Skip to main content
Log in

The Application of Delivery Systems for DNA Methyltransferase Inhibitors

  • Review Article
  • Published:
BioDrugs Aims and scope Submit manuscript

Abstract

DNA methylation, which often occurs at the cytosine residue of cytosine-guanine dinucleotides, is critical for the control of gene expression and mitotic inheritance in eukaryotes. DNA methylation silences gene expression either by directly hindering the access of transcription factors to the target DNA, or through recruitment of histone deacetylases to remodel the chromatin structure to an inactive state. Aberrant hypermethylation of tumor suppressor genes is commonly associated with the development of cancer. A number of anti-cancer agents have been developed that function through demethylation, reversing regional hypermethylation to restore the expression of tumor suppressor genes. Azacitidine and decitabine are used in the clinic, but their applications are limited to myelodysplastic syndrome and other blood-related diseases. Despite the potency of these drugs, their broader clinical application is restricted by cytotoxicity, nonspecific targeting, structural instability, catabolism, and poor bioavailability. Further improvements in the delivery systems for these drugs could overcome the issues associated with inefficient bioavailability, whilst facilitating the administration of combinations of demethylating agents and histone deacetylase inhibitors to enhance efficacy. This review focuses on the current limitations of existing demethylating agents and highlights possible approaches using recent developments in drug delivery systems to improve the clinical potential of these drugs.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1
Table I
Fig. 2
Fig. 3
Table II
Table III
Fig. 4

Similar content being viewed by others

References

  1. Hotchkiss RD. The quantitative separation of purines, pyrimidines, and nucleosides by paper chromatography. J Biol Chem 1948; 175: 315–32

    CAS  PubMed  Google Scholar 

  2. Wu JC, Santi DV. On the mechanism and inhibition of DNA cytosine methyltransferases. Prog Clinic Biol Res 1985; 198: 119–29

    CAS  Google Scholar 

  3. Antequera F, Bird A. Number of CpG islands and genes in human and mouse. Proc Natl Acad Sci U S A 1993; 90: 11995–9

    Article  CAS  PubMed  Google Scholar 

  4. Jeltsch A. Beyond Watson and Crick: DNA methylation and molecular enzymology of DNA methyltransferases. ChemBioChem 2002; 3 (4): 274–93

    Article  CAS  PubMed  Google Scholar 

  5. Rodenhiser D, Mann M. Epigenetics and human disease: translating basic biology into clinical applications. CMAJ 2006; 174 (3): 341–8

    Article  PubMed  Google Scholar 

  6. Holliday R, Pugh JE. DNA modification mechanisms and gene activity during development. Science 1975; 187: 226–32

    Article  CAS  PubMed  Google Scholar 

  7. Ellis L, Atadja PW, Johnstone RW. Epigenetics in cancer: targeting chromatin modifications. Mol Cancer Ther 2009; 8: 1409–20

    Article  CAS  PubMed  Google Scholar 

  8. Bird A. DNA methylation patterns epigenetic memory. Genes Dev 2002; 16: 6–21

    Article  CAS  PubMed  Google Scholar 

  9. Jones PA, Baylin SB. The fundamental role of epigenetic events in cancer. Nat Rev Genet 2002; 3: 415–28

    Article  CAS  PubMed  Google Scholar 

  10. de Caceres II, Battagli C, Esteller M, et al. Tumor cell-specific BRCA1 and RASSF1A hypermethylation in serum, plasma, and peritoneal fluid from ovarian cancer patients. Cancer Res 2004; 64 (18): 6476–81

    Article  Google Scholar 

  11. Esteller M, Corn PG, Baylin SB, et al. A gene hypermethylation profile of human cancer. Cancer Res 2001; 61 (8): 3225–9

    CAS  PubMed  Google Scholar 

  12. Whitman SP, Hackanson B, Liyanarachchi S, et al. DNA hypermethylation and epigenetic silencing of the tumor suppressor gene, SLC5A8, in acute myeloid leukemia with the MLL partial tandem duplication. Blood 2008; 112 (5): 2013–6

    Article  CAS  PubMed  Google Scholar 

  13. Herman JG, Merlo A, Mao L, et al. Inactivation of the CDKN2/p16/MTS1 gene is frequently associated with aberrant DNA methylation in all common human cancers. Cancer Res 1995; 55 (20): 4525–30

    CAS  PubMed  Google Scholar 

  14. Grady WM, Carethers JM. Genomic and epigenetic instability in colorectal cancer pathogenesis. Gastroenterology 2008; 135 (4): 1079–99

    Article  CAS  PubMed  Google Scholar 

  15. Bestor T. The DNA methyltransferases of mammals. Hum Mol Genet 2000; 9: 2395–402

    Article  CAS  PubMed  Google Scholar 

  16. Roll JD, Rivenbark A, Jones W, et al. DNMT3b overexpression contributes to a hypermethylator phenotype in human breast cancer cell lines [abstract]. Mol Cancer 2008; 7: 15

    Article  PubMed  CAS  Google Scholar 

  17. Biniszkiewicz D, Gribnau J, Ramsahoye B, et al. DNMT1 overexpression causes genomic hypermethylation, loss of imprinting, and embryonic lethality. Mol Cell Biol 2002; 22 (7): 2124–35

    Article  CAS  PubMed  Google Scholar 

  18. Lin R-K, Hsu H-S, Chang J-W, et al. Alteration of DNA methyltransferases contributes to 5′CpG methylation and poor prognosis in lung cancer. Lung Cancer 2007; 55 (2): 205–13

