Skip to main content
Top
Published in: BioDrugs 4/2011

01-08-2011 | Review Article

The Application of Delivery Systems for DNA Methyltransferase Inhibitors

Authors: Ms Sue Ping Lim, Paul Neilsen, Raman Kumar, Andrew Abell, David F. Callen

Published in: BioDrugs | Issue 4/2011

Login to get access

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.
Literature
1.
go back to reference Hotchkiss RD. The quantitative separation of purines, pyrimidines, and nucleosides by paper chromatography. J Biol Chem 1948; 175: 315–32PubMed Hotchkiss RD. The quantitative separation of purines, pyrimidines, and nucleosides by paper chromatography. J Biol Chem 1948; 175: 315–32PubMed
2.
go back to reference Wu JC, Santi DV. On the mechanism and inhibition of DNA cytosine methyltransferases. Prog Clinic Biol Res 1985; 198: 119–29 Wu JC, Santi DV. On the mechanism and inhibition of DNA cytosine methyltransferases. Prog Clinic Biol Res 1985; 198: 119–29
3.
go back to reference Antequera F, Bird A. Number of CpG islands and genes in human and mouse. Proc Natl Acad Sci U S A 1993; 90: 11995–9PubMedCrossRef Antequera F, Bird A. Number of CpG islands and genes in human and mouse. Proc Natl Acad Sci U S A 1993; 90: 11995–9PubMedCrossRef
4.
go back to reference Jeltsch A. Beyond Watson and Crick: DNA methylation and molecular enzymology of DNA methyltransferases. ChemBioChem 2002; 3 (4): 274–93PubMedCrossRef Jeltsch A. Beyond Watson and Crick: DNA methylation and molecular enzymology of DNA methyltransferases. ChemBioChem 2002; 3 (4): 274–93PubMedCrossRef
5.
go back to reference Rodenhiser D, Mann M. Epigenetics and human disease: translating basic biology into clinical applications. CMAJ 2006; 174 (3): 341–8PubMedCrossRef Rodenhiser D, Mann M. Epigenetics and human disease: translating basic biology into clinical applications. CMAJ 2006; 174 (3): 341–8PubMedCrossRef
6.
go back to reference Holliday R, Pugh JE. DNA modification mechanisms and gene activity during development. Science 1975; 187: 226–32PubMedCrossRef Holliday R, Pugh JE. DNA modification mechanisms and gene activity during development. Science 1975; 187: 226–32PubMedCrossRef
7.
go back to reference Ellis L, Atadja PW, Johnstone RW. Epigenetics in cancer: targeting chromatin modifications. Mol Cancer Ther 2009; 8: 1409–20PubMedCrossRef Ellis L, Atadja PW, Johnstone RW. Epigenetics in cancer: targeting chromatin modifications. Mol Cancer Ther 2009; 8: 1409–20PubMedCrossRef
9.
go back to reference Jones PA, Baylin SB. The fundamental role of epigenetic events in cancer. Nat Rev Genet 2002; 3: 415–28PubMedCrossRef Jones PA, Baylin SB. The fundamental role of epigenetic events in cancer. Nat Rev Genet 2002; 3: 415–28PubMedCrossRef
10.
go back to reference 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): 6476–81CrossRef 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): 6476–81CrossRef
11.
go back to reference Esteller M, Corn PG, Baylin SB, et al. A gene hypermethylation profile of human cancer. Cancer Res 2001; 61 (8): 3225–9PubMed Esteller M, Corn PG, Baylin SB, et al. A gene hypermethylation profile of human cancer. Cancer Res 2001; 61 (8): 3225–9PubMed
12.
go back to reference 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): 2013–6PubMedCrossRef 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): 2013–6PubMedCrossRef
13.
go back to reference 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): 4525–30PubMed 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): 4525–30PubMed
14.
go back to reference Grady WM, Carethers JM. Genomic and epigenetic instability in colorectal cancer pathogenesis. Gastroenterology 2008; 135 (4): 1079–99PubMedCrossRef Grady WM, Carethers JM. Genomic and epigenetic instability in colorectal cancer pathogenesis. Gastroenterology 2008; 135 (4): 1079–99PubMedCrossRef
16.
go back to reference 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: 15PubMedCrossRef 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: 15PubMedCrossRef
17.
go back to reference 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): 2124–35PubMedCrossRef 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): 2124–35PubMedCrossRef
18.
go back to reference 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): 205–13PubMedCrossRef 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): 205–13PubMedCrossRef
19.
