Skip to main content
Log in

DNA Methylation as a Therapeutic Target in Hematologic Disorders: Recent Results in Older Patients with Myelodysplasia and Acute Myeloid Leukemia

  • Published:
International Journal of Hematology Aims and scope Submit manuscript

Abstract

DNA methylation provides a major epigenetic code (besides histone modification) of the lineage- and developmentspecific genes (such as regulators of differentiation in the hematopoietic lineages) that control expression of normal cells. However, DNA methylation is also involved in malignancies because aberrant methylating gene activity occurs during leukemic transformation.Thus, genes such as tumor suppressor genes, growth-regulatory genes, and adhesion molecules are often silenced in various hematopoietic malignancies by epigenetic inactivation via DNA hypermethylation. This inactivation is frequently seen not only in transformed cell lines but also in primary leukemia cells. Because this defect is amenable to reversion by pharmacologic means, agents that inhibit DNA methylation have been developed to specifically target this hypermethylation defect in leukemia and preleukemia cases. The most clinically advanced agents, the azanucleosides 5-azacytidine and 5-aza-2′-deoxycytidine (decitabine), were discovered more than 25 years ago, when their methylation-inhibitory activities, even at low concentrations, became apparent.Although both of these agents, like cytarabine, had been clinically used until then at high doses, the redevelopment of these agents for low-dose schedules has revealed very interesting clinical activities for treating myelodysplasia (MDS) and acute myeloid leukemia (AML). Because these diseases occur mostly in patients over 60 years of age, low-dose schedules with these compounds provide a very promising approach in such patient groups by virtue of their low nonhematologic toxicity profiles. In the present review, we describe the development of treatments that target DNA hypermethylation in MDS and AML, and clinical results are presented. In addition, pharmacologic DNA demethylation may be viewed as a platform for biological modification of malignant cells to become sensitized (or resensitized) to secondary signals, such as differentiating signals (retinoids, vitamin D3) and hormonal signals (eg, estrogen receptor in breast cancer cells, androgen receptor in prostate cancer cells). Finally, an in vitro synergism between the reactivating potency of demethylating agents and inhibitors of histone deacetylation has been tested in several pilot studies of AML and MDS treatment. Finally, gene reactivation by either group of compounds results in therapeutically meaningful reactivation of fetal hemoglobin in patients with severe hemoglobinopathies, extending the therapeutic range of derepressive epigenetic agents to nonmalignant hematopoietic disorders.

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.

Similar content being viewed by others

References

  1. Bird AP. CpG-rich islands and the function of DNA methylation. Nature. 1986;321:209–213.

    Article  CAS  PubMed  Google Scholar 

  2. Cross SH, Bird AP. CpG islands and genes. Curr Opin Genet Dev. 1995;5:309–314.

    Article  CAS  PubMed  Google Scholar 

  3. Gardiner-Garden M, Frommer M. CpG islands in vertebrate genomes. J Mol Biol. 1987;196:261–282.

    Article  CAS  PubMed  Google Scholar 

  4. Graff JR, Herman JG, Myohanen S, Baylin SB,Vertino PM. Mapping patterns of CpG island methylation in normal and neoplastic cells implicates both upstream and downstream regions in de novo methylation. J Biol Chem. 1997;272:22322–22329.

    Article  CAS  PubMed  Google Scholar 

  5. Baylin SB, Herman JG, Graff JR, Vertino PM, Issa JP. Alterations in DNA methylation: a fundamental aspect of neoplasia. Adv Cancer Res. 1998;72:141–196.

    Article  CAS  PubMed  Google Scholar 

  6. Barlow DP. Gametic imprinting in mammals. Science. 1995;270:1610–1613.

    Article  CAS  PubMed  Google Scholar 

  7. Goto T, Monk M. Regulation of X-chromosome inactivation in development in mice and humans. Microbiol Mol Biol Rev. 1998;62:362–378.

    PubMed  PubMed Central  CAS  Google Scholar 

  8. Bird AP, Wolffe AP. Methylation-induced repression: belts, braces, and chromatin. Cell. 1999;99:451–454.

    Article  CAS  PubMed  Google Scholar 

  9. Jones PL, Veenstra GJ, Wade PA, et al. Methylated DNA and MeCP2 recruit histone deacetylase to repress transcription. Nat Genet. 1998;19:187–189.

    Article  CAS  PubMed  Google Scholar 

  10. Jones PL, Wolffe AP. Relationships between chromatin organization and DNA methylation in determining gene expression. Semin Cancer Biol. 1999;9:339–347.

