全部 标题 作者
关键词 摘要

OALib Journal期刊
ISSN: 2333-9721
费用:99美元

查看量下载量

相关文章

更多...

The Interface between BCR-ABL-Dependent and -Independent Resistance Signaling Pathways in Chronic Myeloid Leukemia

DOI: 10.1155/2012/671702

Full-Text   Cite this paper   Add to My Lib

Abstract:

Chronic myeloid leukemia (CML) is a clonal hematopoietic disorder characterized by the presence of the Philadelphia chromosome which resulted from the reciprocal translocation between chromosomes 9 and 22. The pathogenesis of CML involves the constitutive activation of the BCR-ABL tyrosine kinase, which governs malignant disease by activating multiple signal transduction pathways. The BCR-ABL kinase inhibitor, imatinib, is the front-line treatment for CML, but the emergence of imatinib resistance and other tyrosine kinase inhibitors (TKIs) has called attention for additional resistance mechanisms and has led to the search for alternative drug treatments. In this paper, we discuss our current understanding of mechanisms, related or unrelated to BCR-ABL, which have been shown to account for chemoresistance and treatment failure. We focus on the potential role of the influx and efflux transporters, the inhibitor of apoptosis proteins, and transcription factor-mediated signals as feasible molecular targets to overcome the development of TKIs resistance in CML. 1. Introduction Chronic myeloid leukemia (CML) is a myeloproliferative disorder that results from the reciprocal translocation of the ABL1 oncogene on chromosome 9 with the breakpoint cluster region (BCR) gene on chromosome 22 [t(9; 22)], leading to the formation of the BCR-ABL oncoprotein. The shortened chromosome 22 formed by this translocation is the Philadelphia (Ph) chromosome. The BCR-ABL fusion oncogene, which is responsible for the pathogenesis of CML, has greatly enhanced ABL1 tyrosine kinase constitutive activity [1]. CML is characterized by a biphasic evolutive course. Most patients are diagnosed in the chronic phase (CML-CP), which is characterized by the absence of symptoms in half of the patients. However, a prominent leukocytosis is frequently observed by routine testing. In the other half of patients, symptoms are common and include splenomegaly, weight loss, lethargy, and anemia [2]. The disease may progress either directly to blast phase (BP) or through an intermediate accelerated phase (AP). The time course for progression to BP is variable and the molecular mechanisms underlying disease progression are extremely complex. BCR-ABL-dependent pathways to blast transformation include an increase in genomic instability, telomere shortening, loss of tumor-suppressor function, and inhibition of tumor suppressors with cell regulatory functions [2, 3]. In order to identify prognostic factors for CML patients, many clinical and biological characteristics have been analyzed. Sokal risk score

