Elsevier

Leukemia Research

Volume 36, Issue 10, October 2012, Pages 1311-1314
Leukemia Research

Nilotinib and imatinib inhibit cytarabine cellular uptake: Implications for combination therapy

https://doi.org/10.1016/j.leukres.2012.06.012Get rights and content

Abstract

The tyrosine kinase inhibitor (TKI) imatinib has been used for a decade to treat chronic myeloid leukemia (CML). A very efficient response is obtained with patients in chronic phase, but its efficacy in late phase patients is often transient. The combination of imatinib or of the new TKI nilotinib with cytarabine is a new treatment approach proposed for CML. We have investigated the effect of imatinib and nilotinib on cytarabine uptake, and have found that both molecules inhibit cytarabine transport. These results should impact on the design of clinical trials that investigate the combination of TKIs and nucleoside analogs.

Introduction

Chronic myeloid leukemia (CML) is characterized by a reciprocal translocation between chromosomes 9 and 22. This genetic modification leads to the generation of the bcr-abl fusion gene that encodes a chimeric protein with constitutive tyrosine kinase activity [1]. Imatinib (Gleevec) is a very efficient inhibitor of the Bcr-Abl fusion protein that has been used for a decade to treat CML patients and acute lymphoblastic leukemia (ALL) patients that harbor a very similar chromosome translocation. An efficient and sustained response to imatinib is obtained in patients with CML in chronic phase but its efficacy is generally transient in late stages CML (accelerated and blast crisis phases) [2], [3], [4]. Moreover, imatinib is not an optimal treatment for 30–35% of the patients in chronic phase; some patients have to discontinue treatment due to side effects, and others lose or do not achieve a complete cytogenetic response, which significantly increases the risk of disease progression [2]. Thus, new therapeutic approaches that increase the cytogenetic response would be beneficial to CML patients.

Nilotinib (Tasigna) is a rationally designed Bcr-Abl inhibitor with greater potency and specificity than imatinib [1]. Furthermore, it is active against several Bcr-Abl mutants that are resistant to imatinib [1]. Nilotinib efficacy was first demonstrated in patients who failed imatinib therapy but its superiority over imatinib has been also confirmed in newly diagnosed CML patients [1], [5], [6], [7].

Treatments that combine imatinib with other chemotherapeutic agents are also actively investigated in CML and ALL patients. The combination of imatinib with the nucleoside analog cytarabine was an obvious regimen to test in the clinic since cytarabine combined to interferon alpha was the previous standard of care for CML before the imatinib era [8], and that several in vitro studies reported synergistic activities with imatinib and cytarabine [9], [10], [11]. A similar combination treatment could be envisaged for nilotinib that is also able to synergize with cytarabine [12].

Nucleosides use specific transporters to enter into cells, with the equilibrative nucleoside transporter 1 (ENT1) being the main cytarabine transporter; its low abundance in acute myeloid leukemia blasts correlates with a lack of response to cytarabine treatment [13]. Few years ago it was reported that some p38 MAPK inhibitors could block nucleoside transport, and that this inhibition occurs in a p38 MAPK-independent manner [14]. This finding was later extended to other types of kinase inhibitors that have different protein targets. Imatinib was tested and it was among the most efficient to block nucleoside transport; it inhibited uridine and thymidine transport by almost 70% at a 10 μM concentration [15]. In another study, it was also reported that imatinib would inhibit the uptake of the nucleosidic analog fludarabine with a similar order of magnitude [16].

We then hypothesized that a combination treatment with cytarabine and imatinib or nilotinib would not be optimal for the treatment of CML if both drugs were used simultaneously. Thus, we investigated to which extent imatinib and nilotinib would inhibit the cellular uptake of the natural nucleoside thymidine, and the nucleoside analog cytarabine in CML cells.

Section snippets

Cell lines and cell culture

K562 (CCL-243) and MEG-01 (CRL-2021) cell lines were obtained from ATCC. Both cell lines were cultured in RPMI-1640 (Wisent, St-Bruno, Canada) supplemented with 10% fetal calf serum, 100 U/ml penicillin and 100 μg/ml streptomycin at 37 °C in a humidified atmosphere with 5% CO2.

Uptake assays of [3H]-thymidine and [3H]-cytarabine

K562 or MEG-01 cells (4 × 105 cells per sample) were incubated at 37 °C for 15 min with 5 μl imatinib or nilotinib at different concentrations, in 0.5 ml of transport buffer (20 mM Tris/HCl, 3 mM K2HPO4, 1 mM MgCl2·6H2O, 2 mM CaCl2, 5 

Imatinib inhibits thymidine and cytarabine uptake in CML cells

The effect of imatinib on nucleoside transport was first examined with thymidine. At a 10-μM concentration, imatinib was able to inhibit the entry of radiolabeled thymidine in K562 cells by 76%. Imatinib was still efficient and blocked by 43% the entry of thymidine at 1 μM, but its effect on thymidine transport at 0.1 μM was not significant (Fig. 1a). With the MEG-01 CML cell line, thymidine entry was inhibited by 72 and 52% with 10 and 1 μM imatinib, respectively. As with K562 cells, the effect

Discussion

At a common regimen of 400 mg daily, imatinib reaches a mean peak concentration of 4.6 μM and a mean serum trough concentration of 1.5 μM [1]. We report that 1 μM imatinib inhibits cytarabine by 23% in K562 cells, and by 25% in MEG-01 cells (Fig. 1c and d). These results are in line with those reported previously with uridine in K562 cells and fludarabine with human T-lymphocytes [15], [16]. Results from a French clinical trial and from the HOVON-51 study that investigate the upfront combination

Conflict of interest statement

The authors declare no conflict of interest in connection with this work.

Acknowledgements

Contributions. JBS, KG and MC performed experiments, and POdeL and MC designed the study and wrote the manuscript.

References (19)

There are more references available in the full text version of this article.

Cited by (6)

  • Development and validation of a sensitive LC–MS/MS method for simultaneous determination of eight tyrosine kinase inhibitors and its application in mice pharmacokinetic studies

    2018, Journal of Pharmaceutical and Biomedical Analysis
    Citation Excerpt :

    Hence, more effective therapies via combinational approach are warranted in order to improve the therapeutic outcomes. In fact, several preclinical and clinical trials of combining different TKIs are actively ongoing [11,14–16]. Moreover, TKIs show high interpatient pharmacokinetics variability, possibly due to food intake, co-administered drug, disease or other factors.

  • Selective inhibition of human equilibrative and concentrative nucleoside transporters by BCR-ABL kinase inhibitors: Identification of key hENT1 amino acid residues for interaction with BCR-ABL kinase inhibitors

    2016, Journal of Biological Chemistry
    Citation Excerpt :

    Accumulation of [3H]cytarabine, [3H]cladribine, or [3H]fludarabine was inhibited in CEM cells by all five BCR-ABL TKIs but to different extents. Earlier results (26) showed inhibition of accumulation of fludarabine triphosphate in T-lymphocytes exposed to imatinib and fludarabine simultaneously and similar results were later shown by Naud et al. (20) with imatinib and nilotinib on cytarabine uptake in chronic myelogenous leukemia cell lines. Our results with first, second, and third generation BCR-ABL TKIs in a T-lymphocytic cell line are consistent, and illustrate potential difficulties in administering BCR-ABL TKIs with nucleoside drugs in combination therapies.

View full text