    Article  CAS  PubMed  Google Scholar 

  11. Jaenisch R, Bird A. Epigenetic regulation of gene expression: how the genome integrates intrinsic and environmental signals. Nat Genet. 2003;33(suppl):245–254.

    Article  CAS  PubMed  Google Scholar 

  12. Kouzarides T. Histone methylation in transcriptional control. Curr Opin Genet Dev. 2002;12:198–209.

    Article  CAS  PubMed  Google Scholar 

  13. Pfeifer GP, Steigerwald S, Boehm TLJ, Drahovsky D. DNA methylation levels in acute human leukemia. Cancer Lett. 1988;39:185–192.

    Article  CAS  PubMed  Google Scholar 

  14. Wahlfors J, Hiltunen H, Heinonen E, Hämäläinen E, Alhoionen L, Jänne J. Genomic hypomethylation in human chronic lymphocytic leukemia. Blood. 1992;80:2074–2080.

    PubMed  CAS  Google Scholar 

  15. Lyko F, Stach D, Brenner A, et al. Quantitative analysis of DNA methylation in chronic lymphocytic leukemia patients. Electrophoresis. 2004;25:1–6.

    Article  CAS  Google Scholar 

  16. Lübbert M, Brugger W, Mertelsmann R, Kanz L. Developmental regulation of myeloid gene expression and demethylation during ex vivo culture of peripheral blood progenitor cells. Blood. 1996;87:447–455.

    PubMed  Google Scholar 

  17. Sakashita K, Koike K, Kinoshita T, et al. Dynamic DNA methylation change in the CpG island region of p15 during human myeloid development. J Clin Invest. 2001;108:1195–1204.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Chim CS, Liang R, Kwong YL. Hypermethylation of gene promoters in hematological neoplasia. Hematol Oncol. 2002;20:167–176.

    Article  CAS  PubMed  Google Scholar 

  19. Lehmann U, Brakensiek K, Kreipe H. Role of epigenetic changes in hematological malignancies. Ann Hematol. 2004;83:137–152.

    Article  PubMed  Google Scholar 

  20. Drexler HG. Review of alterations of the cyclin-dependent kinase inhibitor INK4 family genes p15, p16, p18 and p19 in human leukemia-lymphoma cells. Leukemia. 1998;12:845–859.

    Article  CAS  PubMed  Google Scholar 

  21. Lübbert M. Gene silencing of the p15/INK4B cell-cycle inhibitor by hypermethylation: an early or later epigenetic alteration in myelodysplastic syndromes? Leukemia. 2003;17:1762–1764.

    Article  CAS  PubMed  Google Scholar 

  22. Di Croce L, Raker VA, Corsaro M, et al. Methyltransferase recruitment and DNA hypermethylation of target promoters by an oncogenic transcription factor. Science. 2002;295:1079–1082.

    Article  PubMed  Google Scholar 

  23. Momparler RL, Cote S, Eliopoulos N. Pharmacological approach for optimization of the dose schedule of 5-aza-2′-deoxycytidine (decitabine) for the therapy of leukemia. Leukemia. 1997;11(suppl 1):1–6.

    Article  CAS  Google Scholar 

  24. Lübbert M. DNA methylation inhibitors in the treatment of leukemias, myelodysplastic syndromes and hemoglobinopathies: clinical results and possible mechanisms of action. Curr Top Microbiol Immunol. 2000;249:135–164.

    PubMed  Google Scholar 

  25. Miller KB, Kim K, Morrison FS, et al. The evaluation of low-dose cytarabine in the treatment of myelodysplastic syndromes: a phase- III intergroup study. Ann Hematol. 1992;65:162–168.

    Article  CAS  PubMed  Google Scholar 

  26. Silverman LR, Holland JF, Weinberg RS, et al. Effects of treatment with 5-azacytidine on the in vivo and in vitro hematopoiesis in patients with myelodysplastic syndromes. Leukemia. 1993;7(suppl 1):21–29.

    PubMed  Google Scholar 

  27. Silverman LR, Demakos EP, Peterson BL, et al. Randomized controlled trial of azacitidine in patients with the myelodysplastic syn drome:A study of the Cancer and Leukemia Group B. J Clin Oncol. 2002;20:2429–2440.

    Article  CAS  PubMed  Google Scholar 

  28. Kornblith AB, Herndon JE 2nd, Silverman LR, et al. Impact of azacytidine on the quality of life of patients with myelodysplastic syndrome treated in a randomized phase III trial: a Cancer and Leukemia Group B study. J Clin Oncol. 2002;20:2441–2452.