20.
go back to reference 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: 5483–95PubMedCrossRef 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: 5483–95PubMedCrossRef
21.
go back to reference Singal R, Ginder GD. DNA methylation. Blood 1999; 93 (12): 4059–70PubMed Singal R, Ginder GD. DNA methylation. Blood 1999; 93 (12): 4059–70PubMed
22.
go back to reference 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: 61–5PubMedCrossRef 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: 61–5PubMedCrossRef
23.
go back to reference Hermann A, Gowher H, Jeltsch A. Biochemistry and biology of mammalian DNA methyltransferases. Cell Mol Life Sci 2004; 61 (19): 2571–87PubMedCrossRef Hermann A, Gowher H, Jeltsch A. Biochemistry and biology of mammalian DNA methyltransferases. Cell Mol Life Sci 2004; 61 (19): 2571–87PubMedCrossRef
24.
go back to reference Penterman J, Uzawa R, Fischer RL. Genetic interactions between DNA demethylation and methylation in arabidopsis. Plant Physiol 2007; 145 (4): 1549–57PubMedCrossRef Penterman J, Uzawa R, Fischer RL. Genetic interactions between DNA demethylation and methylation in arabidopsis. Plant Physiol 2007; 145 (4): 1549–57PubMedCrossRef
25.
go back to reference Wu SC, Zhang Y. Active DNA demethylation: many roads lead to Rome. Nat Rev Mol Cell Biol 2010; 11 (9): 607–20PubMedCrossRef Wu SC, Zhang Y. Active DNA demethylation: many roads lead to Rome. Nat Rev Mol Cell Biol 2010; 11 (9): 607–20PubMedCrossRef
26.
27.
go back to reference 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): 6107–12CrossRef 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): 6107–12CrossRef
28.
go back to reference Issa J-PJ. DNA methylation as a therapeutic target in cancer. Clin Cancer Res 2007; 13 (6): 1634–7PubMedCrossRef Issa J-PJ. DNA methylation as a therapeutic target in cancer. Clin Cancer Res 2007; 13 (6): 1634–7PubMedCrossRef
29.
go back to reference Lu Q, Qiu X, Hu N, et al. Epigenetics, disease, and therapeutic interventions. Ageing Res Rev 2006; 5 (4): 449–67PubMedCrossRef Lu Q, Qiu X, Hu N, et al. Epigenetics, disease, and therapeutic interventions. Ageing Res Rev 2006; 5 (4): 449–67PubMedCrossRef
30.
go back to reference 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: 267–78PubMedCrossRef 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: 267–78PubMedCrossRef
31.
go back to reference Amatori S, Bagaloni I, Donati B, et al. DNA demethylating antineoplastic strategies. Genes Cancer 2010; 1 (3): 197–209PubMedCrossRef Amatori S, Bagaloni I, Donati B, et al. DNA demethylating antineoplastic strategies. Genes Cancer 2010; 1 (3): 197–209PubMedCrossRef
33.
go back to reference Li LH, Buskirk HH, Reineke LM. Cytotoxicity and mode of action of 5-azacytidine on L1210 leukemia. Cancer Res 1970; 30: 2760–9PubMed Li LH, Buskirk HH, Reineke LM. Cytotoxicity and mode of action of 5-azacytidine on L1210 leukemia. Cancer Res 1970; 30: 2760–9PubMed
34.
go back to reference 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): 2385–99PubMedCrossRef 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): 2385–99PubMedCrossRef
35.
go back to reference Mai A, Altucci L. Epi-drugs to fight cancer: from chemistry to cancer treatment, the road ahead. Int J Biochem Cell Biol 2009; 41: 199–213PubMedCrossRef Mai A, Altucci L. Epi-drugs to fight cancer: from chemistry to cancer treatment, the road ahead. Int J Biochem Cell Biol 2009; 41: 199–213PubMedCrossRef
36.
go back to reference Kaminskas E, Farrell A, Abraham S, et al. Approval summary: azacitidine for treatment of myelodysplastic syndrome subtypes. Clin Cancer Res 2005; 11: 3604–8PubMedCrossRef Kaminskas E, Farrell A, Abraham S, et al. Approval summary: azacitidine for treatment of myelodysplastic syndrome subtypes. Clin Cancer Res 2005; 11: 3604–8PubMedCrossRef
37.
