 Haematol 153: 386–392. doi: 10.1111/j.1365-2141.2010.08479.x 21418176

33. van der Ploeg LH, Flavell RA (1980) DNA methylation in the human gamma delta beta-globin locus in erythroid and nonerythroid tissues. Cell 19: 947–958. 6247075

34. 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 A 80: 6907–6911. 6316333

35. 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 Biol 203: 439–455. 3199442

36. DeSimone J, Heller P, Hall L, Zwiers D (1982) 5-Azacytidine stimulates fetal hemoglobin synthesis in anemic baboons. Proc Natl Acad Sci U S A 79: 4428–4431. 6181507

37. 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 Med 307: 1469–1475. doi: 10.1056/NEJM198212093072401 6183586

38. 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. Blood 62: 370–380. 6191799

39. 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 A 80: 4842–4846. 6192443

40. 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. Blood 66: 527–532. 2411310

41. Lowrey CH, Nienhuis AW (1993) Brief report: treatment with azacitidine of patients with end-stage beta-thalassemia. N Engl J Med 329: 845–848. doi: 10.1056/NEJM199309163291205 7689171

42. 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. Blood 92: 30b–30b.

43. 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. Blood 102: 3865–3870. doi: 10.1182/blood-2003-05-1738 12907443

44. 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 A 110: 6518–6523. doi: 10.1073/pnas.1303976110 23576758

45. 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 Biol 31: 3298–3311. doi: 10.1128/MCB.05310-11 21670149

46. Santi DV, Garrett CE, Barr PJ (1983) On the mechanism of inhibition of DNA-cytosine methyltransferases by cytosine analogs. Cell 33: 9–10. 6205762

47. 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 Res 46: 5511–5517. 2428479

48. 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 Res 35: 4301–4312. doi: 10.1093/nar/gkm432 17576694

49. 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. Nature 524: 114–118. doi: 10.1038/nature14948 26200337

50. 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 Commun 7: 11040. doi: 10.1038/ncomms11040 27010513

51. Olivieri NF, Saunthararajah Y, Thayalasuthan V, Kwiatkowski J, Ware RE, Kuypers FA, et al. (2011) A pilot study of subcutaneous decitabine in beta-thalassemia intermedia. Blood 118: 2708–2711. doi: 10.1182/blood-2011-03-341909 21700776

52. 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 125: 1043–1055. doi: 10.1172/JCI78789 25621498

53. 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. Blood 103: 4102–4110. doi: 10.1182/blood-2003-07-2431 14976039

54. 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 Ther 9: 1536–1543. doi: 10.1158/1535-7163.MCT-10-0191 20501800

55. 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. Blood 99: 3905–3908. 12010787

56. 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 Spectrom 20: 1117–1126. doi: 10.1002/rcm.2423 16523529

57. 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 Res 35: e31. doi: 10.1093/nar/gkl1156 17264127

58. Camiener GW, Smith CG (1965) Studies of the enzymatic deamination of cytosine arabinoside. I. Enzyme distribution and species specificity. Biochem Pharmacol 14: 1405–1416. 4956026

59. 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 Rep 59: 459–465. 54211

60. DeSimone J, Heller P, Molokie RE, Hall L, Zwiers D (1985) Tetrahydrouridine, cytidine analogues, and hemoglobin F. Am J Hematol 18: 283–288. 2579548

61. 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 Res 14: 3529–3535. doi: 10.1158/1078-0432.CCR-07-4885 18519786

62. 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. Oncotarget 3: 1137–1145. doi: 10.18632/oncotarget.597 23087155

63. 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 Res 42: 1736–1739. 6896013

64. 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. Leukemia 5: 991–998. 1961042

65. Wong PP, Currie VE, Mackey RW, Krakoff IH, Tan CT, Burchenal JH, et al. (1979) Phase I evaluation of tetrahydrouridine combined with cytosine arabinoside. Cancer Treat Rep 63: 1245–1249. 383291