References

[1]  J. M. Goldman and J. V. Melo, “Mechanisms of disease: chronic myeloid leukemia—advances in biology and new approaches to treatment,” New England Journal of Medicine, vol. 349, no. 15, pp. 1451–1464, 2003.
[2]  D. Perrotti, C. Jamieson, J. Goldman, and T. Skorski, “Chronic myeloid leukemia: mechanisms of blastic transformation,” Journal of Clinical Investigation, vol. 120, no. 7, pp. 2254–2264, 2010.
[3]  J. V. Melo and D. J. Barnes, “Chronic myeloid leukaemia as a model of disease evolution in human cancer,” Nature Reviews Cancer, vol. 7, no. 6, pp. 441–453, 2007.
[4]  J. C. Hernández-Boluda, B. Bellosillo, M. C. Vela, D. Colomer, A. Alvarez-Larrán, and F. Cervantes, “Survivin expression in the progression of chronic myeloid leukemia: a sequential study in 16 patients,” Leukemia and Lymphoma, vol. 46, no. 5, pp. 717–722, 2005.
[5]  R. Hehlmann, H. Heimpel, J. Hasford et al., “Randomized comparison of busulfan and hydroxyurea in chronic myelogenous leukemia: prolongation of survival by hydroxyurea,” Blood, vol. 82, no. 2, pp. 398–407, 1993.
[6]  F. Bonifazi, A. de Vivo, G. Rosti et al., “Chronic myeloid leukemia and interferon-α: a study of complete cytogenetic responders,” Blood, vol. 98, no. 10, pp. 3074–3081, 2001.
[7]  A. Gratwohl, R. Brand, J. Apperley et al., “Allogeneic hematopoietic stem cell transplantation for chronic myeloid leukemia in Europe 2006: transplant activity, long-term data and current results. An analysis by the chronic leukemia working party of the European Group for Blood and Marrow Transplantation (EBMT),” Haematologica, vol. 91, no. 4, pp. 513–521, 2006.
[8]  T. Schindler, W. Bornmann, P. Pellicena, W. T. Miller, B. Clarkson, and J. Kuriyan, “Structural mechanism for STI-571 inhibition of abelson tyrosine kinase,” Science, vol. 289, no. 5486, pp. 1938–1942, 2000.
[9]  H. M. Kantarjian, J. E. Cortes, S. O'Brien et al., “Long-term survival benefit and improved complete cytogenetic and molecular response rates with imatinib mesylate in Philadelphia chromosome-positive chronic-phase chronic myeloid leukemia after failure of interferon-α,” Blood, vol. 104, no. 7, pp. 1979–1988, 2004.
[10]  J. Pinilla-Ibarz, J. Cortes, and M. J. Mauro, “Intolerance to tyrosine kinase inhibitors in chronic myeloid leukemia,” Cancer, vol. 117, no. 4, pp. 688–697, 2011.
[11]  M. Baccarani, G. Saglio, J. Goldman et al., “Evolving concepts in the management of chronic myeloid leukemia: recommendations from an expert panel on behalf of the European LeukemiaNet,” Blood, vol. 108, no. 6, pp. 1809–1820, 2006.
[12]  G. Saglio, H. Kantarjian, T. Holyoake, A. Ranganathan, and J. E. Cortés, “Proceedings of the third global workshop on chronic myeloid leukemia,” Clinical Lymphoma, Myeloma and Leukemia, vol. 10, no. 6, pp. 443–451, 2010.
[13]  M. Breccia, F. Efficace, and G. Alimena, “Imatinib treatment in chronic myelogenous leukemia: what have we learned so far?” Cancer Letters, vol. 300, no. 2, pp. 115–121, 2011.
[14]  A. M. Eiring, J. S. Khorashad, K. Morley, and M. W. Deininger, “Advances in the treatment of chronic myeloid leukemia,” BMC Medicine, vol. 9, article 99, 2011.
[15]  J. V. Melo and C. Chuah, “Resistance to imatinib mesylate in chronic myeloid leukaemia,” Cancer Letters, vol. 249, no. 2, pp. 121–132, 2007.
[16]  D. Bixby and M. Talpaz, “Mechanisms of resistance to tyrosine kinase inhibitors in chronic myeloid leukemia and recent therapeutic strategies to overcome resistance,” Hematology, pp. 461–476, 2009.
[17]  A. Quintás-Cardama and J. Cortes, “Molecular biology of BCR-ABL1-positive chronic myeloid leukemia,” Blood, vol. 113, no. 8, pp. 1619–1630, 2009.
[18]  S. Roychowdhury and M. Talpaz, “Managing resistance in chronic myeloid leukemia,” Blood Reviews, vol. 25, no. 6, pp. 279–290, 2011.
[19]  K. Eechoute, A. Sparreboom, H. Burger et al., “Drug transporters and imatinib treatment: Implications for clinical practice,” Clinical Cancer Research, vol. 17, no. 3, pp. 406–415, 2011.
[20]  S. V. Ambudkar, S. Dey, C. A. Hrycyna, M. Ramachandra, I. Pastan, and M. M. Gottesman, “Biochemical, cellular, and pharmacological aspects of the multidrug transporter,” Annual Review of Pharmacology and Toxicology, vol. 39, pp. 361–398, 1999.
[21]  G. D. Kruh, “Introduction to resistance to anticancer agents,” Oncogene, vol. 22, no. 47, pp. 7262–7264, 2003.
[22]  N. Kartner, D. Evernden-Porelle, G. Bradley, and V. Ling, “Detection of P-glycoprotein in multidrug-resistant cell lines by monoclonal antibodies,” Nature, vol. 316, no. 6031, pp. 820–823, 1985.
[23]  M. M. Gottesman, T. Fojo, and S. E. Bates, “Multidrug resistance in cancer: role of ATP-dependent transporters,” Nature Reviews Cancer, vol. 2, no. 1, pp. 48–58, 2002.
[24]  P. M. Chaudhary and I. B. Roninson, “Expression and activity of P-glycoprotein, a multidrug efflux pump, in human hematopoietic stem cells,” Cell, vol. 66, no. 1, pp. 85–94, 1991.
[25]  W. T. Klimecki, B. W. Futscher, T. M. Grogan, and W. S. Dalton, “P-glycoprotein expression and function in circulating blood cells from normal volunteers,” Blood, vol. 83, no. 9, pp. 2451–2458, 1994.
[26]  A. H. Schinkel, U. Mayer, E. Wagenaar et al., “Normal viability and altered pharmacokinetics in mice lacking MDR1-type (drug-transporting) P-glycoproteins,” Proceedings of the National Academy of Sciences of the United States of America, vol. 94, no. 8, pp. 4028–4033, 1997.
[27]  M. F. Fromm, “Importance of P-glycoprotein at blood-tissue barriers,” Trends in Pharmacological Sciences, vol. 25, no. 8, pp. 423–429, 2004.
[28]  G. Luurtsema, J. Verbeek, M. Lubberink et al., “Carbon-11 labeled tracers for in vivo imaging of P-glycoprotein function: kinetics, advantages and disadvantages,” Current Topics in Medicinal Chemistry, vol. 10, no. 17, pp. 1820–1833, 2010.
[29]  K. S. Lown, R. R. Mayo, A. B. Leichtman et al., “Role of intestinal P-glycoprotein (MDR1) in interpatient variation in the oral bioavailability of cyclosporine,” Clinical Pharmacology and Therapeutics, vol. 62, no. 3, pp. 248–260, 1997.
[30]  A. H. Schinkel, “P-glycoprotein, a gatekeeper in the blood-brain barrier,” Advanced Drug Delivery Reviews, vol. 36, no. 2-3, pp. 179–194, 1999.
[31]  B. Sarkadi, L. Homolya, G. Szakács, and A. Váradi, “Human multidrug resistance ABCB and ABCG transporters: participation in a chemoimmunity defense system,” Physiological Reviews, vol. 86, no. 4, pp. 1179–1236, 2006.
[32]  I. Svoboda-Beusan, R. Kusec, K. Bendelja et al., “The relevance of multidrug resistance-associated P-glycoprotein expression in the treatment response of B-cell chronic lymphocytic leukemia,” Haematologica, vol. 85, no. 12, pp. 1261–1267, 2000.