go back to reference 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): 1635–40PubMedCrossRef 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): 1635–40PubMedCrossRef
38.
go back to reference Stresemann C, Lyko F. Modes of action of the DNA methyltransferase inhibitors azacytidine and decitabine. Int J Cancer 2008; 123: 8–13PubMedCrossRef Stresemann C, Lyko F. Modes of action of the DNA methyltransferase inhibitors azacytidine and decitabine. Int J Cancer 2008; 123: 8–13PubMedCrossRef
39.
go back to reference Piskala A, Som F. Synthesis of 1-glycosyl derivatives of 5-azauracil and 5-azacytosine. Collect Czech Chem Commun 1964; 29: 2060–76 Piskala A, Som F. Synthesis of 1-glycosyl derivatives of 5-azauracil and 5-azacytosine. Collect Czech Chem Commun 1964; 29: 2060–76
40.
go back to reference Biard DSF, Cordier A, Sarasin A. Establishment of a human cell line for the detection of demethylating agents. Exp Cell Res 1992; 200 (2): 263–71PubMedCrossRef Biard DSF, Cordier A, Sarasin A. Establishment of a human cell line for the detection of demethylating agents. Exp Cell Res 1992; 200 (2): 263–71PubMedCrossRef
41.
go back to reference Caterina M, Emanuela SA, Giuseppa P, et al. Epigenetic therapy in myelodysplastic syndromes. Eur J Haematol 2010; 84 (6): 463–73CrossRef Caterina M, Emanuela SA, Giuseppa P, et al. Epigenetic therapy in myelodysplastic syndromes. Eur J Haematol 2010; 84 (6): 463–73CrossRef
42.
go back to reference 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): 763–9PubMed 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): 763–9PubMed
43.
go back to reference 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): 690–5PubMedCrossRef 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): 690–5PubMedCrossRef
44.
go back to reference 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: 2041–8PubMed 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: 2041–8PubMed
45.
go back to reference 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: 1–10CrossRef 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: 1–10CrossRef
46.
go back to reference 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: 187–99CrossRef 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: 187–99CrossRef
47.
go back to reference 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 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
48.
go back to reference 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: 2344–53PubMed 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: 2344–53PubMed
49.
go back to reference 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: 4534–8PubMed 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: 4534–8PubMed
50.
go back to reference Valinluck V, Sowers LC. Endogenous cytosine damage products alter the site selectivity of human DNA maintenance methyltransferase DNMT1. Cancer Res 2007; 67 (3): 946–50PubMedCrossRef Valinluck V, Sowers LC. Endogenous cytosine damage products alter the site selectivity of human DNA maintenance methyltransferase DNMT1. Cancer Res 2007; 67 (3): 946–50PubMedCrossRef
51.
go back to reference 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): 3057–64PubMedCrossRef 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): 3057–64PubMedCrossRef
52.
53.
go back to reference 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): 4744–8CrossRef 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): 4744–8CrossRef
54.
go back to reference Klimasauskas S, Kumar S, Roberts RJ, et al. HhaI methyltransferase flips its target base out of the DNA helix. Cell 1994; 76: 357–69PubMedCrossRef Klimasauskas S, Kumar S, Roberts RJ, et al. HhaI methyltransferase flips its target base out of the DNA helix. Cell 1994; 76: 357–69PubMedCrossRef
55.
go back to reference Yoo CB, Cheng JC, Jones PA. Zebularine: a new drug for epigenetic therapy. Biochem Soc Trans 2004; 32: 910–2PubMedCrossRef Yoo CB, Cheng JC, Jones PA. Zebularine: a new drug for epigenetic therapy. Biochem Soc Trans 2004; 32: 910–2PubMedCrossRef
56.
go back to reference 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): 233–40CrossRef 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): 233–40CrossRef
57.
go back to reference Cheng JC, Yoo CB, Weisenberger DJ, et al. Preferential response of cancer cells to zebularine. Cancer Cell 2004; 6 (2): 151–8PubMedCrossRef Cheng JC, Yoo CB, Weisenberger DJ, et al. Preferential response of cancer cells to zebularine. Cancer Cell 2004; 6 (2): 151–8PubMedCrossRef
58.
