DOI QR코드

DOI QR Code

Use of deferasirox, an iron chelator, to overcome imatinib resistance of chronic myeloid leukemia cells

  • Kim, Dae Sik (Division of Hematology and Oncology, Department of Internal Medicine, Korea University School of Medicine) ;
  • Na, Yoo Jin (Graduate School of Medicine, Korea University School of Medicine) ;
  • Kang, Myoung Hee (Graduate School of Medicine, Korea University School of Medicine) ;
  • Yoon, Soo-Young (Department of Laboratory Medicine, Korea University School of Medicine) ;
  • Choi, Chul Won (Division of Hematology and Oncology, Department of Internal Medicine, Korea University School of Medicine)
  • Received : 2015.02.02
  • Accepted : 2015.03.30
  • Published : 2016.03.01

Abstract

Background/Aims: The treatment of chronic myeloid leukemia (CML) has achieved impressive success since the development of the Bcr-Abl tyrosine kinase inhibitor, imatinib mesylate. Nevertheless, resistance to imatinib has been observed, and a substantial number of patients need alternative treatment strategies. Methods: We have evaluated the effects of deferasirox, an orally active iron chelator, and imatinib on K562 and KU812 human CML cell lines. Imatinib-resistant CML cell lines were created by exposing cells to gradually increasing concentrations of imatinib. Results: Co-treatment of cells with deferasirox and imatinib induced a synergistic dose-dependent inhibition of proliferation of both CML cell lines. Cell cycle analysis showed an accumulation of cells in the subG1 phase. Western blot analysis of apoptotic proteins showed that co-treatment with deferasirox and imatinib induced an increased expression of apoptotic proteins. These tendencies were clearly identified in imatinib-resistant CML cell lines. The results also showed that co-treatment with deferasirox and imatinib reduced the expression of Bcr-Abl, phosphorylated Bcr-Abl, nuclear $factor-{\kappa}B$ ($NF-{\kappa}B$) and ${\beta}-catenin$. Conclusions: We observed synergistic effects of deferasirox and imatinib on both imatinib-resistant and imatinib-sensitive cell lines. These effects were due to induction of apoptosis and cell cycle arrest by down-regulated expression of $NF-{\kappa}B$ and ${\beta}-catenin$ levels. Based on these results, we suggest that a combination treatment of deferasirox and imatinib could be considered as an alternative treatment option for imatinib-resistant CML.