[33]  M. Ohsawa, Y. Ikura, H. Fukushima et al., “Immunohistochemical expression of multidrug resistance proteins as a predictor of poor response to chemotherapy and prognosis in patients with nodal diffuse large B-cell lymphoma,” Oncology, vol. 68, no. 4–6, pp. 422–431, 2005.
[34]  J. J. Louren?o, R. C. Maia, M. A. M. Scheiner, F. C. Vasconcelos, and M. A. M. Moreira, “Genomic variation at the MDR1 promoter and P-glycoprotein expression and activity in AML patients,” Leukemia Research, vol. 32, no. 6, pp. 976–979, 2008.
[35]  T. M. Grogan, C. M. Spier, S. E. Salmon et al., “P-glycoprotein expression in human plasma cell myeloma: correlation with prior chemotherapy,” Blood, vol. 81, no. 2, pp. 490–495, 1993.
[36]  S. V. Ambudkar, Z. E. Sauna, M. M. Gottesman, and G. Szakacs, “A novel way to spread drug resistance in tumor cells: functional intercellular transfer of P-glycoprotein (ABCB1),” Trends in Pharmacological Sciences, vol. 26, no. 8, pp. 385–387, 2005.
[37]  A. Levchenko, B. M. Mehta, X. Niu et al., “Intercellular transfer of P-glycoprotein mediates acquired multidrug resistance in tumor cells,” Proceedings of the National Academy of Sciences of the United States of America, vol. 102, no. 6, pp. 1933–1938, 2005.
[38]  M. Bebawy, V. Combes, E. Lee et al., “Membrane microparticles mediate transfer of P-glycoprotein to drug sensitive cancer cells,” Leukemia, vol. 23, no. 9, pp. 1643–1649, 2009.
[39]  L. J. Robinson, W. K. Roberts, T. T. Ling, D. Lamming, S. S. Sternberg, and P. D. Roepe, “Human MDR 1 Protein overexpression delays the apoptotic cascade in chinese hamster ovary fibroblasts,” Biochemistry, vol. 36, no. 37, pp. 11169–11178, 1997.
[40]  M. J. Smyth, E. Krasovskis, V. R. Sutton, and R. W. Johnstone, “The drug efflux protein, P-glycoprotein, additionally protects drug-resistant tumor cells from multiple forms of caspase-dependent apoptosis,” Proceedings of the National Academy of Sciences of the United States of America, vol. 95, no. 12, pp. 7024–7029, 1998.
[41]  R. W. Johnstone, E. Cretney, and M. J. Smyth, “P-glycoprotein protects leukemia cells against caspase-dependent, but not caspase-independent, cell death,” Blood, vol. 93, no. 3, pp. 1075–1085, 1999.
[42]  M. Pallis, J. Turzanski, M. Grundy, C. Seedhouse, and N. Russell, “Resistance to spontaneous apoptosis in acute myeloid leukaemia blasts is associated with P-glycoprotein expression and function, but not with the presence of FLT3 internal tandem duplications,” British Journal of Haematology, vol. 120, no. 6, pp. 1009–1016, 2003.
[43]  P. S. Souza, F. C. Vasconcelos, F. R. de Souza Reis, G. N. De Moraes, and R. C. Maia, “P-glycoprotein and survivin simultaneously regulate vincristine-induced apoptosis in chronic myeloid leukemia cells,” International Journal of Oncology, vol. 39, no. 4, pp. 925–933, 2011.
[44]  Y. Kuwazuru, A. Yoshimura, S. Hanada et al., “Expression of the multidrug transporter, P-glycoprotein, in chronic myelogenous leukaemia cells in blast crisis,” British Journal of Haematology, vol. 74, no. 1, pp. 24–29, 1990.
[45]  A. Stavrovskaya, A. Turkina, N. Sedyakhina et al., “Prognostic value of P-glycoprotein and leukocyte differentiation antigens in chronic meloid leukemia,” Leukemia and Lymphoma, vol. 28, no. 5-6, pp. 469–482, 1998.
[46]  A. F. List, K. J. Kopecky, C. L. Willman et al., “Cyclosporine inhibition of P-glycoprotein in chronic myeloid leukemia blast phase,” Blood, vol. 100, no. 5, pp. 1910–1912, 2002.
[47]  E. Weisberg and J. D. Griffin, “Mechanism of resistance to the ABL tyrosine kinase inhibitor STI571 in BCR/ABL-transformed hematopoietic cell lines,” Blood, vol. 95, no. 11, pp. 3498–3505, 2000.
[48]  F. C. Vasconcelos, K. L. Silva, P. S. Souza et al., “Variation of MDR proteins expression and activity levels according to clinical status and evolution of CML patients,” Cytometry, B, vol. 80, no. 3, pp. 158–166, 2011.
[49]  F. X. Mahon, M. W. Deininger, B. Schultheis, et al., “Selection and characterization of BCR-ABL positive cell lines with differential sensitivity to the signal transduction inhibitor STI571: diverse mechanisms of resistance,” Blood, vol. 96, pp. 1070–1079, 2000.
[50]  N. Widmer, H. Rumpold, G. Untergasser, A. Fayet, T. Buclin, and L. A. Decosterd, “Resistance reversal by RNAi silencing of MDR1 in CML cells associated with increase in imatinib intracellular levels,” Leukemia, vol. 21, no. 7, pp. 1561–1562, 2007.
[51]  Y. Assef, F. Rubio, G. Coló, S. del Mónaco, M. A. Costas, and B. A. Kotsias, “Imatinib resistance in multidrug-resistant K562 human leukemic cells,” Leukemia Research, vol. 33, no. 5, pp. 710–716, 2009.
[52]  F. X. Mahon, F. Belloc, V. Lagarde et al., “MDR1 gene overexpression confers resistance to imatinib mesylate in leukemia cell line models,” Blood, vol. 101, no. 6, pp. 2368–2373, 2003.
[53]  H. Rumpold, A. M. Wolf, K. Gruenewald, G. Gastl, E. Gunsilius, and D. Wolf, “RNAi-mediated knockdown of P-glycoprotein using a transposon-based vector system durably restores imatinib sensitivity in imatinib-resistant CML cell lines,” Experimental Hematology, vol. 33, no. 7, pp. 767–775, 2005.
[54]  T. Illmer, M. Schaich, U. Platzbecker et al., “P-glycoprotein-mediated drug efflux is a resistance mechanism of chronic myelogenous leukemia cells to treatment with imatinib mesylate,” Leukemia, vol. 18, no. 3, pp. 401–408, 2004.
[55]  Y. Zong, S. Zhou, and B. P. Sorrentino, “Loss of P-glycoprotein expression in hematopoietic stem cells does not improve responses to imatinib in a murine model of chronic myelogenous leukemia,” Leukemia, vol. 19, no. 9, pp. 1590–1596, 2005.
[56]  T. P. Stromskaya, E. Y. Rybalkina, S. S. Kruglov et al., “Role of P-glycoprotein in evolution of populations of chronic myeloid leukemia cells treated with imatinib,” Biochemistry, vol. 73, no. 1, pp. 29–37, 2008.
[57]  S. Hatziieremia, N. E. Jordanides, T. L. Holyoake, J. C. Mountford, and H. G. J?rgensen, “Inhibition of MDR1 does not sensitize primitive chronic myeloid leukemia CD34+ cells to imatinib,” Experimental Hematology, vol. 37, no. 6, pp. 692–700, 2009.
[58]  H. Gurney, M. Wong, R. L. Balleine et al., “Imatinib disposition and ABCB1 (MDR1, P-glycoprotein) genotype,” Clinical Pharmacology and Therapeutics, vol. 82, no. 1, pp. 33–40, 2007.
[59]  C. Kimchi-Sarfaty, J. M. Oh, I. W. Kim et al., “A "silent" polymorphism in the MDR1 gene changes substrate specificity,” Science, vol. 315, no. 5811, pp. 525–528, 2007.
[60]  S. Dulucq, S. Bouchet, B. Turcq et al., “Multidrug resistance gene (MDR1) polymorphisms are associated with major molecular responses to standard-dose imatinib in chronic myeloid leukemia,” Blood, vol. 112, no. 5, pp. 2024–2027, 2008.
[61]  L. N. Ni, J. Y. Li, K. R. Miao et al., “Multidrug resistance gene (MDR1) polymorphisms correlate with imatinib response in chronic myeloid leukemia,” Medical Oncology, vol. 28, no. 1, pp. 265–269, 2011.