    Article  PubMed  Google Scholar 

  19. Bestor TH, Verdine GL. DNA methyltransferases. Curr Opin Cell Biol 1994; 6 (3): 380–9

    Article  CAS  PubMed  Google Scholar 

  20. Christman JK. 5-Azacytidine and 5-aza-2′-deoxycytidine as inhibitors of DNA methylation: mechanistic studies and their implications for cancer therapy. Oncogene 2002; 21: 5483–95

    Article  CAS  PubMed  Google Scholar 

  21. Singal R, Ginder GD. DNA methylation. Blood 1999; 93 (12): 4059–70

    CAS  PubMed  Google Scholar 

  22. Robert M-F, Morin S, Beaulieu N, et al. DNMT1 is required to maintain CpG methylation and aberrant gene silencing in human cancer cells. Nat Genet 2002; 33: 61–5

    Article  PubMed  CAS  Google Scholar 

  23. Hermann A, Gowher H, Jeltsch A. Biochemistry and biology of mammalian DNA methyltransferases. Cell Mol Life Sci 2004; 61 (19): 2571–87

    Article  CAS  PubMed  Google Scholar 

  24. Penterman J, Uzawa R, Fischer RL. Genetic interactions between DNA demethylation and methylation in arabidopsis. Plant Physiol 2007; 145 (4): 1549–57

    Article  CAS  PubMed  Google Scholar 

  25. Wu SC, Zhang Y. Active DNA demethylation: many roads lead to Rome. Nat Rev Mol Cell Biol 2010; 11 (9): 607–20

    Article  CAS  PubMed  Google Scholar 

  26. Ooi SKT, Bestor TH. The colorful history of active DNA demethylation. Cell 2008; 133 (7): 1145–8

    Article  CAS  PubMed  Google Scholar 

  27. Ramchandani S, Bhattacharya SK, Cervoni N, et al. DNA methylation is a reversible biological signal. Proc Nat Acad Sci U S A 1999; 96 (11): 6107–12

    Article  CAS  Google Scholar 

  28. Issa J-PJ. DNA methylation as a therapeutic target in cancer. Clin Cancer Res 2007; 13 (6): 1634–7

    Article  CAS  PubMed  Google Scholar 

  29. Lu Q, Qiu X, Hu N, et al. Epigenetics, disease, and therapeutic interventions. Ageing Res Rev 2006; 5 (4): 449–67

    Article  CAS  PubMed  Google Scholar 

  30. Teodoridis JM, Strathdee G, Brown R. Epigenetic silencing mediated by CpG island methylation: potential as a therapeutic target and as a biomarker. Drug Resist Updat 2004; 7: 267–78

    Article  CAS  PubMed  Google Scholar 

  31. Amatori S, Bagaloni I, Donati B, et al. DNA demethylating antineoplastic strategies. Genes Cancer 2010; 1 (3): 197–209

    Article  CAS  PubMed  Google Scholar 

  32. Cihak A. Biological effect of 5-azacytidine in eukaryotes. Oncology 1974; 30: 405–22

    Article  CAS  PubMed  Google Scholar 

  33. Li LH, Buskirk HH, Reineke LM. Cytotoxicity and mode of action of 5-azacytidine on L1210 leukemia. Cancer Res 1970; 30: 2760–9

    CAS  PubMed  Google Scholar 

  34. Sellis D, Provata A, Almirantis Y. Alu and LINE1 distributions in the human chromosomes: evidence of global genomic organization expressed in the form of power laws. Mol Biol Evol 2007; 24 (11): 2385–99

    Article  CAS  PubMed  Google Scholar 

  35. Mai A, Altucci L. Epi-drugs to fight cancer: from chemistry to cancer treatment, the road ahead. Int J Biochem Cell Biol 2009; 41: 199–213

    Article  CAS  PubMed  Google Scholar 

  36. Kaminskas E, Farrell A, Abraham S, et al. Approval summary: azacitidine for treatment of myelodysplastic syndrome subtypes. Clin Cancer Res 2005; 11: 3604–8

    Article  CAS  PubMed  Google Scholar 

  37. Issa J-PJ, Garcia-Manero G, Giles FJ, et al. Phase 1 study of low-dose prolonged exposure schedules of the hypomethylating agent 5-aza-2′-deoxycytidine (decitabine) in hematopoietic malignancies. Blood 2004; 103 (5): 1635–40

    Article  CAS  PubMed  Google Scholar 

  38. Stresemann C, Lyko F. Modes of action of the DNA methyltransferase inhibitors azacytidine and decitabine. Int J Cancer 2008; 123: 8–13

    Article  CAS  PubMed  Google Scholar 

  39. Piskala A, Som F. Synthesis of 1-glycosyl derivatives of 5-azauracil and 5-azacytosine. Collect Czech Chem Commun 1964; 29: 2060–76

    CAS  Google Scholar 

  40. Biard DSF, Cordier A, Sarasin A. Establishment of a human cell line for the detection of demethylating agents. Exp Cell Res 1992; 200 (2): 263–71

    Article  CAS  PubMed  Google Scholar 

  41. Caterina M, Emanuela SA, Giuseppa P, et al. Epigenetic therapy in myelodysplastic syndromes. Eur J Haematol 2010; 84 (6): 463–73

    Article  CAS  Google Scholar 

  42. Christman JK, Mendelsohn N, Herzog D, et al. Effect of 5-azacytidine on differentiation and DNA methylation in human promyelocyte leukemia cells (HL-60). Cancer Res 1983; 43 (2): 763–9