Keywords

Acknowledgement

Supported by : Korea University

References

  1. Faderl S, Talpaz M, Estrov Z, O'Brien S, Kurzrock R, Kantarjian HM. The biology of chronic myeloid leukemia. N Engl J Med 1999;341:164-172. https://doi.org/10.1056/NEJM199907153410306
  2. Buchdunger E, Zimmermann J, Mett H, et al. Inhibition of the Abl protein-tyrosine kinase in vitro and in vivo by a 2-phenylaminopyrimidine derivative. Cancer Res 1996;56:100-104.
  3. Druker BJ, Tamura S, Buchdunger E, et al. Effects of a selective inhibitor of the Abl tyrosine kinase on the growth of Bcr-Abl positive cells. Nat Med 1996;2:561-566. https://doi.org/10.1038/nm0596-561
  4. O'Brien SG, Guilhot F, Larson RA, et al. Imatinib compared with interferon and low-dose cytarabine for newly diagnosed chronic-phase chronic myeloid leukemia. N Engl J Med 2003;348:994-1004. https://doi.org/10.1056/NEJMoa022457
  5. Deininger M, O'Brien SG, Guilhot F, et al. International randomized study of interferon vs STI571 (IRIS) 8-year follow up: sustained survival and low risk for progression or events in patients with newly diagnosed chronic myeloid leukemia in chronic phase (CML-CP) treated with imatinib. Blood 2009;114:1126.
  6. Gambacorti-Passerini CB, Gunby RH, Piazza R, Galietta A, Rostagno R, Scapozza L. Molecular mechanisms of resistance to imatinib in Philadelphia-chromosome-positive leukaemias. Lancet Oncol 2003;4:75-85. https://doi.org/10.1016/S1470-2045(03)00979-3
  7. Karvela M, Helgason GV, Holyoake TL. Mechanisms and novel approaches in overriding tyrosine kinase inhibitor resistance in chronic myeloid leukemia. Expert Rev Anticancer Ther 2012;12:381-392. https://doi.org/10.1586/era.12.10
  8. Tauchi T, Ohyashiki K. Molecular mechanisms of resistance of leukemia to imatinib mesylate. Leuk Res 2004;28 Suppl 1:S39-S45. https://doi.org/10.1016/j.leukres.2003.10.007
  9. Kantarjian H, Shah NP, Hochhaus A, et al. Dasatinib versus imatinib in newly diagnosed chronic-phase chronic myeloid leukemia. N Engl J Med 2010;362:2260-2270. https://doi.org/10.1056/NEJMoa1002315
  10. Saglio G, Kim DW, Issaragrisil S, et al. Nilotinib versus imatinib for newly diagnosed chronic myeloid leukemia. N Engl J Med 2010;362:2251-2259. https://doi.org/10.1056/NEJMoa0912614
  11. Cilloni D, Saglio G. Molecular pathways: BCR-ABL. Clin Cancer Res 2012;18:930-937. https://doi.org/10.1158/1078-0432.CCR-10-1613
  12. Choi JG, Kim JL, Park J, et al. Effects of oral iron chelator deferasirox on human malignant lymphoma cells. Korean J Hematol 2012;47:194-201. https://doi.org/10.5045/kjh.2012.47.3.194
  13. Kim JL, Kang HN, Kang MH, Yoo YA, Kim JS, Choi CW. The oral iron chelator deferasirox induces apoptosis in myeloid leukemia cells by targeting caspase. Acta Haematol 2011;126:241-245. https://doi.org/10.1159/000330608
  14. Le NT, Richardson DR. The role of iron in cell cycle progression and the proliferation of neoplastic cells. Biochim Biophys Acta 2002;1603:31-46.
  15. Kalinowski DS, Richardson DR. The evolution of iron chelators for the treatment of iron overload disease and cancer. Pharmacol Rev 2005;57:547-583. https://doi.org/10.1124/pr.57.4.2
  16. Buss JL, Greene BT, Turner J, Torti FM, Torti SV. Iron chelators in cancer chemotherapy. Curr Top Med Chem 2004;4:1623-1635. https://doi.org/10.2174/1568026043387269
  17. List AF, Baer MR, Steensma DP, et al. Deferasirox reduces serum ferritin and labile plasma iron in RBC transfusion- dependent patients with myelodysplastic syndrome. J Clin Oncol 2012;30:2134-2139. https://doi.org/10.1200/JCO.2010.34.1222
  18. Fukushima T, Kawabata H, Nakamura T, et al. Iron chelation therapy with deferasirox induced complete remission in a patient with chemotherapy-resistant acute monocytic leukemia. Anticancer Res 2011;31:1741-1744.