[62]  R. Bhatia, M. Holtz, N. Niu et al., “Persistence of malignant hematopoietic progenitors in chronic myelogenous leukemia patients in complete cytogenetic remission following imatinib mesylate treatment,” Blood, vol. 101, no. 12, pp. 4701–4707, 2003.
[63]  M. Dohse, C. Scharenberg, S. Shukla et al., “Comparison of ATP-binding cassette transporter interactions with the tyrosine kinase inhibitors imatinib, nilotinib, and dasatinib,” Drug Metabolism and Disposition, vol. 38, no. 8, pp. 1371–1380, 2010.
[64]  C. Hegedus, C. ?zvegy-Laczka, A. á. Apáti et al., “Interaction of nilotinib, dasatinib and bosutinib with ABCB1 and ABCG2: implications for altered anti-cancer effects and pharmacological properties,” British Journal of Pharmacology, vol. 158, no. 4, pp. 1153–1164, 2009.
[65]  F. X. Mahon, S. Hayette, V. Lagarde et al., “Evidence that resistance to nilotinib may be due to BCR-ABL, Pgp, or Src kinase overexpression,” Cancer Research, vol. 68, no. 23, pp. 9809–9816, 2008.
[66]  A. Giannoudis, A. Davies, C. M. Lucas, R. J. Harris, M. Pirmohamed, and R. E. Clark, “Effective dasatinib uptake may occur without human organic cation transporter 1 (hOCT1): implications for the treatment of imatinib-resistant chronic myeloid leukemia,” Blood, vol. 112, no. 8, pp. 3348–3354, 2008.
[67]  D. K. Hiwase, V. Saunders, D. Hewett et al., “Dasatinib cellular uptake and efflux in chronic myeloid leukemia cells: therapeutic implications,” Clinical Cancer Research, vol. 14, no. 12, pp. 3881–3888, 2008.
[68]  N. L. Bartlett, B. L. Lum, G. A. Fisher et al., “Phase I trial of doxorubicin with cyclosporine as a modulator of multidrug resistance,” Journal of Clinical Oncology, vol. 12, no. 4, pp. 835–842, 1994.
[69]  A. Garrigues, J. Nugier, S. Orlowski, and E. Ezan, “A high-throughput screening microplate test for the interaction of drugs with P-glycoprotein,” Analytical Biochemistry, vol. 305, no. 1, pp. 106–114, 2002.
[70]  I. Frydrych, P. Mlejnek, and P. Dolezel, “Cyclosporin A sensitises Bcr-Abl positive cells to imatinib mesylate independently of P-glycoprotein expression,” Toxicology in Vitro, vol. 23, no. 8, pp. 1482–1490, 2009.
[71]  R. C. Maia, M. K. Carri?o, C. E.N.P. Klumb et al., “Clinical approach to circumvention of multidrug resistance in refractory leukemic patients: association of cyclosporin A with etoposide,” Journal of Experimental and Clinical Cancer Research, vol. 16, no. 4, pp. 419–424, 1997.
[72]  R. C. Maia, H. Noronha, F. C. Vasconcelos, and V. M. Rumjanek, “Interaction of cyclosporin A and etoposide. Clinical and in vitro assessment in blast phase of chronic myeloid leukaemia,” Clinical and Laboratory Haematology, vol. 19, no. 3, pp. 215–217, 1997.
[73]  Z. Ni, M. E. Mark, X. Cai, and Q. Mao, “Fluorescence resonance energy transfer (FRET) analysis demonstrates dimer/oligomer formation of the human breast cancer resistance protein (BCRP/ABCG2) in intact cells,” International Journal of Biochemistry and Molecular Biology, vol. 1, no. 1, pp. 1–11, 2010.
[74]  P. A. Fetsch, A. Abati, T. Litman et al., “Localization of the ABCG2 mitoxantrone resistance-associated protein in normal tissues,” Cancer Letters, vol. 235, no. 1, pp. 84–92, 2006.
[75]  M. Nakagawa, E. Schneider, K. H. Dixon et al., “Reduced intracellular drug accumulation in the absence of P-glycoprotein (MDR1) overexpression in mitoxantrone-resistant human MCF-7 breast cancer cells,” Cancer Research, vol. 52, no. 22, pp. 6175–6181, 1992.
[76]  C. Lemos, G. Jansen, and G. J. Peters, “Drug transporters: recent advances concerning BCRP and tyrosine kinase inhibitors,” British Journal of Cancer, vol. 98, no. 5, pp. 857–862, 2008.
[77]  Y. Honjo, C. A. Hrycyna, Q. W. Yan et al., “Acquired mutations in the MXR/BCRP/ABCP gene alter substrate specificity in MXR/BCRP/ABCP-overexpressing cells,” Cancer Research, vol. 61, no. 18, pp. 6635–6639, 2001.
[78]  M. Miwa, S. Tsukahara, E. Ishikawa, S. Asada, Y. Imai, and Y. Sugimoto, “Single amino acid substitutions in the transmembrane domains of breast cancer resistance protein (BCRP) alter cross resistance patterns in transfectants,” International Journal of Cancer, vol. 107, no. 5, pp. 757–763, 2003.
[79]  C. ?zvegy-Laczka, T. Hegedus, G. Várady et al., “High-affinity interaction of tyrosine kinase inhibitors with the ABCG2 multidrug transporter,” Molecular Pharmacology, vol. 65, no. 6, pp. 1485–1495, 2004.
[80]  S. Shukla, Z. E. Sauna, and S. V. Ambudkar, “Evidence for the interaction of imatinib at the transport-substrate site(s) of the multidrug-resistance-linked ABC drug transporters ABCB1 (P-glycoprotein) and ABCG2,” Leukemia, vol. 22, no. 2, pp. 445–447, 2008.
[81]  P. J. Houghton, G. S. Germain, F. C. Harwood et al., “Imatinib mesylate is a potent inhibitor of the ABCG2 (BCRP) transporter and reverses resistance to topotecan and SN-38 in vitro,” Cancer Research, vol. 64, no. 7, pp. 2333–2337, 2004.
[82]  H. Burger, H. van Tol, A. W. M. Boersma et al., “Imatinib mesylate (STI571) is a substrate for the breast cancer resistance protein (BCRP)/ABCG2 drug pump,” Blood, vol. 104, no. 9, pp. 2940–2942, 2004.
[83]  C. Brendel, C. Scharenberg, M. Dohse et al., “Imatinib mesylate and nilotinib (AMN107) exhibit high-affinity interaction with ABCG2 on primitive hematopoietic stem cells,” Leukemia, vol. 21, no. 6, pp. 1267–1275, 2007.
[84]  S. M. Graham, H. G. J?rgensen, E. Allan et al., “Primitive, quiescent, Philadelphia-positive stem cells from patients with chronic myeloid leukemia are insensitive to STI571 in vitro,” Blood, vol. 99, no. 1, pp. 319–325, 2002.
[85]  T. Nakanishi, K. Shiozawa, B. A. Hassel, and D. D. Ross, “Complex interaction of BCRP/ABCG2 and imatinib in BCR-ABL-expressing cells: BCRP-mediated resistance to imatinib is attenuated by imatinib-induced reduction of BCRP expression,” Blood, vol. 108, no. 2, pp. 678–684, 2006.
[86]  M. A. Goodell, K. Brose, G. Paradis, A. S. Conner, and R. C. Mulligan, “Isolation and functional properties of murine hematopoietic stem cells that are replicating in vivo,” Journal of Experimental Medicine, vol. 183, no. 4, pp. 1797–1806, 1996.
[87]  S. Zhou, J. D. Schuetz, K. D. Bunting et al., “The ABC transporter BCRP1/ABCG2 is expressed in a wide variety of stem cells and is a molecular determinant of the side-population phenotype,” Nature Medicine, vol. 7, no. 9, pp. 1028–1034, 2001.
[88]  C. W. Scharenberg, M. A. Harkey, and B. Torok-Storb, “The ABCG2 transporter is an efficient hoechst 33342 efflux pump and is preferentially expressed by immature human hematopoietic progenitors,” Blood, vol. 99, no. 2, pp. 507–512, 2002.
[89]  J. W. Jonker, M. Buitelaar, E. Wagenaar et al., “The breast cancer resistance protein protects against a major chlorophyll-derived dietary phototoxin and protoporphyria,” Proceedings of the National Academy of Sciences of the United States of America, vol. 99, no. 24, pp. 15649–15654, 2002.