    CAS  PubMed  Google Scholar 

  43. Borthakur G, El Ahdab S, Ravandi F, et al. Activity of decitabine in patients with myelodysplastic syndrome previously treated with azacitidine. Leuk Lymphoma 2008; 49 (4): 690–5

    Article  CAS  PubMed  Google Scholar 

  44. Creusot F, Acs G, Christman JK. Inhibition of DNA methyltransferase and induction of Friend erythroleukemia cell differentiation by 5-azacytidine and 5-aza-2′-deoxycytidine. J Biol Chem 1982; 257: 2041–8

    CAS  PubMed  Google Scholar 

  45. Lemaire M, Chabot GG, Raynal N, et al. Importance of dose-schedule of 5-aza-2′-deoxycytidine for epigenetic therapy of cancer. BMC Cancer 2008; 8: 1–10

    Article  CAS  Google Scholar 

  46. Patel K, Dickson J, Din S, et al. Targeting of 5-aza-2′-deoxycytidine residues by chromatin-associated DNMT1 induces proteasomal degradation of the free enzyme. Nucl Acids Res 2010; 38: 187–99

    Article  CAS  Google Scholar 

  47. Beumer JH, Eiseman JL, Parise RA, et al. Pharmacokinetics, metabolism, and oral bioavailability of the DNA methyltransferase inhibitor 5-fluoro-2′-deoxycytidine in mice. Cancer Ther Preclin 2006; 12: 7483–91

    CAS  Google Scholar 

  48. Boothman DA, Briggle TV, Greer S. Protective, tumor-selective dual pathway activation of 5-fluoro-2′-deoxycytidine provided by tetrahydrouridine in mice bearing mammary adenocarcinoma-77. Cancer Res 1987; 47: 2344–53

    CAS  PubMed  Google Scholar 

  49. Kaysen J, Spriggs D, Kufe D. Incorporation of 5-fluorodeoxycytidine and metabolites into nucleic acids of human MCF-7 breast carcinoma cells. Cancer Res 1986; 46: 4534–8

    CAS  PubMed  Google Scholar 

  50. Valinluck V, Sowers LC. Endogenous cytosine damage products alter the site selectivity of human DNA maintenance methyltransferase DNMT1. Cancer Res 2007; 67 (3): 946–50

    Article  CAS  PubMed  Google Scholar 

  51. Valinluck V, Liu P, Kang Jr JI, et al. 5-Halogenated pyrimidine lesions within a CpG sequence context mimic 5-methylcytosine by enhancing the binding of the methyl-CpG-binding domain of methyl-CpG-binding protein 2 (MeCP2). Nucl Acids Res 2005; 33 (9): 3057–64

    Article  CAS  PubMed  Google Scholar 

  52. Issa J-PJ, Kantarjian HM. Targeting DNA methylation. Clin Cancer Res 2009; 15 (12): 3938–46

    Article  CAS  PubMed  Google Scholar 

  53. Smith SS, Kaplan BE, Sowers LC, et al. Mechanism of human methyl-directed DNA methyltransferase and the fidelity of cytosine methylation. Proc Nat Acad Sci U S A 1992; 89 (10): 4744–8

    Article  CAS  Google Scholar 

  54. Klimasauskas S, Kumar S, Roberts RJ, et al. HhaI methyltransferase flips its target base out of the DNA helix. Cell 1994; 76: 357–69

    Article  CAS  PubMed  Google Scholar 

  55. Yoo CB, Cheng JC, Jones PA. Zebularine: a new drug for epigenetic therapy. Biochem Soc Trans 2004; 32: 910–2

    Article  CAS  PubMed  Google Scholar 

  56. Yoo CB, Chuang JC, Byun H-M, et al. Long-term epigenetic therapy with oral zebularine has minimal side effects and prevents intestinal tumors in mice. Cancer Prevent Res 2008; 1 (4): 233–40

    Article  CAS  Google Scholar 

  57. Cheng JC, Yoo CB, Weisenberger DJ, et al. Preferential response of cancer cells to zebularine. Cancer Cell 2004; 6 (2): 151–8

    Article  CAS  PubMed  Google Scholar 

  58. Cheng JC, Weisenberger DJ, Gonzales FA, et al. Continuous zebularine treatment effectively sustains demethylation in human bladder cancer cells. Mol Cell Bio 2004; 24: 1270–8

    Article  CAS  Google Scholar 

  59. Votruba I, Holy A, Wightman RH. The mechanism of inhibition of DNA synthesis in Escherichia coli by pyrimidin-2-one beta-D-ribofuranoside. Biochim Biophys Acta 1973; 324: 12–23

    Google Scholar 

  60. Zhou L, Cheng X, Connolly BA, et al. Zebularine: a novel DNA methylation inhibitor that forms a covalent complex with DNA methyltransferases. J Mol Biol 2002; 321: 591-9

    Google Scholar 

  61. Cheng JC, Matsen CB, Gonzales FA, et al. Inhibition of DNA methylation and reactivation of silenced genes by zebularine. J Natl Cancer Inst 2003; 95: 399–409

    Article  CAS  PubMed  Google Scholar 

  62. Kim CH, Marquez VE, Mao DT, et al. Synthesis of pyrimidin-2-one nucleosides as acid-stable inhibitors of cytidine deaminase. J Med Chem 1986; 29 (8): 1374–80

    Article  CAS  PubMed  Google Scholar 

  63. Champion C, Guianvarc’h D, Sénamaud-Beaufort C, et al. Mechanistic insights on the inhibition of C5 DNA methyltransferases by zebularine [abstract]. PLoS ONE 2010; 5 (8): e12388