go back to reference Cheng JC, Weisenberger DJ, Gonzales FA, et al. Continuous zebularine treatment effectively sustains demethylation in human bladder cancer cells. Mol Cell Bio 2004; 24: 1270–8CrossRef Cheng JC, Weisenberger DJ, Gonzales FA, et al. Continuous zebularine treatment effectively sustains demethylation in human bladder cancer cells. Mol Cell Bio 2004; 24: 1270–8CrossRef
59.
go back to reference 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 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
60.
go back to reference 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 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
61.
go back to reference 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: 399–409PubMedCrossRef 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: 399–409PubMedCrossRef
62.
go back to reference 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): 1374–80PubMedCrossRef 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): 1374–80PubMedCrossRef
63.
go back to reference 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): e12388PubMedCrossRef 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): e12388PubMedCrossRef
64.
go back to reference Kurkjian C, Kummar S, Murgo A. DNA methylation: its role in cancer development and therapy. J Curr Probl Cancer 2008; 32: 187–235CrossRef Kurkjian C, Kummar S, Murgo A. DNA methylation: its role in cancer development and therapy. J Curr Probl Cancer 2008; 32: 187–235CrossRef
65.
go back to reference 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: 121–33PubMedCrossRef 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: 121–33PubMedCrossRef
66.
go back to reference 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): 7086–90PubMedCrossRef 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): 7086–90PubMedCrossRef
67.
go back to reference 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): 2021–34PubMedCrossRef 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): 2021–34PubMedCrossRef
68.
go back to reference Jones PA, Taylor SM. Cellular differentiation, cytidine analogs and DNA methylation. Cell 1980; 20: 85–93PubMedCrossRef Jones PA, Taylor SM. Cellular differentiation, cytidine analogs and DNA methylation. Cell 1980; 20: 85–93PubMedCrossRef
69.
go back to reference Rhee I, Bachman KE, Park BH, et al. DNMT1 and DNMT3b cooperate to silence genes in human cancer cells. Nature 2002; 416 (6880): 552–6PubMedCrossRef Rhee I, Bachman KE, Park BH, et al. DNMT1 and DNMT3b cooperate to silence genes in human cancer cells. Nature 2002; 416 (6880): 552–6PubMedCrossRef
70.
go back to reference Howell PM, Liu Z, Khong HT. Demethylating agents in the treatment of cancer. Pharmaceuticals 2010; 3 (7): 2022–44CrossRef Howell PM, Liu Z, Khong HT. Demethylating agents in the treatment of cancer. Pharmaceuticals 2010; 3 (7): 2022–44CrossRef
71.
go back to reference Daher GC, Harris BE, Diasio RB. Metabolism of pyrimidine analogues and their nucleosides. Pharmacol Ther 1990; 48 (2): 189–222PubMedCrossRef Daher GC, Harris BE, Diasio RB. Metabolism of pyrimidine analogues and their nucleosides. Pharmacol Ther 1990; 48 (2): 189–222PubMedCrossRef
72.
go back to reference 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): 752–71PubMedCrossRef 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): 752–71PubMedCrossRef
73.
go back to reference Davidson S, Crowther P, Radley J, et al. Cytotoxicity of 5-aza-2′-deoxycytidine in a mammalian cell system. Eur J Cancer 1992; 28: 362–8PubMedCrossRef Davidson S, Crowther P, Radley J, et al. Cytotoxicity of 5-aza-2′-deoxycytidine in a mammalian cell system. Eur J Cancer 1992; 28: 362–8PubMedCrossRef
74.
go back to reference 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): 5495–503PubMedCrossRef 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): 5495–503PubMedCrossRef
75.
go back to reference Stresemann C, Brueckner B, Musch T, et al. Functional diversity of DNA methyltransferase inhibitors in human cancer cell lines. Cancer Res 2006; 66 (5): 2794–800PubMedCrossRef Stresemann C, Brueckner B, Musch T, et al. Functional diversity of DNA methyltransferase inhibitors in human cancer cell lines. Cancer Res 2006; 66 (5): 2794–800PubMedCrossRef
76.
go back to reference Yoo CB, Jones PA. Epigenetic therapy of cancer: past, present and future. Nat Rev Drug Discov 2006; 5 (1): 37–50PubMedCrossRef Yoo CB, Jones PA. Epigenetic therapy of cancer: past, present and future. Nat Rev Drug Discov 2006; 5 (1): 37–50PubMedCrossRef
77.
go back to reference Issa J-P. Epigenetic changes in the myelodysplastic syndrome. Hematol Oncol Clin N Am 2010; 24 (2): 317–30CrossRef Issa J-P. Epigenetic changes in the myelodysplastic syndrome. Hematol Oncol Clin N Am 2010; 24 (2): 317–30CrossRef
78.