[90]  S. Zhou, J. J. Morris, Y. Barnes, L. Lan, J. D. Schuetz, and B. P. Sorrentino, “Bcrp1 gene expression is required for normal numbers of side population stem cells in mice, and confers relative protection to mitoxantrone in hematopoietic cells in vivo,” Proceedings of the National Academy of Sciences of the United States of America, vol. 99, no. 19, pp. 12339–12344, 2002.
[91]  P. Breedveld, D. Pluim, G. Cipriani et al., “The effect of Bcrp1 (Abcg2) on the in vivo pharmacokinetics and brain penetration of imatinib mesylate (Gleevec): implications for the use of breast cancer resistance protein and P-glycoprotein inhibitors to enable the brain penetration of imatinib in patients,” Cancer Research, vol. 65, no. 7, pp. 2577–2582, 2005.
[92]  Y. Imai, M. Nakane, K. Kage et al., “C421A polymorphism in the human breast cancer resistance protein gene is associated with low expression of Q141K protein and low-level drug resistance,” Molecular Cancer Therapeutics, vol. 1, no. 8, pp. 611–616, 2002.
[93]  A. Tamura, M. Watanabe, H. Saito et al., “Functional validation of the genetic polymorphisms of human ATP-binding cassette (ABC) transporter ABCG2: identification of alleles that are defective in porphyrin transport,” Molecular Pharmacology, vol. 70, no. 1, pp. 287–296, 2006.
[94]  S. Mizuarai, N. Aozasa, and H. Kotani, “Single nucleotide polymorphisms result in impaired membrane localization and reduced atpase activity in multidrug transporter ABCG2,” International Journal of Cancer, vol. 109, no. 2, pp. 238–246, 2004.
[95]  N. Takahashi and M. Miura, “Therapeutic drug monitoring of imatinib for chronic myeloid leukemia patients in the chronic phase,” Pharmacology, vol. 87, no. 5-6, pp. 241–248, 2011.
[96]  H. Koepsell, K. Lips, and C. Volk, “Polyspecific organic cation transporters: structure, function, physiological roles, and biopharmaceutical implications,” Pharmaceutical Research, vol. 24, no. 7, pp. 1227–1251, 2007.
[97]  M. A. Hediger, M. F. Romero, J. B. Peng, A. Rolfs, H. Takanaga, and E. A. Bruford, “The ABCs of solute carriers: physiological, pathological and therapeutic implications of human membrane transport proteins,” Pflugers Archiv European Journal of Physiology, vol. 447, no. 5, pp. 465–468, 2004.
[98]  L. Wang, A. Giannoudis, S. Lane, P. Williamson, M. Pirmohamed, and R. E. Clark, “Expression of the uptake drug transporter hOCT1 is an important clinical determinant of the response to imatinib in chronic myeloid leukemia,” Clinical Pharmacology and Therapeutics, vol. 83, no. 2, pp. 258–264, 2008.
[99]  J. Thomas, L. Wang, R. E. Clark, and M. Pirmohamed, “Active transport of imatinib into and out of cells: implications for drug resistance,” Blood, vol. 104, no. 12, pp. 3739–3745, 2004.
[100]  D. L. White, V. A. Saunders, P. Dang et al., “Most CML patients who have a suboptimal response to imatinib have low OCT-1 activity: higher doses of imatinib may overcome the negative impact of low OCT-1 activity,” Blood, vol. 110, no. 12, pp. 4064–4072, 2007.
[101]  L. C. Crossman, B. J. Druker, M. W. N. Deininger, M. Pirmohamed, L. Wang, and R. E. Clark, “hOCT 1 and resistance to imatinib,” Blood, vol. 106, no. 3, pp. 1133–1134, 2005.
[102]  S. Hu, R. M. Franke, K. K. Filipski et al., “Interaction of imatinib with human organic ion carriers,” Clinical Cancer Research, vol. 14, no. 10, pp. 3141–3148, 2008.
[103]  N. E. Crook, R. J. Clem, and L. K. Miller, “An apoptosis-inhibiting baculovirus gene with a zinc finger-like motif,” Journal of Virology, vol. 67, no. 4, pp. 2168–2174, 1993.
[104]  Q. L. Deveraux and J. C. Reed, “IAP family proteins—suppressors of apoptosis,” Genes and Development, vol. 13, no. 3, pp. 239–252, 1999.
[105]  A. Wrzesień-Ku?, P. Smolewski, A. Sobczak-Pluta, A. Wierzbowska, and T. Robak, “The inhibitor of apoptosis protein family and its antagonists in acute leukemias,” Apoptosis, vol. 9, no. 6, pp. 705–715, 2004.
[106]  Q. L. Deveraux, R. Takahashi, G. S. Salvesen, and J. C. Reed, “X-linked IAP is a direct inhibitor of cell-death proteases,” Nature, vol. 388, no. 6639, pp. 300–304, 1997.
[107]  B. P. Eckelman, G. S. Salvesen, and F. L. Scott, “Human inhibitor of apoptosis proteins: why XIAP is the black sheep of the family,” EMBO Reports, vol. 7, no. 10, pp. 988–994, 2006.
[108]  F. L. Scott, J. B. Denault, S. J. Riedl, H. Shin, M. Renatus, and G. S. Salvesen, “XIAP inhibits caspase-3 and -7 using two binding sites: evolutionary conserved mechanism of IAPs,” EMBO Journal, vol. 24, no. 3, pp. 645–655, 2005.
[109]  A. M. Verhagen, P. G. Ekert, M. Pakusch et al., “Identification of DIABLO, a mammalian protein that promotes apoptosis by binding to and antagonizing IAP proteins,” Cell, vol. 102, no. 1, pp. 43–53, 2000.
[110]  P. Liston, W. G. Fong, N. L. Kelly et al., “Identification of XAF1 as an antagonist of XIAP anti-Caspase activity,” Nature Cell Biology, vol. 3, no. 2, pp. 128–133, 2001.
[111]  M. Holcik and R. G. Korneluk, “XIAP, the guardian angel,” Nature Reviews Molecular Cell Biology, vol. 2, no. 7, pp. 550–556, 2001.
[112]  A. R. Mufti, E. Burstein, and C. S. Duckett, “XIAP: cell death regulation meets copper homeostasis,” Archives of Biochemistry and Biophysics, vol. 463, no. 2, pp. 168–174, 2007.
[113]  S. Galbán and C. S. Duckett, “XIAP as a ubiquitin ligase in cellular signaling,” Cell Death and Differentiation, vol. 17, no. 1, pp. 54–60, 2010.
[114]  H. Harlin, S. B. Reffey, C. S. Duckett, T. Lindsten, and C. B. Thompson, “Characterization of XIAP-deficient mice,” Molecular and Cellular Biology, vol. 21, no. 10, pp. 3604–3608, 2001.
[115]  B. Vischioni, P. van der Valk, S. W. Span, F. A. E. Kruyt, J. A. Rodriguez, and G. Giaccone, “Expression and localization of inhibitor of apoptosis proteins in normal human tissues,” Human Pathology, vol. 37, no. 1, pp. 78–86, 2006.
[116]  S. S. Liu, B. K. Tsang, A. N. Cheung, et al., “Anti-apoptotic proteins, apoptotic and proliferative parameters and their prognostic significance in cervical carcinoma,” European Journal of Cancer, vol. 37, no. 9, pp. 1104–1110, 2001.
[117]  B. Z. Carter, S. M. Kornblau, T. Tsao et al., “Caspase-independent cell death in AML: caspase inhibition in vitro with pan-caspase inhibitors or in vivo by XIAP or Survivin does not affect cell survival or prognosis,” Blood, vol. 102, no. 12, pp. 4179–4186, 2003.
[118]  I. Tamm, S. Richter, F. Scholz et al., “Xiap expression correlates with monocytic differentiation in adult de novo AML: impact on prognosis,” Hematology Journal, vol. 5, no. 6, pp. 489–495, 2004.