    Article  PubMed  CAS  Google Scholar 

  64. Kurkjian C, Kummar S, Murgo A. DNA methylation: its role in cancer development and therapy. J Curr Probl Cancer 2008; 32: 187–235

    Article  Google Scholar 

  65. Ben-Kasus T, Ben-Zvi Z, Marquez VE, et al. Metabolic activation of zebularine, a novel DNA methylation inhibitor, in human bladder carcinoma cells. Biochem Pharmacol 2005; 70: 121–33

    Article  CAS  PubMed  Google Scholar 

  66. Mund C, Hackanson Br, Stresemann C, et al. Characterization of DNA demethylation effects induced by 5-aza-2′-deoxycytidine in patients with myelodysplastic syndrome. Cancer Res 2005; 65 (16): 7086–90

    Article  CAS  PubMed  Google Scholar 

  67. Sheikhnejad G, Brank A, Christman JK, et al. Mechanism of inhibition of DNA (cytosine C5)-methyltransferases by oligodeoxyribonucleotides containing 5,6-dihydro-5-azacytosine. J Mol Biol 1999; 285 (5): 2021–34

    Article  CAS  PubMed  Google Scholar 

  68. Jones PA, Taylor SM. Cellular differentiation, cytidine analogs and DNA methylation. Cell 1980; 20: 85–93

    Article  CAS  PubMed  Google Scholar 

  69. Rhee I, Bachman KE, Park BH, et al. DNMT1 and DNMT3b cooperate to silence genes in human cancer cells. Nature 2002; 416 (6880): 552–6

    Article  CAS  PubMed  Google Scholar 

  70. Howell PM, Liu Z, Khong HT. Demethylating agents in the treatment of cancer. Pharmaceuticals 2010; 3 (7): 2022–44

    Article  CAS  Google Scholar 

  71. Daher GC, Harris BE, Diasio RB. Metabolism of pyrimidine analogues and their nucleosides. Pharmacol Ther 1990; 48 (2): 189–222

    Article  CAS  PubMed  Google Scholar 

  72. Palii SS, Van Emburgh BO, Sankpal UT, et al. DNA methylation inhibitor 5-aza-2′-deoxycytidine induces reversible genome-wide DNA damage that is distinctly influenced by DNA methyltransferases 1 and 3B. Mol Cell Biol 2008; 28 (2): 752–71

    Article  CAS  PubMed  Google Scholar 

  73. Davidson S, Crowther P, Radley J, et al. Cytotoxicity of 5-aza-2′-deoxycytidine in a mammalian cell system. Eur J Cancer 1992; 28: 362–8

    Article  CAS  PubMed  Google Scholar 

  74. Yang AS, Doshi KD, Choi S-W, et al. DNA methylation changes after 5-aza-2′-deoxycytidine therapy in patients with leukemia. Cancer Res 2006; 66 (10): 5495–503

    Article  CAS  PubMed  Google Scholar 

  75. Stresemann C, Brueckner B, Musch T, et al. Functional diversity of DNA methyltransferase inhibitors in human cancer cell lines. Cancer Res 2006; 66 (5): 2794–800

    Article  CAS  PubMed  Google Scholar 

  76. Yoo CB, Jones PA. Epigenetic therapy of cancer: past, present and future. Nat Rev Drug Discov 2006; 5 (1): 37–50

    Article  CAS  PubMed  Google Scholar 

  77. Issa J-P. Epigenetic changes in the myelodysplastic syndrome. Hematol Oncol Clin N Am 2010; 24 (2): 317–30

    Article  Google Scholar 

  78. Issa J-PJ, Kantarjian HM, Kirkpatrick P. Azacitidine. Nat Rev Drug Discov 2005; 4 (4): 275–6

    Article  CAS  PubMed  Google Scholar 

  79. Kaminskas E, Farrell AT, Wang Y-C, et al. FDA drug approval summary: azacitidine (5-azacytidine, VidazaTM) for injectable suspension. Oncologist 2005; 10 (3): 176–82

    Article  CAS  PubMed  Google Scholar 

  80. Blum W. How much? How frequent? How long? A clinical guide to new therapies in myelodysplastic syndromes. Hematology 2010; 1: 314–21

    Article  Google Scholar 

  81. Appleton K, Mackay HJ, Judson I, et al. Phase I and pharmacodynamic trial of the DNA methyltransferase inhibitor decitabine and carboplatin in solid tumors. J Clin Oncol 2007; 25 (29): 4603–9

    Article  CAS  PubMed  Google Scholar 

  82. Stewart DJ, Issa J-P, Kurzrock R, et al. Decitabine effect on tumor global DNA methylation and other parameters in a phase I trial in refractory solid tumors and lymphomas. Clin Cancer Res 2009; 15 (11): 3881–8

    Article  CAS  PubMed  Google Scholar 

  83. Cowan LA, Talwar S, Yang AS. Will DNA methylation inhibitors work in solid tumors? A review of the clinical experience with azacitidine and decitabine in solid tumors. Epigenomics 2010; 2 (1): 71–86

    Article  CAS  PubMed  Google Scholar 

  84. O’Reilly EM, Abou-Alfa GK. Comparison of gemcitabine plus platinum analog with gemcitabine alone in advanced pancreatic cancer. Nat Clin Prac Oncol 2008; 5 (6): 312–3

    Google Scholar 

  85. Castellano S, Kuck D, Sala M, et al. Constrained analogues of procaine as novel small molecule inhibitors of DNA methyltransferase-1. J Med Chem 2008; 51 (7): 2321–5