79.
go back to reference Kaminskas E, Farrell AT, Wang Y-C, et al. FDA drug approval summary: azacitidine (5-azacytidine, VidazaTM) for injectable suspension. Oncologist 2005; 10 (3): 176–82PubMedCrossRef Kaminskas E, Farrell AT, Wang Y-C, et al. FDA drug approval summary: azacitidine (5-azacytidine, VidazaTM) for injectable suspension. Oncologist 2005; 10 (3): 176–82PubMedCrossRef
80.
go back to reference Blum W. How much? How frequent? How long? A clinical guide to new therapies in myelodysplastic syndromes. Hematology 2010; 1: 314–21CrossRef Blum W. How much? How frequent? How long? A clinical guide to new therapies in myelodysplastic syndromes. Hematology 2010; 1: 314–21CrossRef
81.
go back to reference 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): 4603–9PubMedCrossRef 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): 4603–9PubMedCrossRef
82.
go back to reference 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): 3881–8PubMedCrossRef 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): 3881–8PubMedCrossRef
83.
go back to reference 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): 71–86PubMedCrossRef 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): 71–86PubMedCrossRef
84.
go back to reference 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 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
85.
go back to reference 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): 2321–5PubMedCrossRef 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): 2321–5PubMedCrossRef
86.
go back to reference 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): 44PubMedCrossRef 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): 44PubMedCrossRef
87.
go back to reference 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): 4277–85PubMedCrossRef 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): 4277–85PubMedCrossRef
88.
go back to reference 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): 3866–73CrossRef 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): 3866–73CrossRef
89.
go back to reference 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: 298–311PubMedCrossRef 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: 298–311PubMedCrossRef
90.
go back to reference 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): 1124–7PubMedCrossRef 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): 1124–7PubMedCrossRef
91.
go back to reference 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): 7563–70PubMed 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): 7563–70PubMed
92.
go back to reference 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): 1515–20PubMedCrossRef 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): 1515–20PubMedCrossRef
93.
go back to reference Liu Z, Xie Z, Jones W, et al. Curcumin is a potent DNA hypomethylation agent. Bioorg Med Chem Lett 2009; 19 (3): 706–9PubMedCrossRef Liu Z, Xie Z, Jones W, et al. Curcumin is a potent DNA hypomethylation agent. Bioorg Med Chem Lett 2009; 19 (3): 706–9PubMedCrossRef
94.
go back to reference 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): 4491–9PubMedCrossRef 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): 4491–9PubMedCrossRef
95.
go back to reference Siedlecki P, Boy RG, Musch T, et al. Discovery of two novel, small-molecule inhibitors of DNA methylation. J Med Chem 2005; 49 (2): 678–83CrossRef Siedlecki P, Boy RG, Musch T, et al. Discovery of two novel, small-molecule inhibitors of DNA methylation. J Med Chem 2005; 49 (2): 678–83CrossRef
96.
go back to reference 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): 6305–11PubMedCrossRef 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): 6305–11PubMedCrossRef
97.
go back to reference Esteller M. DNA methylation and cancer therapy: new developments and expectations. Curr Opin Oncol 2005; 17 (1): 55–60PubMedCrossRef Esteller M. DNA methylation and cancer therapy: new developments and expectations. Curr Opin Oncol 2005; 17 (1): 55–60PubMedCrossRef
98.
go back to reference 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): 4984–9PubMed 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): 4984–9PubMed
99.
go back to reference Lee BH, Yegnasubramanian S, Lin X, et al. Procainamide is a specific inhibitor of DNA methyltransferase 1. J Biol Chem 2005; 280: 40749–56PubMedCrossRef Lee BH, Yegnasubramanian S, Lin X, et al. Procainamide is a specific inhibitor of DNA methyltransferase 1. J Biol Chem 2005; 280: 40749–56PubMedCrossRef
100.
go back to reference Chatterjee K, Parmley WW, Massie B, et al. Oral hydralazine therapy for chronic refractory heart failure. Circulation 1976; 54 (6): 879–83PubMedCrossRef Chatterjee K, Parmley WW, Massie B, et al. Oral hydralazine therapy for chronic refractory heart failure. Circulation 1976; 54 (6): 879–83PubMedCrossRef
101.