    Article  CAS  PubMed  Google Scholar 

  29. Wijermans PW, Krulder JW, Huijgens PC, Neve P. Continuous infusion of low-dose 5-aza-2′-deoxycytidine in elderly patients with high-risk myelodysplastic syndrome. Leukemia. 1997;11(suppl 1):19–23.

    Article  CAS  Google Scholar 

  30. Wijermans PW, Lübbert M, Verhoef G. Low dose decitabine for elderly high risk MDS patients: who will respond? [abstract]. Blood. 2002;100:96a.

    Article  Google Scholar 

  31. Lübbert M, Wijermans PW, Kunzmann R, et al. Cytogenetic responses in high-risk myelodysplastic syndrome following lowdose treatment with the DNA methylation inhibitor 5-aza-2′- deoxycytidine. Br J Haematol. 2001;114:349–357.

    Article  PubMed  Google Scholar 

  32. Hiddemann W, Kern W, Schoch C, et al. Management of acute myeloid leukemia in elderly patients. J Clin Oncol. 1999;17:3569–3576.

    Article  CAS  PubMed  Google Scholar 

  33. Pinto A, Zagonel V, Attadia V, et al. 5-Aza-2′-deoxycytidine as a differentiation inducer in acute myeloid leukaemias and myelodysplastic syndromes of the elderly. Bone Marrow Transplant. 1989;4(suppl 3):28–32.

    PubMed  Google Scholar 

  34. Issa JP, Garcia-Manero G, Giles FJ, et al. Phase 1 study of lowdose prolonged exposure schedules of the hypomethylating agent 5-aza-2′-deoxycytidine (decitabine) in hematopoietic malignancies. Blood. 2004;103:1635–1640.

    Article  CAS  PubMed  Google Scholar 

  35. Trus MR, Bordeleau LJ, San-Marina S, Minden MD. 5-Aza-2′- deoxycytidine (5-Aza-2-Cdr) reverses CpG methylation of the retinoic acid receptor ′ (RAR ′) promoter and restores retinoid responsiveness in non M3 acute myelogenous leukemia (AML) blasts [abstract]. Blood. 2001;98. Abstract 1925.

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

    Article  PubMed  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Lowrey CH, Nienhuis AW. Brief report: treatment with azacitidine of patients with end-stage β-thalassemia. N Engl J Med. 1993;329:845–848.

    Article  CAS  PubMed  Google Scholar 

  39. Koshy H, Molokie R, Dom L, et al. Augmentation of fetal hemoglobin (HbF) levels by low dose short duration 5-aza-2′-deoxycytidine (decitabine) administration in sickle cell anemia patients who had no HbF elevation following hydroxyurea therapy [abstract]. Blood. 1998;92(suppl 1):306b.

    Google Scholar 

  40. Saunthararajah Y, Hillery CA, Lavelle D, et al. Effects of 5-aza-2′- deoxycytidine on fetal hemoglobin levels, red cell adhesion, and hematopoietic differentiation in patients with sickle cell disease. Blood. 2003;102:3865–3870.

    Article  CAS  PubMed  Google Scholar 

  41. Lavelle D, DeSimone J, Hankewych M, Kousnetzova T, Chen YH. Decitabine induces cell cycle arrest at the G1 phase via p21 (WAF1) and the G2/M phase via the p38 MAP kinase pathway. Leuk Res. 2003;27:999–1007.

    Article  CAS  PubMed  Google Scholar 

  42. Chan AT, Tao Q, Robertson KD, et al. Azacitidine induces demethylation of the Epstein-Barr virus genome in tumors. J Clin Oncol. 2004;22:1373–1381.

    Article  CAS  PubMed  Google Scholar 

  43. Gore SD,Weng LJ, Figg WD, et al. Impact of prolonged infusions of the putative differentiating agent sodium phenylbutyrate on myelodysplastic syndromes and acute myeloid leukemia. Clin Cancer Res. 2002;8:963–970.

    PubMed  CAS  Google Scholar 

  44. Cameron EE, Bachman KE, Myohanen S, Herman JG, Baylin SB. Synergy of demethylation and histone deacetylase inhibition in the re-expression of genes silenced in cancer. Nat Genet. 1999;21:103–107.

    Article  CAS  PubMed  Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Michael Lübbert.

About this article

Cite this article

Rüter, B., Wijermans, P.W. & Lübbert, M. DNA Methylation as a Therapeutic Target in Hematologic Disorders: Recent Results in Older Patients with Myelodysplasia and Acute Myeloid Leukemia. Int J Hematol 80, 128–135 (2004). https://doi.org/10.1532/IJH97.04094

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1532/IJH97.04094

Key words

Navigation