    Article  CAS  PubMed  Google Scholar 

  86. Zambrano P, Segura-Pacheco B, Perez-Cardenas E, et al. A phase I study of hydralazine to demethylate and reactivate the expression of tumor suppressor genes [abstract]. BMC Cancer 2005; 5 (1): 44

    Article  PubMed  CAS  Google Scholar 

  87. Datta J, Ghoshal K, Denny WA, et al. A new class of quinoline-based DNA hypomethylating agents reactivates tumor suppressor genes by blocking DNA methyltransferase 1 activity and inducing its degradation. Cancer Res 2009; 69 (10): 4277–85

    Article  CAS  PubMed  Google Scholar 

  88. Pina IC, Gautschi JT, Wang G-Y-S, et al. Psammaplins from the sponge pseudoceratina purpurea: inhibition of both histone deacetylase and DNA methyltransferase. J Organ Chem 2003; 68 (10): 3866–73

    Article  CAS  Google Scholar 

  89. Cui X, Wakai T, Shirai Y, et al. Arsenic trioxide inhibits DNA methyltransferase and restores methylation-silenced genes in human liver cancer cells. Hum Pathol 2006; 37: 298–311

    Article  CAS  PubMed  Google Scholar 

  90. Suzuki T, Tanaka R, Hamada S, et al. Design, synthesis, inhibitory activity, and binding mode study of novel DNA methyltransferase 1 inhibitors. Bioorg Med Chem Lett 2010; 20 (3): 1124–7

    Article  CAS  PubMed  Google Scholar 

  91. Fang MZ, Wang Y, Ai N, et al. Tea polyphenol (−)-epigallocatechin-3-gallate inhibits DNA methyltransferase and reactivates methylation-silenced genes in cancer cell lines. Cancer Res 2003; 63 (22): 7563–70

    CAS  PubMed  Google Scholar 

  92. Chuang JC, Yoo CB, Kwan JM, et al. Comparison of biological effects of non-nucleoside DNA methylation inhibitors versus 5-aza-2′-deoxycytidine. Mol Cancer Ther 2005; 4 (10): 1515–20

    Article  CAS  PubMed  Google Scholar 

  93. Liu Z, Xie Z, Jones W, et al. Curcumin is a potent DNA hypomethylation agent. Bioorg Med Chem Lett 2009; 19 (3): 706–9

    Article  PubMed  CAS  Google Scholar 

  94. Dhillon N, Aggarwal BB, Newman RA, et al. Phase II trial of curcumin in patients with advanced pancreatic cancer. Clin Cancer Res 2008; 14 (14): 4491–9

    Article  CAS  PubMed  Google Scholar 

  95. Siedlecki P, Boy RG, Musch T, et al. Discovery of two novel, small-molecule inhibitors of DNA methylation. J Med Chem 2005; 49 (2): 678–83

    Article  CAS  Google Scholar 

  96. Brueckner B, Garcia Boy R, Siedlecki P, et al. Epigenetic reactivation of tumor suppressor genes by a novel small-molecule inhibitor of human DNA methyltransferases. Cancer Res 2005; 65 (14): 6305–11

    Article  CAS  PubMed  Google Scholar 

  97. Esteller M. DNA methylation and cancer therapy: new developments and expectations. Curr Opin Oncol 2005; 17 (1): 55–60

    Article  CAS  PubMed  Google Scholar 

  98. Villar-Garea A, Fraga MF, Espada J, et al. Procaine is a DNA-demethylating agent with growth-inhibitory effects in human cancer cells. Cancer Res 2003; 63 (16): 4984–9

    CAS  PubMed  Google Scholar 

  99. Lee BH, Yegnasubramanian S, Lin X, et al. Procainamide is a specific inhibitor of DNA methyltransferase 1. J Biol Chem 2005; 280: 40749–56

    Article  CAS  PubMed  Google Scholar 

  100. Chatterjee K, Parmley WW, Massie B, et al. Oral hydralazine therapy for chronic refractory heart failure. Circulation 1976; 54 (6): 879–83

    Article  CAS  PubMed  Google Scholar 

  101. Cornacchia E, Golbus J, Maybaum J, et al. Hydralazine and procainamide inhibit T cell DNA methylation and induce autoreactivity. J Immunol 1988; 140 (7): 2197–200

    CAS  PubMed  Google Scholar 

  102. Candelaria M, Gallardo-Rincón D, Arce C, et al. A phase II study of epigenetic therapy with hydralazine and magnesium valproate to overcome chemotherapy resistance in refractory solid tumors. Ann Oncol 2007; 18 (9): 1529–38

    Article  CAS  PubMed  Google Scholar 

  103. Song Y, Zhang C. Hydralazine inhibits human cervical cancer cell growth in vitro in association with APC demethylation and re-expression. Cancer Chem Pharmacol 2009; 63 (4): 605–13

    Article  CAS  Google Scholar 

  104. Chavez-Blanco A, Perez-Plasencia C, Perez-Cardenas E, et al. Antineoplastic effects of the DNA methylation inhibitor hydralazine and the histone deacetylase inhibitor valproic acid in cancer cell lines [abstract]. Cancer Cell Int 2006; 6 (1): 2

    Article  PubMed  CAS  Google Scholar 

  105. Amato R. Inhibition of DNA methylation by antisense oligonucleotide MG98 as cancer therapy. Clin Genitourin Cancer 2007; 5 (7): 422–6