66. Ho DH, Bodey GP, Hall SW, Benjamin RS, Brown NS, Freireich EJ, et al. (1978) Clinica, pharmacology of tetrahydrouridine. J Clin Pharmacol 18: 259–265. 641214

67. Kreis W, Woodcock TM, Gordon CS, Krakoff IH (1977) Tetrahydrouridine: Physiologic disposition and effect upon deamination of cytosine arabinoside in man. Cancer Treat Rep 61: 1347–1353. 589600

68. 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 Pharmacol 31: 481–484. 8453688

69. Goldenthal EI, Cookson KM, Geil RG, Wazeter FX (1974) Preclinical toxicologic evaluation of tetrahydrouridine (NSC-112907) in beagle dogs and rhesus monkeys. Cancer Chemother Rep3 5: 15–16.

70. 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. Blood 119: 1240–1247. doi: 10.1182/blood-2011-08-371690 22160381

71. 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. Epigenetics 4: 176–184. 19421002

72. 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

73. 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 A 81: 6993–6997. 6209710

74. 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 Res 38: 4313–4324. doi: 10.1093/nar/gkq187 20348135

75. 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 Chem 257: 2041–2048. 6173384

76. 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 Program 2013: 511–521. doi: 10.1182/asheducation-2013.1.511 24319226

77. 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 Res 12: 5777–5785. doi: 10.1158/1078-0432.CCR-06-0669 17020984

78. 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 Pharmacol 69: 1457–1466.

79. 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 Oncol 29: 2521–2527. doi: 10.1200/JCO.2010.34.4226 21576646

80. 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 Res 15: 3881–3888. doi: 10.1158/1078-0432.CCR-08-2196 19470736

81. 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. Cancer 116: 4043–4053. doi: 10.1002/cncr.25204 20564122

82. 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 Haemostasis 100: 864–870. 18989531

83. Zhang D, Xu C, Manwani D, Frenette PS (2016) Neutrophils, platelets, and inflammatory pathways at the nexus of sickle cell disease pathophysiology. Blood 127: 801–809. doi: 10.1182/blood-2015-09-618538 26758915

84. 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. Haematologica 93: 941–942. doi: 10.3324/haematol.12460 18460647

85. Gladwin MT, Kato GJ (2008) Hemolysis-associated hypercoagulability in sickle cell disease: the plot (and blood) thickens! Haematologica 93: 1–3. doi: 10.3324/haematol.12318 18166776

86. 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. Haematologica 93: 20–26. doi: 10.3324/haematol.11763 18166781

87. Whelihan MF, Zachary V, Orfeo T, Mann KG (2012) Prothrombin activation in blood coagulation: the erythrocyte contribution to thrombin generation. Blood 120: 3837–3845. doi: 10.1182/blood-2012-05-427856 22968460

88. 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 Haematol 98: 51–56. 9233563

89. 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. Haematologica 94: 1520–1527. doi: 10.3324/haematol.2009.010546 19648162

90. 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 Haemost 11: 2187–2189. doi: 10.1111/jth.12423 24119168

91. Chuncharunee S, Archararit N, Ungkanont A, Jootar S, Angchaisuksiri P, Bunyaratavej A, et al. (2000) Etiology and incidence of thrombotic and hemorrhagic disorders in Thai patients with extreme thrombocytosis. J Med Assoc Thai 83 Suppl 1: S95–100.

92. Hathirat P, Mahaphan W, Chuansumrit A, Pintadit P, Sasanakul W, Isarangkura P (1993) Platelet counts in thalassemic children before and after splenectomy. Southeast Asian J Trop Med Public Health 24 Suppl 1: 213–215.