go back to reference Cornacchia E, Golbus J, Maybaum J, et al. Hydralazine and procainamide inhibit T cell DNA methylation and induce autoreactivity. J Immunol 1988; 140 (7): 2197–200PubMed Cornacchia E, Golbus J, Maybaum J, et al. Hydralazine and procainamide inhibit T cell DNA methylation and induce autoreactivity. J Immunol 1988; 140 (7): 2197–200PubMed
102.
go back to reference 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): 1529–38PubMedCrossRef 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): 1529–38PubMedCrossRef
103.
go back to reference 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): 605–13CrossRef 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): 605–13CrossRef
104.
go back to reference 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): 2PubMedCrossRef 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): 2PubMedCrossRef
105.
go back to reference Amato R. Inhibition of DNA methylation by antisense oligonucleotide MG98 as cancer therapy. Clin Genitourin Cancer 2007; 5 (7): 422–6PubMedCrossRef Amato R. Inhibition of DNA methylation by antisense oligonucleotide MG98 as cancer therapy. Clin Genitourin Cancer 2007; 5 (7): 422–6PubMedCrossRef
106.
go back to reference 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): 2444–9PubMedCrossRef 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): 2444–9PubMedCrossRef
107.
go back to reference 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): 159–67PubMedCrossRef 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): 159–67PubMedCrossRef
108.
go back to reference 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): 766–74PubMedCrossRef 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): 766–74PubMedCrossRef
109.
go back to reference 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): 3354–60PubMed 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): 3354–60PubMed
110.
go back to reference 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): 292–301PubMedCrossRef 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): 292–301PubMedCrossRef
111.
go back to reference 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): 393–5PubMedCrossRef 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): 393–5PubMedCrossRef
112.
go back to reference 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): 27–33CrossRef 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): 27–33CrossRef
113.
go back to reference Simmons TL, Andrianasolo E, McPhail K, et al. Marine natural products as anticancer drugs. Mol Cancer Ther 2005; 4 (2): 333–42PubMed Simmons TL, Andrianasolo E, McPhail K, et al. Marine natural products as anticancer drugs. Mol Cancer Ther 2005; 4 (2): 333–42PubMed
114.
go back to reference 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): 56PubMedCrossRef 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): 56PubMedCrossRef
115.
go back to reference 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): 238–48PubMedCrossRef 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): 238–48PubMedCrossRef
116.
go back to reference 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): 1256–64PubMedCrossRef 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): 1256–64PubMedCrossRef
117.
go back to reference Yoo CB, Jeong S, Egger G, et al. Delivery of 5-aza-2′-deoxycytidine to cells using oligodeoxynucleotides. Cancer Res 2007; 67 (13): 6400–8PubMedCrossRef Yoo CB, Jeong S, Egger G, et al. Delivery of 5-aza-2′-deoxycytidine to cells using oligodeoxynucleotides. Cancer Res 2007; 67 (13): 6400–8PubMedCrossRef
118.
go back to reference 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): 1443–50PubMedCrossRef 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): 1443–50PubMedCrossRef
119.
go back to reference 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): 52–7PubMedCrossRef 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): 52–7PubMedCrossRef
120.
go back to reference Kantarjian HM, Issa J-PJ. Decitabine dosing schedules. Sem Hematol 2005; 42: S17–22CrossRef Kantarjian HM, Issa J-PJ. Decitabine dosing schedules. Sem Hematol 2005; 42: S17–22CrossRef
121.
go back to reference 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 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
122.
go back to reference 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): 597–602PubMedCrossRef 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): 597–602PubMedCrossRef
123.
go back to reference 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): 3906–11PubMedCrossRef 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): 3906–11PubMedCrossRef
124.
go back to reference 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): 3897–905PubMedCrossRef 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): 3897–905PubMedCrossRef
125.
go back to reference 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 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
126.
go back to reference 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): 1327–8PubMedCrossRef 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): 1327–8PubMedCrossRef
127.
go back to reference 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): 411–6PubMedCrossRef 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): 411–6PubMedCrossRef
128.
go back to reference 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): 92PubMedCrossRef 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): 92PubMedCrossRef
129.