    Article  CAS  PubMed  Google Scholar 

  106. Klisovic RB, Stock W, Cataland S, et al. A phase I biological study of MG98, an oligodeoxynucleotide antisense to DNA methyltransferase 1, in patients with high-risk myelodysplasia and acute myeloid leukemia. Clin Cancer Res 2008; 14 (8): 2444–9

    Article  CAS  PubMed  Google Scholar 

  107. Winquist E, Knox J, Ayoub J-P, et al. Phase II trial of DNA methyltransferase 1 inhibition with the antisense oligonucleotide MG98 in patients with metastatic renal carcinoma: a National Cancer Institute of Canada Clinical Trials Group investigational new drug study. Invest New Drugs 2006; 24 (2): 159–67

    Article  CAS  PubMed  Google Scholar 

  108. Stewart DJ, Donehower RC, Eisenhauer EA, et al. A phase I pharmacokinetic and pharmacodynamic study of the DNA methyltransferase 1 inhibitor MG98 administered twice weekly. Ann Oncol 2003; 14 (5): 766–74

    Article  CAS  PubMed  Google Scholar 

  109. Shen Z-X, Chen G-Q, Ni J-H, et al. Use of arsenic trioxide (As2O3) in the treatment of acute promyelocytic leukemia (APL): II. Clinical efficacy and pharmacokinetics in relapsed patients. Blood 1997; 89 (9): 3354–60

    CAS  PubMed  Google Scholar 

  110. Florea A-M, Splettstoesser F, Büsselberg D. Arsenic trioxide (As2O3) induced calcium signals and cytotoxicity in two human cell lines: SY-5Y neuroblastoma and 293 embryonic kidney (HEK). Toxicol Appl Pharmacol 2007; 220 (3): 292–301

    Article  CAS  PubMed  Google Scholar 

  111. Pham NB, Butler MS, Quinn RJ. Isolation of psammaplin A 11′-sulfate and bisaprasin 11′-sulfate from the marine sponge Aplysinella rhax. J Nat Prod 2000; 63 (3): 393–5

    Article  CAS  PubMed  Google Scholar 

  112. Ahn MY, Jung JH, Na YJ, et al. A natural histone deacetylase inhibitor, Psammaplin A, induces cell cycle arrest and apoptosis in human endometrial cancer cells. Gyn Oncol 2008; 108 (1): 27–33

    Article  CAS  Google Scholar 

  113. Simmons TL, Andrianasolo E, McPhail K, et al. Marine natural products as anticancer drugs. Mol Cancer Ther 2005; 4 (2): 333–42

    CAS  PubMed  Google Scholar 

  114. Sigalotti L, Covre A, Fratta E, et al. Epigenetics of human cutaneous melanoma: setting the stage for new therapeutic strategies. J Transl Med 2010; 8 (1): 56

    Article  PubMed  CAS  Google Scholar 

  115. Byun H-M, Choi SH, Laird PW, et al. 2′-Deoxy-N4-[2- (4-nitrophenyl) ethoxycarbonyl]-5-azacytidine: a novel inhibitor of DNA methyltransferase that requires activation by human carboxylesterase 1. Cancer Lett 2008; 266 (2): 238–48

    Article  CAS  PubMed  Google Scholar 

  116. Brueckner B, Rius M, Markelova MR, et al. Delivery of 5-azacytidine to human cancer cells by elaidic acid esterification increases therapeutic drug efficacy. Mol Cancer Ther 2010; 9 (5): 1256–64

    Article  CAS  PubMed  Google Scholar 

  117. Yoo CB, Jeong S, Egger G, et al. Delivery of 5-aza-2′-deoxycytidine to cells using oligodeoxynucleotides. Cancer Res 2007; 67 (13): 6400–8

    Article  CAS  PubMed  Google Scholar 

  118. Chuang JC, Warner SL, Vollmer D, et al. S1 10, a 5-aza-2′-deoxycytidine-containing dinucleotide, is an effective DNA methylation inhibitor in vivo and can reduce tumor growth. Mol Cancer Ther 2010; 9 (5): 1443–50

    Article  CAS  PubMed  Google Scholar 

  119. Kantarjian H, Oki Y, Garcia-Manero G, et al. Results of a randomized study of 3 schedules of low-dose decitabine in higher-risk myelodysplastic syndrome and chronic myelomonocytic leukemia. Blood 2007; 109 (1): 52–7

    Article  CAS  PubMed  Google Scholar 

  120. Kantarjian HM, Issa J-PJ. Decitabine dosing schedules. Sem Hematol 2005; 42: S17–22

    Article  CAS  Google Scholar 

  121. Daskalakis M, Nguyen TT, Nguyen C, et al. Demethylation of a hyper-methylated P15/INK4B gene in patients with myelodysplastic syndrome by 5-aza-2′-deoxycytidine (decitabine) treatment. Blood 2002; 100: 2957–64

    Google Scholar 

  122. Marcucci G, Silverman L, Eller M, et al. Bioavailability of azacitidine subcutaneous versus intravenous in patients with the myelodysplastic syndromes. J Clin Pharmacol 2005; 45 (5): 597–602

    Article  CAS  PubMed  Google Scholar 

  123. Rudek MA, Zhao M, He P, et al. Pharmacokinetics of 5-azacitidine administered with phenylbutyrate in patients with refractory solid tumors or hematologic malignancies. J Clin Oncol 2005; 23 (17): 3906–11

    Article  CAS  PubMed  Google Scholar 

  124. Samlowski WE, Leachman SA, Wade M, et al. Evaluation of a 7-day continuous intravenous infusion of decitabine: inhibition of promoter-specific and global genomic DNA methylation. J Clin Oncol 2005; 23 (17): 3897–905