  19. Messa E, Carturan S, Maffe C, et al. Deferasirox is a powerful NF-kappaB inhibitor in myelodysplastic cells and in leukemia cell lines acting independently from cell iron deprivation by chelation and reactive oxygen species scavenging. Haematologica 2010;95:1308-1316. https://doi.org/10.3324/haematol.2009.016824
  20. Bedford MR, Ford SJ, Horniblow RD, Iqbal TH, Tselepis C. Iron chelation in the treatment of cancer: a new role for deferasirox? J Clin Pharmacol 2013;53:885-891.
  21. Breccia M, Alimena G. Efficacy and safety of deferasirox in myelodysplastic syndromes. Ann Hematol 2013;92:863-870. https://doi.org/10.1007/s00277-013-1703-7
  22. Song S, Christova T, Perusini S, et al. Wnt inhibitor screen reveals iron dependence of $\beta$-catenin signaling in cancers. Cancer Res 2011;71:7628-7639. https://doi.org/10.1158/0008-5472.CAN-11-2745
  23. Chou TC, Talalay P. Quantitative analysis of dose-effect relationships: the combined effects of multiple drugs or enzyme inhibitors. Adv Enzyme Regul 1984;22:27-55. https://doi.org/10.1016/0065-2571(84)90007-4
  24. Chou TC. Drug combination studies and their synergy quantification using the Chou-Talalay method. Cancer Res 2010;70:440-446. https://doi.org/10.1158/0008-5472.CAN-09-1947
  25. Keutgens A, Robert I, Viatour P, Chariot A. Deregulated NF-kappaB activity in haematological malignancies. Biochem Pharmacol 2006;72:1069-1080. https://doi.org/10.1016/j.bcp.2006.06.011
  26. Gasparini C, Celeghini C, Monasta L, Zauli G. NF-${\kappa}B$ pathways in hematological malignancies. Cell Mol Life Sci 2014;71:2083-2102. https://doi.org/10.1007/s00018-013-1545-4
  27. Reuther JY, Reuther GW, Cortez D, Pendergast AM, Baldwin AS Jr. A requirement for NF-kappaB activation in Bcr-Abl-mediated transformation. Genes Dev 1998;12:968-981. https://doi.org/10.1101/gad.12.7.968
  28. Cilloni D, Messa F, Arruga F, et al. The NF-kappaB pathway blockade by the IKK inhibitor PS1145 can overcome imatinib resistance. Leukemia 2006;20:61-67. https://doi.org/10.1038/sj.leu.2403998
  29. Lounnas N, Frelin C, Gonthier N, et al. NF-kappaB inhibition triggers death of imatinib-sensitive and imatinib- resistant chronic myeloid leukemia cells including T315I Bcr-Abl mutants. Int J Cancer 2009;125:308-317. https://doi.org/10.1002/ijc.24294
  30. Wei YL, Liang Y, Xu L, Zhao XY. The antiproliferation effect of berbamine on k562 resistant cells by inhibiting NF-kappaB pathway. Anat Rec (Hoboken) 2009;292:945-950. https://doi.org/10.1002/ar.20924
  31. Hu Y, Swerdlow S, Duffy TM, Weinmann R, Lee FY, Li S. Targeting multiple kinase pathways in leukemic progenitors and stem cells is essential for improved treatment of Ph+ leukemia in mice. Proc Natl Acad Sci U S A 2006;103:16870-16875. https://doi.org/10.1073/pnas.0606509103
  32. Hu Y, Chen Y, Douglas L, Li S. beta-Catenin is essential for survival of leukemic stem cells insensitive to kinase inhibition in mice with BCR-ABL-induced chronic myeloid leukemia. Leukemia 2009;23:109-116. https://doi.org/10.1038/leu.2008.262
  33. Coluccia AM, Vacca A, Dunach M, et al. Bcr-Abl stabilizes beta-catenin in chronic myeloid leukemia through its tyrosine phosphorylation. EMBO J 2007;26:1456-1466. https://doi.org/10.1038/sj.emboj.7601485

Cited by

  1. Iron metabolism and drug resistance in cancer vol.30, pp.5, 2016, https://doi.org/10.1007/s10534-017-0037-7
  2. MPT0B002, a novel microtubule inhibitor, downregulates T315I mutant Bcr-Abl and induces apoptosis of imatinib-resistant chronic myeloid leukemia cells vol.35, pp.4, 2016, https://doi.org/10.1007/s10637-017-0457-9
  3. Ribonucleotide Reductase Inhibitor 3-AP Induces Oncogenic Virus Infected Cell Death and Represses Tumor Growth vol.9, pp.23, 2016, https://doi.org/10.7150/jca.27437
  4. Metformin enhances the cytotoxic effect of nilotinib and overcomes nilotinib resistance in chronic myeloid leukemia cells vol.36, pp.suppl1, 2016, https://doi.org/10.3904/kjim.2019.336