130.
go back to reference Beisler JA. Isolation, characterization, and properties of a labile hydrolysis product of the antitumor nucleoside, 5-azacytidine. J Med Chem 1978; 21: 204–8PubMedCrossRef Beisler JA. Isolation, characterization, and properties of a labile hydrolysis product of the antitumor nucleoside, 5-azacytidine. J Med Chem 1978; 21: 204–8PubMedCrossRef
131.
go back to reference 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): 1194–204PubMedCrossRef 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): 1194–204PubMedCrossRef
132.
go back to reference Nair LS, Laurencin CT. Biodegradable polymers as biomaterials. Prog Polymer Sci 2007; 32 (8–9): 762–98CrossRef Nair LS, Laurencin CT. Biodegradable polymers as biomaterials. Prog Polymer Sci 2007; 32 (8–9): 762–98CrossRef
133.
go back to reference 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): 6288–308PubMedCrossRef 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): 6288–308PubMedCrossRef
134.
go back to reference Veronese FM, Pasut G. PEGylation, successful approach to drug delivery. Drug Discov Today 2005; 10 (21): 1451–8PubMedCrossRef Veronese FM, Pasut G. PEGylation, successful approach to drug delivery. Drug Discov Today 2005; 10 (21): 1451–8PubMedCrossRef
135.
go back to reference 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): 847–52PubMedCrossRef 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): 847–52PubMedCrossRef
136.
go back to reference 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): 995–1021PubMedCrossRef 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): 995–1021PubMedCrossRef
137.
go back to reference 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): 15880–7PubMedCrossRef 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): 15880–7PubMedCrossRef
138.
go back to reference Qin T, Jelinek J, Si J, et al. Mechanisms of resistance to 5-aza-2′-deoxycytidine in human cancer cell lines. Blood 2009; 113: 659–67PubMedCrossRef Qin T, Jelinek J, Si J, et al. Mechanisms of resistance to 5-aza-2′-deoxycytidine in human cancer cell lines. Blood 2009; 113: 659–67PubMedCrossRef
139.
140.
go back to reference Takeda E, Weber G. Role of ribonucleotide reductase in expression of the neoplastic program. Life Sci 1981; 28 (9): 1007–14PubMedCrossRef Takeda E, Weber G. Role of ribonucleotide reductase in expression of the neoplastic program. Life Sci 1981; 28 (9): 1007–14PubMedCrossRef
141.
go back to reference 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): 1857–61PubMedCrossRef 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): 1857–61PubMedCrossRef
142.
go back to reference 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: 4132–5PubMed 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: 4132–5PubMed
143.
go back to reference Galmarini CM, Mackey JR, Dumontet C. Nucleoside analogues: mechanism of drug resistance and reversal strategies. Leukemia 2001; 15: 875–90PubMedCrossRef Galmarini CM, Mackey JR, Dumontet C. Nucleoside analogues: mechanism of drug resistance and reversal strategies. Leukemia 2001; 15: 875–90PubMedCrossRef
144.
go back to reference 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: 751–6PubMedCrossRef 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: 751–6PubMedCrossRef
145.
go back to reference Szyf M. The role of DNA hypermethylation and demethylation in cancer and cancer therapy. Curr Oncol 2008; 15: 72–5PubMedCrossRef Szyf M. The role of DNA hypermethylation and demethylation in cancer and cancer therapy. Curr Oncol 2008; 15: 72–5PubMedCrossRef
146.
go back to reference Agrawal A, Murphy RF, Agrawal DK. DNA methylation in breast and colorectal cancers. Mod Pathol 2007; 20 (7): 711–21PubMedCrossRef Agrawal A, Murphy RF, Agrawal DK. DNA methylation in breast and colorectal cancers. Mod Pathol 2007; 20 (7): 711–21PubMedCrossRef
148.
go back to reference 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): e8340PubMedCrossRef 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): e8340PubMedCrossRef
149.
go back to reference Gaudet F, Hodgson JG, Eden A, et al. Induction of tumors in mice by genomic hypomethylation. Science 2003; 300 (5618): 489–92PubMedCrossRef Gaudet F, Hodgson JG, Eden A, et al. Induction of tumors in mice by genomic hypomethylation. Science 2003; 300 (5618): 489–92PubMedCrossRef
150.
go back to reference Kantarjian H, Issa J-PJ, Rosenfeld CS, et al. Decitabine improves patient outcomes in myelodysplastic syndromes. Cancer 2006; 106 (8): 1794–803PubMedCrossRef Kantarjian H, Issa J-PJ, Rosenfeld CS, et al. Decitabine improves patient outcomes in myelodysplastic syndromes. Cancer 2006; 106 (8): 1794–803PubMedCrossRef
151.