    Article  CAS  PubMed  Google Scholar 

  125. Odenike JEG, van Besien D, Huo P, et al., University of Chicago Phase II Consortium. Phase II trial of decitabine in myelofibrosis with myeloid metaplasia. 2007 ASCO Annual Meeting Proceedings part I; 2007 June 20. J Clin Oncol 2007; 25: 7008

    Google Scholar 

  126. Chabot GG, Bouchard J, Momparler RL. Kinetics of deamination of 5-aza-2′-deoxycytidine and cytosine arabinoside by human liver cytidine deaminase and its inhibition by 3-deazauridine, thymidine or uracil arabinoside. Biochem Pharmacol 1983; 32 (7): 1327–8

    Article  CAS  PubMed  Google Scholar 

  127. Lemaire M, Momparler L, Raynal N, et al. Inhibition of cytidine deaminase by zebularine enhances the antineoplastic action of 5-aza-2′-deoxycytidine. Cancer Chemother Pharmacol 2009; 63 (3): 411–6

    Article  CAS  PubMed  Google Scholar 

  128. Lavelle D, Saunthararajah Y, Vaitkus K, et al. S110, a novel decitabine dinucleotide, increases fetal hemoglobin levels in baboons (P. anubis) [abstract]. J Transl Med 2010; 8 (1): 92

    Article  PubMed  CAS  Google Scholar 

  129. Pannier AK, Shea LD. Controlled release systems for DNA delivery. Mol Ther 2004; 10 (1): 19–26

    Article  CAS  PubMed  Google Scholar 

  130. Beisler JA. Isolation, characterization, and properties of a labile hydrolysis product of the antitumor nucleoside, 5-azacytidine. J Med Chem 1978; 21: 204–8

    Article  CAS  PubMed  Google Scholar 

  131. Rogstad DK, Herring JL, Theruvathu JA, et al. Chemical decomposition of 5-aza-2′-deoxycytidine (decitabine): kinetic analyses and identification of products by NMR, HPLC, and mass spectrometry. Chem Res Toxicol 2009; 22 (6): 1194–204

    Article  CAS  PubMed  Google Scholar 

  132. Nair LS, Laurencin CT. Biodegradable polymers as biomaterials. Prog Polymer Sci 2007; 32 (8–9): 762–98

    Article  CAS  Google Scholar 

  133. Knop K, Hoogenboom R, Fischer D, et al. Poly (ethylene glycol) in drug delivery: pros and cons as well as potential alternatives. Angew Chem Int Ed Engl 2010; 49 (36): 6288–308

    Article  CAS  PubMed  Google Scholar 

  134. Veronese FM, Pasut G. PEGylation, successful approach to drug delivery. Drug Discov Today 2005; 10 (21): 1451–8

    Article  CAS  PubMed  Google Scholar 

  135. Argemí A, Vega A, Subra-Paternault P, et al. Characterization of azacytidine/poly (1-lactic) acid particles prepared by supercritical antisolvent precipitation. J Pharm Biomed Anal 2009; 50 (5): 847–52

    Article  PubMed  CAS  Google Scholar 

  136. Young JD, Yao SYM, Sun L, et al. Human equilibrative nucleoside transporter (ENT) family of nucleoside and nucleobase transporter proteins. Xenobiotica 2008; 38 (7–8): 995–1021

    Article  CAS  PubMed  Google Scholar 

  137. Baldwin SA, Yao SYM, Hyde RJ, et al. Functional characterization of novel human and mouse equilibrative nucleoside transporters (hENT3 and mENT3) located in intracellular membranes. J Biol Chem 2005; 280 (16): 15880–7

    Article  CAS  PubMed  Google Scholar 

  138. Qin T, Jelinek J, Si J, et al. Mechanisms of resistance to 5-aza-2′-deoxycytidine in human cancer cell lines. Blood 2009; 113: 659–67

    Article  CAS  PubMed  Google Scholar 

  139. Jordan A, Reichard P. Ribonucleotide reductases. Ann Rev Biochem 1998; 67 (1): 71–98

    Article  CAS  PubMed  Google Scholar 

  140. Takeda E, Weber G. Role of ribonucleotide reductase in expression of the neoplastic program. Life Sci 1981; 28 (9): 1007–14

    Article  CAS  PubMed  Google Scholar 

  141. Bouffard DY, Laliberté J, Momparler RL. Kinetic studies on 2′, 2′-difluorodeoxycytidine (gemcitabine) with purified human deoxycytidine kinase and cytidine deaminase. Biochem Pharmacol 1993; 45 (9): 1857–61

    Article  CAS  PubMed  Google Scholar 

  142. Tanaka M, Yoshida S, Saneyoshi M, et al. Utilization of 5-fluoro-2′-deoxyuridine triphosphate and 5-fluoro-2′-deoxycytidine triphosphate in DNA synthesis by DNA polymerases alpha and beta from calf thymus. Cancer Res 1981; 41: 4132–5

    CAS  PubMed  Google Scholar 

  143. Galmarini CM, Mackey JR, Dumontet C. Nucleoside analogues: mechanism of drug resistance and reversal strategies. Leukemia 2001; 15: 875–90

    Article  CAS  PubMed  Google Scholar 

  144. Shichijo S, Yamada A, Sagawa K, et al. Induction of MAGE genes in lymphoid cells by demethylating agent 5-aza-2′-deoxycytidine. Jpn J Cancer Res 1996; 87: 751–6