[119]  I. Tamm, S. Richter, D. Oltersdorf et al., “High expression levels of x-linked inhibitor of apoptosis protein and survivin correlate with poor overall survival in childhood de novo acute myeloid leukemia,” Clinical Cancer Research, vol. 10, no. 11, pp. 3737–3744, 2004.
[120]  Y. Zhang, J. Zhu, Y. Tang et al., “X-linked inhibitor of apoptosis positive nuclear labeling: a new independent prognostic biomarker of breast invasive ductal carcinoma,” Diagnostic Pathology, vol. 6, no. 1, article 49, 2011.
[121]  G. Fang, C. N. Kim, C. L. Perkins et al., “CGP57148B (STI-571) induces differentiation and apoptosis and sensitizes Bcr-Abl-positive human leukemia cells to apoptosis due to antileukemic drugs,” Blood, vol. 96, no. 6, pp. 2246–2253, 2000.
[122]  J. S. Kim, H. K. Jeung, J. W. Cheong et al., “Apicidin potentiates the imatinib-induced apoptosis of Bcr-Abl-positive human leukaemia cells by enhancing the activation of mitochondria-dependent caspase cascades,” British Journal of Haematology, vol. 124, no. 2, pp. 166–178, 2004.
[123]  K. Airiau, F.-X. Mahon, M. Josselin, M. Jeanneteau, B. Turcq, and F. Belloc, “ABT-737 increases tyrosine kinase inhibitor-induced apoptosis in chronic myeloid leukemia cells through XIAP downregulation and sensitizes CD34+ CD38- population to imatinib,” Experimental Hematology. In press.
[124]  X. S. Hao, J. H. Hao, F. T. Liu, A. C. Newland, and L. Jia, “Potential mechanisms of leukemia cell resistance to TRAIL-induced apopotosis,” Apoptosis, vol. 8, no. 6, pp. 601–607, 2003.
[125]  D. C. McManus, C. A. Lefebvre, G. Cherton-Horvat et al., “Loss of XIAP protein expression by RNAi and antisense approaches sensitizes cancer cells to functionally diverse chemotherapeutics,” Oncogene, vol. 23, no. 49, pp. 8105–8117, 2004.
[126]  A. D. Schimmer, S. Dalili, R. A. Batey, and S. J. Riedl, “Targeting XIAP for the treatment of malignancy,” Cell Death and Differentiation, vol. 13, no. 2, pp. 179–188, 2006.
[127]  A. D. Schimmer, E. H. Estey, G. Borthakur et al., “Phase I/II trial of AEG35156 X-linked inhibitor of apoptosis protein antisense oligonucleotide combined with idarubicin and cytarabine in patients with relapsed or primary refractory acute myeloid leukemia,” Journal of Clinical Oncology, vol. 27, no. 28, pp. 4741–4746, 2009.
[128]  R. T. Lima, L. M. Martins, J. E. Guimar?es, C. Sambade, and M. H. Vasconcelos, “Chemosensitization effects of XIAP downregulation in K562 leukemia cells,” Journal of Chemotherapy, vol. 18, no. 1, pp. 98–102, 2006.
[129]  H. Seca, R. T. Lima, J. E. Guimar?es, and M. H. Vasconcelos, “Simultaneous targeting of P-gp and XIAP with siRNAs increases sensitivity of P-gp overexpressing CML cells to imatinib,” Hematology, vol. 16, no. 2, pp. 100–108, 2011.
[130]  P. S. de Souza, F. da Cunha Vasconcelos, L. F. R. Silva, and R. C. Maia, “Cyclosporine A enables vincristine-induced apoptosis during reversal of multidrug resistance phenotype in chronic myeloid leukemia cells,” Tumor Biology. In press.
[131]  G. Ambrosini, C. Adida, and D. C. Altieri, “A novel anti-apoptosis gene, survivin, expressed in cancer and lymphoma,” Nature Medicine, vol. 3, no. 8, pp. 917–921, 1997.
[132]  F. Li, G. Ambrosini, E. Y. Chu et al., “Control of apoptosis and mitotic spindle checkpoint by survivin,” Nature, vol. 396, no. 6711, pp. 580–584, 1998.
[133]  F. Li and X. Ling, “Survivin study: an update of “what is the next wave?”,” Journal of Cellular Physiology, vol. 208, no. 3, pp. 476–486, 2006.
[134]  M. Speletas, N. Argentou, V. Karanikas et al., “Survivin isoform expression patterns in CML patients correlate with resistance to imatinib and progression, but do not trigger cytolytic responses,” Clinical Immunology, vol. 139, no. 2, pp. 155–163, 2011.
[135]  D. C. Altieri, “Validating survivin as a cancer therapeutic target,” Nature Reviews Cancer, vol. 3, no. 1, pp. 46–54, 2003.
[136]  S. Fukuda and L. M. Pelus, “Survivin, a cancer target with an emerging role in normal adult tissues,” Molecular Cancer Therapeutics, vol. 5, no. 5, pp. 1087–1098, 2006.
[137]  S. Fukuda and L. M. Pelus, “Regulation of the inhibitor-of-apoptosis family member survivin in normal cord blood and bone marrow CD34+ cells by hematopoietic growth factors: Implication of survivin expression in normal hematopoiesis,” Blood, vol. 98, no. 7, pp. 2091–2100, 2001.
[138]  J. M. Gerber, L. Qin, J. Kowalski et al., “Characterization of chronic myeloid leukemia stem cells,” American Journal of Hematology, vol. 86, no. 1, pp. 31–37, 2011.
[139]  G. V. Helgason, G. A. R. Young, and T. L. Holyoake, “Targeting chronic myeloid leukemia stem cells,” Current Hematologic Malignancy Reports, vol. 5, no. 2, pp. 81–87, 2010.
[140]  A. Mori, H. Wada, Y. Nishimura, T. Okamoto, Y. Takemoto, and E. Kakishita, “Expression of the antiapoptosis gene survivin in human leukemia,” International Journal of Hematology, vol. 75, no. 2, pp. 161–165, 2002.
[141]  A. Badran, A. Yoshida, Yujiwano et al., “Expression of the antiapoptotic gene survivin in chronic myeloid leukemia,” Anticancer Research, vol. 23, no. 1, pp. 589–592, 2003.
[142]  E. Conte, F. Stagno, P. Guglielmo, A. Scuto, C. Consoli, and A. Messina, “Survivin expression in chronic myeloid leukemia,” Cancer Letters, vol. 225, no. 1, pp. 105–110, 2005.
[143]  M. Zhara, H. Mourad, G. Farouk, M. Elbatch, S. Ezzat, and W. Sami, “Molecular detection of survivin expression, antiapoptotic gene, and other prognostic markers, how they are correlated and how it could be of prognostic value in chronic myeloid leukemia patient,” The Egyptian Journal of Immunology, vol. 14, no. 2, pp. 51–62, 2007.
[144]  B. Z. Carter, D. H. Mak, W. D. Schober et al., “Regulation of survivin expression through BCR-Abl/MAPK cascade: targeting survivin overcomes imatinib resistance and increases imatinib sensitivity in imatinib-responsive CML cells,” Blood, vol. 107, no. 4, pp. 1555–1563, 2006.
[145]  F. R. D. S. Reis, F. D. C. Vasconcelos, D. L. Pereira, A. Moellman-Coelho, K. L. Silva, and R. C. Maia, “Survivin and P-glycoprotein are associated and highly expressed in late phase chronic myeloid leukemia,” Oncology Reports, vol. 26, no. 2, pp. 471–478, 2011.
[146]  H. Yamamoto, C. Y. Ngan, and M. Monden, “Cancer cells survive with survivin,” Cancer Science, vol. 99, no. 9, pp. 1709–1714, 2008.
[147]  J. K. Kwee, D. G. Luque, A. C. D. S. Ferreira et al., “Modulation of reactive oxygen species by antioxidants in chronic myeloid leukemia cells enhances imatinib sensitivity through survivin downregulation,” Anti-Cancer Drugs, vol. 19, no. 10, pp. 975–981, 2008.
[148]  G. Nestal de Moraes, K. L. Silva, F. D. C. Vasconcelos, and R. C. Maia, “Survivin overexpression correlates with an apoptosis-resistant phenotype in chronic myeloid leukemia cells,” Oncology Reports, vol. 25, no. 6, pp. 1613–1619, 2011.