go back to reference Miyamoto K, Ushijima T. Diagnostic and therapeutic applications of epigenetics. Japanese J Clin Oncol 2005; 35 (6): 293–301CrossRef Miyamoto K, Ushijima T. Diagnostic and therapeutic applications of epigenetics. Japanese J Clin Oncol 2005; 35 (6): 293–301CrossRef
152.
go back to reference 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): 1074–80PubMedCrossRef 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): 1074–80PubMedCrossRef
153.
go back to reference Jager S, Jahnke A, Wilmes T, et al. Leukemia targeting ligands isolated from phage display peptide libraries. Leukemia 2007; 21 (3): 411–20PubMedCrossRef Jager S, Jahnke A, Wilmes T, et al. Leukemia targeting ligands isolated from phage display peptide libraries. Leukemia 2007; 21 (3): 411–20PubMedCrossRef
154.
go back to reference Reece TB, Maxey TS, Kron IL. A prospectus on tissue adhesives. Am J Surg 2001; 182 (2 Suppl. 1): S40–4CrossRef Reece TB, Maxey TS, Kron IL. A prospectus on tissue adhesives. Am J Surg 2001; 182 (2 Suppl. 1): S40–4CrossRef
155.
go back to reference Hamidi M, Azadi A, Rafiei P. Hydrogel nanoparticles in drug delivery. Adv Drug Deliv Rev 2008; 60 (15): 1638–49PubMedCrossRef Hamidi M, Azadi A, Rafiei P. Hydrogel nanoparticles in drug delivery. Adv Drug Deliv Rev 2008; 60 (15): 1638–49PubMedCrossRef
156.
go back to reference 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: 709–19CrossRef 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: 709–19CrossRef
157.
go back to reference 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): 19–29PubMedCrossRef 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): 19–29PubMedCrossRef
158.
go back to reference Suri S, Fenniri H, Singh B. Nanotechnology-based drug delivery systems. J Occup Med Toxicol 2007; 2 (1): 16PubMedCrossRef Suri S, Fenniri H, Singh B. Nanotechnology-based drug delivery systems. J Occup Med Toxicol 2007; 2 (1): 16PubMedCrossRef
159.
go back to reference 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): 243–50PubMedCrossRef 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): 243–50PubMedCrossRef
160.
161.
go back to reference 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: 3373–80PubMedCrossRef 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: 3373–80PubMedCrossRef
162.
go back to reference Galmarini CM, Mackey JR, Dumontet C. Nucleoside analogues and nucleobases in cancer treatment. Lancet Oncol 2002; 3: 415–24PubMedCrossRef Galmarini CM, Mackey JR, Dumontet C. Nucleoside analogues and nucleobases in cancer treatment. Lancet Oncol 2002; 3: 415–24PubMedCrossRef
163.
go back to reference Thierry B, Al-Ejeh F, Khatri A, et al. Multifunctional core-shell magnetic cisplatin nanocarriers. Chem Commun 2009; 47: 7348–50CrossRef Thierry B, Al-Ejeh F, Khatri A, et al. Multifunctional core-shell magnetic cisplatin nanocarriers. Chem Commun 2009; 47: 7348–50CrossRef
164.
go back to reference Kircheis R, Blessing T, Brunner S, et al. Tumor targeting with surface-shielded ligand-polycation DNA complexes. J Control Release 2001; 72 (1–3): 165–70PubMedCrossRef Kircheis R, Blessing T, Brunner S, et al. Tumor targeting with surface-shielded ligand-polycation DNA complexes. J Control Release 2001; 72 (1–3): 165–70PubMedCrossRef
165.
go back to reference 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): 237–46PubMedCrossRef 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): 237–46PubMedCrossRef
Metadata
Title
The Application of Delivery Systems for DNA Methyltransferase Inhibitors
Authors
Ms Sue Ping Lim
Paul Neilsen
Raman Kumar
Andrew Abell
David F. Callen
Publication date
01-08-2011
Publisher
Springer International Publishing
Published in
BioDrugs / Issue 4/2011
Print ISSN: 1173-8804
Electronic ISSN: 1179-190X
DOI
https://doi.org/10.2165/11592770-000000000-00000

Other articles of this Issue 4/2011

BioDrugs 4/2011 Go to the issue

Adis Drug Evaluation

Corifollitropin Alfa