    Article  CAS  PubMed  Google Scholar 

  145. Szyf M. The role of DNA hypermethylation and demethylation in cancer and cancer therapy. Curr Oncol 2008; 15: 72–5

    Article  CAS  PubMed  Google Scholar 

  146. Agrawal A, Murphy RF, Agrawal DK. DNA methylation in breast and colorectal cancers. Mod Pathol 2007; 20 (7): 711–21

    Article  CAS  PubMed  Google Scholar 

  147. Hsieh CL, Jones PA. Meddling with methylation. Nature Cell Biol 2003; 5: 502–4

    Article  CAS  PubMed  Google Scholar 

  148. Hamm CA, Xie H, Costa FF, et al. Global demethylation of rat chondrosarcoma cells after treatment with 5-aza-2′-deoxycytidine results in increased tumorigenicity [abstract]. PLoS ONE 2009; 4 (12): e8340

    Article  PubMed  CAS  Google Scholar 

  149. Gaudet F, Hodgson JG, Eden A, et al. Induction of tumors in mice by genomic hypomethylation. Science 2003; 300 (5618): 489–92

    Article  CAS  PubMed  Google Scholar 

  150. Kantarjian H, Issa J-PJ, Rosenfeld CS, et al. Decitabine improves patient outcomes in myelodysplastic syndromes. Cancer 2006; 106 (8): 1794–803

    Article  CAS  PubMed  Google Scholar 

  151. Miyamoto K, Ushijima T. Diagnostic and therapeutic applications of epigenetics. Japanese J Clin Oncol 2005; 35 (6): 293–301

    Article  Google Scholar 

  152. Senter PD, Beam KS, Mixan B, et al. Identification and activities of human carboxylesterases for the activation of CPT-11, a clinically approved anti-cancer drug. Bioconjug Chem 2001; 12 (6): 1074–80

    Article  CAS  PubMed  Google Scholar 

  153. Jager S, Jahnke A, Wilmes T, et al. Leukemia targeting ligands isolated from phage display peptide libraries. Leukemia 2007; 21 (3): 411–20

    Article  CAS  PubMed  Google Scholar 

  154. Reece TB, Maxey TS, Kron IL. A prospectus on tissue adhesives. Am J Surg 2001; 182 (2 Suppl. 1): S40–4

    Article  Google Scholar 

  155. Hamidi M, Azadi A, Rafiei P. Hydrogel nanoparticles in drug delivery. Adv Drug Deliv Rev 2008; 60 (15): 1638–49

    Article  CAS  PubMed  Google Scholar 

  156. Blanco MD, Trigo RM, Garcia O, et al. Controlled release of cytarabine from poly (2-hydroxyethyl methacrylate-co-N-vinyl-2-pyrrolidone) hydrogels. J Biomaterials Sci Polymer Ed 1997; 8: 709–19

    Article  CAS  Google Scholar 

  157. Yallapu MM, Gupta BK, Jaggi M, et al. Fabrication of curcumin encapsulated PLGA nanoparticles for improved therapeutic effects in metastatic cancer cells. J Colloid Interface Sci 2010; 351 (1): 19–29

    Article  CAS  PubMed  Google Scholar 

  158. Suri S, Fenniri H, Singh B. Nanotechnology-based drug delivery systems. J Occup Med Toxicol 2007; 2 (1): 16

    Article  PubMed  CAS  Google Scholar 

  159. Liu Y, Pan J, Feng S-S. Nanoparticles of lipid monolayer shell and biodegradable polymer core for controlled release of paclitaxel: effects of surfactants on particles size, characteristics and in vitro performance. Int J Pharm 2010; 395 (1–2): 243–50

    Article  CAS  PubMed  Google Scholar 

  160. Farokhzad OC, Langer R. Impact of nanotechnology on drug delivery. ACS Nano 2009; 3: 16–20

    Article  CAS  PubMed  Google Scholar 

  161. Galmarini CM, Warren G, Kohli E, et al. Polymeric nanogels containing the triphosphate form of cytotoxic nucleoside analogues show antitumor activity against breast and colorectal cancer cell lines. Mol Cancer Ther 2008; 7: 3373–80

    Article  CAS  PubMed  Google Scholar 

  162. Galmarini CM, Mackey JR, Dumontet C. Nucleoside analogues and nucleobases in cancer treatment. Lancet Oncol 2002; 3: 415–24

    Article  CAS  PubMed  Google Scholar 

  163. Thierry B, Al-Ejeh F, Khatri A, et al. Multifunctional core-shell magnetic cisplatin nanocarriers. Chem Commun 2009; 47: 7348–50

    Article  CAS  Google Scholar 

  164. Kircheis R, Blessing T, Brunner S, et al. Tumor targeting with surface-shielded ligand-polycation DNA complexes. J Control Release 2001; 72 (1–3): 165–70

    Article  CAS  PubMed  Google Scholar 

  165. Faraasen S, Vörös J, Csúcs G, et al. Ligand-specific targeting of microspheres to phagocytes by surface modification with poly (L-lysine)-grafted polyethylene glycol) conjugate. Pharm Res 2003; 20 (2): 237–46

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgments

No funding has been received for the preparation of this manuscript. The authors have no conflicts of interest directly related to the content of this review.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Sue Ping Lim.

Rights and permissions

Reprints and permissions

About this article

Cite this article

Lim, S.P., Neilsen, P., Kumar, R. et al. The Application of Delivery Systems for DNA Methyltransferase Inhibitors. BioDrugs 25, 227–242 (2011). https://doi.org/10.2165/11592770-000000000-00000

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.2165/11592770-000000000-00000

Keywords

Navigation