[149]  W. Fiskus, Y. Wang, R. Joshi et al., “Cotreatment with vorinostat enhances activity of MK-0457 (VX-680) against acute and chronic myelogenous leukemia cells,” Clinical Cancer Research, vol. 14, no. 19, pp. 6106–6115, 2008.
[150]  S. A. Bright, G. Campiani, M. W. Deininger, M. Lawler, D. C. Williams, and D. M. Zisterer, “Sequential treatment with flavopiridol synergistically enhances pyrrolo-1,5-benzoxazepine-induced apoptosis in human chronic myeloid leukaemia cells including those resistant to imatinib treatment,” Biochemical Pharmacology, vol. 80, no. 1, pp. 31–38, 2010.
[151]  T. Nakahara, A. Kita, K. Yamanaka et al., “Broad spectrum and potent antitumor activities of YM155, a novel small-molecule survivin suppressant, in a wide variety of human cancer cell lines and xenograft models,” Cancer Science, vol. 102, no. 3, pp. 614–621, 2011.
[152]  P. Fortugno, N. R. Wall, A. Giodini et al., “Survivin exists in immunochemically distinct subcellular pools and is involved in spindle microtubule function,” Journal of Cell Science, vol. 115, no. 3, pp. 575–585, 2002.
[153]  B. Gyurkocza, J. Plescia, C. M. Raskett et al., “Antileukemic activity of shepherdin and molecular diversity of Hsp90 inhibitors,” Journal of the National Cancer Institute, vol. 98, no. 15, pp. 1068–1077, 2006.
[154]  Z. Wang, J. Sampath, S. Fukuda, and L. M. Pelus, “Disruption of the inhibitor of apoptosis protein survivin sensitizes BCR-ABL-positive cells to STI571-induced apoptosis,” Cancer Research, vol. 65, no. 18, pp. 8224–8232, 2005.
[155]  S. F. de Mattos, A. Essafi, I. Soeiro et al., “FOXO3a and BCR-ABL regulate cyclin D2 transcription through a STAT5/BCL6-dependent mechanism,” Molecular and Cellular Biology, vol. 24, no. 22, pp. 10058–10071, 2004.
[156]  A. Essafi, S. Fernández De Mattos, Y. A. M. Hassen et al., “Direct transcriptional regulation of Bim by FOXO3a mediates STI571-induced apoptosis in Bcr-Abl-expressing cells,” Oncogene, vol. 24, no. 14, pp. 2317–2329, 2005.
[157]  N. Komatsu, T. Watanabe, M. Uchida et al., “A member of forkhead transcription factor FKHRL1 is a downstream effector of STI571-induced cell cycle arrest in BCR-ABL-expressing cells,” Journal of Biological Chemistry, vol. 278, no. 8, pp. 6411–6419, 2003.
[158]  S. Kikuchi, T. Nagai, M. Kunitama, K. Kirito, K. Ozawa, and N. Komatsu, “Active FKHRL1 overcomes imatinib resistance in chronic myelogenous leukemia-derived cell lines via the production of tumor necrosis factor-related apoptosis-inducing ligand,” Cancer Science, vol. 98, no. 12, pp. 1949–1958, 2007.
[159]  K. U. Birkenkamp, A. Essafi, K. E. van der Vos et al., “FOXO3a induces differentiation of Bcr-Abl-transformed cells through transcriptional down-regulation of Id1,” Journal of Biological Chemistry, vol. 282, no. 4, pp. 2211–2220, 2007.
[160]  Z. Jagani, K. Song, J. L. Kutok et al., “Proteasome inhibition causes regression of leukemia and abrogates BCR-ABL-induced evasion of apoptosis in part through regulation of forkhead tumor suppressors,” Cancer Research, vol. 69, no. 16, pp. 6546–6555, 2009.
[161]  D. Cilloni, F. Messa, F. Arruga et al., “The NF-κB pathway blockade by the IKK inhibitor PS1145 can overcome Imatinib resistance,” Leukemia, vol. 20, no. 1, pp. 61–67, 2006.
[162]  E. A. Duncan, C. A. Goetz, S. J. Stein et al., “IκB kinase β inhibition induces cell death in Imatinib-resistant and T315I Dasatinib-resistant BCR-ABL+ cells,” Molecular Cancer Therapeutics, vol. 7, no. 2, pp. 391–397, 2008.
[163]  N. Lounnas, C. Frelin, N. Gonthier et al., “NF-κB inhibition triggers death of imatinib-sensitive and imatinib-resistant chronic myeloid leukemia cells including T315I Bcr-Abl mutants,” International Journal of Cancer, vol. 125, no. 2, pp. 308–317, 2009.
[164]  M. P. Albero, J. M. Vaquer, E. J. Andreu et al., “Bortezomib decreases Rb phosphorylation and induces caspase-dependent apoptosis in Imatinib-sensitive and-resistant Bcr-Abl1-expressing cells,” Oncogene, vol. 29, no. 22, pp. 3276–3286, 2010.
[165]  Z. Hu, X. F. Pan, F. Q. Wu et al., “Synergy between proteasome inhibitors and imatinib mesylate in chronic myeloid leukemia,” Plos ONE, vol. 4, no. 7, Article ID e6257, 2009.
[166]  R. C. Maia, F. C. Vasconcelos, T. de Sá Bacelar et al., “LQB-118, a pterocarpanquinone structurally related to lapachol [2-hydroxy-3-(3-methyl-2-butenyl)-1,4-naphthoquinone]: a novel class of agent with high apoptotic effect in chronic myeloid leukemia cells,” Investigational New Drugs, vol. 29, no. 6, pp. 1143–1155, 2011.
[167]  M. Gromicho, J. Dinis, M. Magalh?es et al., “Development of imatinib and dasatinib resistance: dynamics of expression of drug transporters ABCB1, ABCC1, ABCG2, MVP, and SLC22A1,” Leukemia and Lymphoma, vol. 52, no. 10, pp. 1980–1990, 2011.
[168]  C. Hirayama, H. Watanabe, R. Nakashima et al., “Constitutive overexpression of P-glycoprotein, rather than breast cancer resistance protein or organic cation transporter 1, contributes to acquisition of imatinib-resistance in K562 cells,” Pharmaceutical Research, vol. 25, no. 4, pp. 827–835, 2008.
[169]  O. Fuchs, “Transcription factor NF-κB inhibitors as single therapeutic agents or in combination with classical chemotherapeutic agents for the treatment of hematologic malignancies,” Current Molecular Pharmacology, vol. 3, no. 3, pp. 98–122, 2010.
[170]  K. C. Arden, “Multiple roles of FOXO transcription factors in mammalian cells point to multiple roles in cancer,” Experimental Gerontology, vol. 41, no. 8, pp. 709–717, 2006.
[171]  R. Sen and D. Baltimore, “Multiple nuclear factors interact with the immunoglobulin enhancer sequences,” Cell, vol. 46, no. 5, pp. 705–716, 1986.
[172]  T. Okamoto, T. Sanda, and K. Asamitsu, “Nk-κb signaling and carcinogenesis,” Current Pharmaceutical Design, vol. 13, no. 5, pp. 447–462, 2007.
[173]  A. K. Mankan, M. W. Lawless, S. G. Gray, D. Kelleher, and R. McManus, “NF-κB regulation: the nuclear response,” Journal of Cellular and Molecular Medicine, vol. 13, no. 4, pp. 631–643, 2009.
[174]  T. Braun, G. Carvalho, C. Fabre, J. Grosjean, P. Fenaux, and G. Kroemer, “Targeting NF-κB in hematologic malignancies,” Cell Death and Differentiation, vol. 13, no. 5, pp. 748–758, 2006.
[175]  T. Lu and G. R. Stark, “Cytokine overexpression and constitutive NFκb in cancer,” Cell Cycle, vol. 3, no. 9, pp. 1114–1117, 2004.
[176]  M. C. Turco, M. F. Romano, A. Petrella, R. Bisogni, P. Tassone, and S. Venuta, “NF-κB/Rel-mediated regulation of apoptosis in hematologic malignancies and normal hematopoietic progenitors,” Leukemia, vol. 18, no. 1, pp. 11–17, 2004.
[177]  D. Cilloni, G. Martinelli, F. Messa, M. Baccarani, and G. Saglio, “Nuclear factor κB as a target for new drug development in myeloid malignancies,” Haematologica, vol. 92, no. 9, pp. 1224–1229, 2007.