93. Setty BN, Kulkarni S, Rao AK, Stuart MJ (2000) Fetal hemoglobin in sickle cell disease: relationship to erythrocyte phosphatidylserine exposure and coagulation activation. Blood 96: 1119–1124. 10910931

94. Setty BN, Kulkarni S, Dampier CD, Stuart MJ (2001) Fetal hemoglobin in sickle cell anemia: relationship to erythrocyte adhesion markers and adhesion. Blood 97: 2568–2573. 11313243

95. Tefferi A (2008) Platelet count in essential thrombocythemia: the more the better? Blood 112: 3526–3527. doi: 10.1182/blood-2008-07-168807 18840727

96. van der Bom JG, Heckbert SR, Lumley T, Holmes CE, Cushman M, Folsom AR, et al. (2009) Platelet count and the risk for thrombosis and death in the elderly. J Thromb Haemost 7: 399–405. doi: 10.1111/j.1538-7836.2008.03267.x 19143922

97. 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

98. 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. Blood 89: 1543–1550. 9057635

99. 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 Oncol 39: 97–108. doi: 10.1053/j.seminoncol.2011.11.011 22289496

100. 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. Leukemia 25: 1739–1750. doi: 10.1038/leu.2011.159 21701495

101. 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. Leukemia 26: 244–254. doi: 10.1038/leu.2011.207 21836612

102. 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 Cell 21: 430–446. doi: 10.1016/j.ccr.2011.12.029 22439938

103. 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

104. 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 Book 37: 812–824. doi: 10.14694/EDBK_174175 28561650

105. Negrotto S, Hu Z, Alcazar O, Ng KP, Triozzi P, Lindner D, et al. (2011) Noncytotoxic differentiation treatment of renal cell cancer. Cancer Res 71: 1431–1441. doi: 10.1158/0008-5472.CAN-10-2422 21303982

106. 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 Cancer 131: 18–29. doi: 10.1002/ijc.26320 21796622

107. 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 Toxicol 33: 75–85.

108. Heinemann V, Plunkett W (1989) Modulation of deoxynucleotide metabolism by the deoxycytidylate deaminase inhibitor 3,4,5,6-tetrahydrodeoxyuridine. Biochem Pharmacol 38: 4115–4121. 2688654

109. 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 Pharmacol 22: 2405–2417. 4200888

110. 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 ONE 1(1): e46. doi: 10.1371/journal.pone.0000046 17183675

111. Platt OS, Orkin SH, Dover G, Beardsley GP, Miller B, Nathan DG (1984) Hydroxyurea enhances fetal hemoglobin production in sickle cell anemia. J Clin Invest 74: 652–656. doi: 10.1172/JCI111464 6205021

112. Saunthararajah Y, Lavelle D, DeSimone J (2004) DNA hypo-methylating agents and sickle cell disease. Brit J Haematol 126: 629–636.

113. 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. Blood 111: 5093–5100. doi: 10.1182/blood-2007-10-117762 18349321

114. 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 A 107: 14639–14644. doi: 10.1073/pnas.1008522107 20679231

115. 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 Res 70: 4470–4480. doi: 10.1158/0008-5472.CAN-09-3028 20460513

116. 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 Res 4: 563–573. doi: 10.1158/1541-7786.MCR-06-0111 16877702

117. Minucci S, Pelicci PG (2006) Histone deacetylase inhibitors and the promise of epigenetic (and more) treatments for cancer. Nat Rev Cancer 6: 38–51. doi: 10.1038/nrc1779 16397526

118. 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 Res 40: 4334–4346. doi: 10.1093/nar/gks031 22278882

119. 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. Oncotarget 7: 58939–58952. doi: 10.18632/oncotarget.10624 27449289

Štítky
Interné lekárstvo

Článok vyšiel v časopise

PLOS Medicine


2017 Číslo 9
Najčítanejšie tento týždeň
Najčítanejšie v tomto čísle
Kurzy

Zvýšte si kvalifikáciu online z pohodlia domova

Získaná hemofilie - Povědomí o nemoci a její diagnostika
nový kurz

Eozinofilní granulomatóza s polyangiitidou
Autori: doc. MUDr. Martina Doubková, Ph.D.

Všetky kurzy
Prihlásenie
Zabudnuté heslo

Zadajte e-mailovú adresu, s ktorou ste vytvárali účet. Budú Vám na ňu zasielané informácie k nastaveniu nového hesla.

Prihlásenie

Nemáte účet?  Registrujte sa

#ADS_BOTTOM_SCRIPTS#