[178]  Z. Lu, Y. Jin, C. Chen, J. Li, Q. Cao, and J. Pan, “Pristimerin induces apoptosis in imatinib-resistant chronic myelogenous leukemia cells harboring T315I mutation by blocking NF-κB signaling and depleting Bcr-Abl,” Molecular Cancer, vol. 9, article 112, 2010.
[179]  S. Luqman and J. M. Pezzuto, “NFκB: a promising target for natural products in cancer chemoprevention,” Phytotherapy Research, vol. 24, no. 7, pp. 949–963, 2010.
[180]  L. J. Crawford, P. Windrum, L. Magill et al., “Proteasome proteolytic profile is linked to Bcr-Abl expression,” Experimental Hematology, vol. 37, no. 3, pp. 357–366, 2009.
[181]  K. H. Kaestner, W. Kn?chel, and D. E. Martínez, “Unified nomenclature for the winged helix/forkhead transcription factors,” Genes and Development, vol. 14, no. 2, pp. 142–146, 2000.
[182]  E. L. Greer and A. Brunet, “FOXO transcription factors at the interface between longevity and tumor suppression,” Oncogene, vol. 24, no. 50, pp. 7410–7425, 2005.
[183]  H. Tran, A. Brunet, E. C. Griffith, and M. E. Greenberg, “The many forks in FOXO's road,” Science's STKE, vol. 2003, no. 172, p. RE5, 2003.
[184]  J. Y. Yang and M. C. Hung, “A new fork for clinical application: targeting forkhead transcription factors in cancer,” Clinical Cancer Research, vol. 15, no. 3, pp. 752–757, 2009.
[185]  L. P. van der Heide, F. M. Jacobs, J. P. Burbach, M. F. M. Hoekman, and M. P. Smidt, “FOXO6 transcriptional activity is regulated by Thr26 and Ser184, independent of nucleo-cytoplasmic shuttling,” Biochemical Journal, vol. 391, no. 3, pp. 623–629, 2005.
[186]  L. P. van der Heide, M. F. M. Hoekman, and M. P. Smidt, “The ins and outs of FOXO shuttling: mechanisms of FOXO translocation and transcriptional regulation,” Biochemical Journal, vol. 380, no. 2, pp. 297–309, 2004.
[187]  H. Huang and D. J. Tindall, “Regulation of FOXO protein stability via ubiquitination and proteasome degradation,” Biochimica et Biophysica Acta, vol. 1813, no. 11, pp. 1961–1964, 2011.
[188]  Y. Zhao, Y. Wang, and W.-G. Zhu, “Applications of post-translational modifications of FOXO family proteins in biological functions,” Journal of Molecular Cell Biology, vol. 3, no. 5, pp. 276–282, 2011.
[189]  T. Skorski, A. Bellacosa, M. Nieborowska-Skorska et al., “Transformation of hematopoietic cells by BCR/ABL requires activation of a PI-3k/Akt-dependent pathway,” EMBO Journal, vol. 16, no. 20, pp. 6151–6161, 1997.
[190]  J. Y. Yang and M. C. Hung, “Deciphering the role of forkhead transcription factors in cancer therapy,” Current Drug Targets, vol. 12, no. 9, pp. 1284–1290, 2011.
[191]  Z. Jagani, A. Singh, and R. Khosravi-Far, “FOXO tumor suppressors and BCR-ABL-induced leukemia: a matter of evasion of apoptosis,” Biochimica et Biophysica Acta, vol. 1785, no. 1, pp. 63–84, 2008.
[192]  K. Naka, T. Hoshii, T. Muraguchi et al., “TGF-β-FOXO signalling maintains leukaemia-initiating cells in chronic myeloid leukaemia,” Nature, vol. 463, no. 7281, pp. 676–680, 2010.
[193]  C. Hurtz, K. Hatzi, L. Cerchietti et al., “BCL6-mediated repression of p53 is critical for leukemia stem cell survival in chronic myeloid leukemia,” Journal of Experimental Medicine, vol. 208, no. 11, pp. 2163–2174, 2011.
[194]  K. Naka, T. Hoshii, and A. Hirao, “Novel therapeutic approach to eradicate tyrosine kinase inhibitor resistant chronic myeloid leukemia stem cells,” Cancer Science, vol. 101, no. 7, pp. 1577–1581, 2010.
[195]  T. Nambu, A. Hamada, R. Nakashima et al., “Association of SLCO1B3 polymorphism with intracellular accumulation of imatinib in leukocytes in patients with chronic myeloid leukemia,” Biological and Pharmaceutical Bulletin, vol. 34, no. 1, pp. 114–119, 2011.
[196]  F. Liu, S. Liu, S. He, Z. Xie, X. Zu, and Y. Jiang, “Survivin transcription is associated with P-glycoprotein/MDR1 overexpression in the multidrug resistance of MCF-7 breast cancer cells,” Oncology Reports, vol. 23, no. 5, pp. 1469–1475, 2010.
[197]  C. D. Netto, A. J. M. da Silva, E. J. S. Salustiano et al., “New Pterocarpanquinones: synthesis, antineoplasic activity on cultured human malignant cell lines and TNF-α modulation in human PBMC cells,” Bioorganic and Medicinal Chemistry, vol. 18, no. 4, pp. 1610–1616, 2010.
[198]  M. Bentires-Alj, V. Barbu, M. Fillet et al., “NF-κB transcription factor induces drug resistance through MDR1 expression in cancer cells,” Oncogene, vol. 22, no. 1, pp. 90–97, 2003.
[199]  H. Shen, W. Xu, Q. Chen, Z. Wu, H. Tang, and F. Wang, “Tetrandrine prevents acquired drug resistance of K562 cells through inhibition of MDR1 gene transcription,” Journal of Cancer Research and Clinical Oncology, vol. 136, no. 5, pp. 659–665, 2010.
[200]  R. C. Y. Hui, A. R. Gomes, D. Constantinidou et al., “The forkhead transcription factor FOXO3a increases phosphoinositide-3 kinase/Akt activity in drug-resistant leukemic cells through induction of PIK3CA expression,” Molecular and Cellular Biology, vol. 28, no. 19, pp. 5886–5898, 2008.
[201]  R. C. Y. Hui, R. E. Francis, S. K. Guest et al., “Doxorubicin activates FOXO3a to induce the expression of multidrug resistance gene ABCB1 (MDR1) in K562 leukemic cells,” Molecular Cancer Therapeutics, vol. 7, no. 3, pp. 670–678, 2008.
[202]  C. Hilmi, L. Larribere, M. Deckert et al., “Involvement of FKHRL1 in melanoma cell survival and death,” Pigment Cell and Melanoma Research, vol. 21, no. 2, pp. 139–146, 2008.
[203]  P. Obexer, J. Hagenbuchner, T. Unterkircher et al., “Repression of BIRC5/survivin by FOXO3/FKHRL1 sensitizes human neuroblastoma cells to DNA damage-induced apoptosis,” Molecular Biology of the Cell, vol. 20, no. 7, pp. 2041–2048, 2009.
[204]  M. Guha, J. Plescia, I. Leav, J. Li, L. R. Languino, and D. C. Altieri, “Endogenous tumor suppression mediated by PTEN involves survivin gene silencing,” Cancer Research, vol. 69, no. 12, pp. 4954–4958, 2009.
[205]  L. Tracey, A. Pérez-Rosado, M. J. Artiga et al., “Expression of the NF-κB targets BCL2 and BIRC5/Survivin characterizes small B-cell and aggressive B-cell lymphomas, respectively,” Journal of Pathology, vol. 206, no. 2, pp. 123–134, 2005.
[206]  X. Shi, Y. Jin, C. Cheng et al., “Triptolide inhibits BCR-ABL transcription and induces apoptosis in STI571-resistant chronic myelogenous leukemia cells harboring T315I mutation,” Clinical Cancer Research, vol. 15, no. 5, pp. 1686–1697, 2009.
[207]  Y. L. Wei, Y. Liang, L. Xu, and X. Y. Zhao, “The antiproliferation effect of berbamine on K562 resistant cells by inhibiting NF-κB pathway,” Anatomical Record, vol. 292, no. 7, pp. 945–950, 2009.
[208]  M. Gyrd-Hansen and P. Meier, “IAPs: from caspase inhibitors to modulators of NF-κB, inflammation and cancer,” Nature Reviews Cancer, vol. 10, no. 8, pp. 561–574, 2010.

Full-Text

comments powered by Disqus

Contact Us

service@oalib.com

QQ:3279437679

WhatsApp +8615387084133

WeChat 1538708413