Elsevier

Biochemical Pharmacology

Volume 78, Issue 2, 15 July 2009, Pages 153-161
Biochemical Pharmacology

Nilotinib (AMN107, Tasigna®) reverses multidrug resistance by inhibiting the activity of the ABCB1/Pgp and ABCG2/BCRP/MXR transporters

https://doi.org/10.1016/j.bcp.2009.04.002Get rights and content

Abstract

Nilotinib, a BCR-Abl tyrosine kinase inhibitor (TKI), was developed to surmount resistance or intolerance to imatinib in patients with Philadelphia positive chronic myelogenous leukemia. Recently, it was shown that several human multidrug resistance (MDR) ATP-binding cassette (ABC) proteins could be modulated by specific TKIs. MDR can produce cancer chemotherapy failure, typically due to overexpression of ABC transporters, which are involved in the extrusion of therapeutic drugs. Here, we report for the first time that nilotinib potentiates the cytotoxicity of widely used therapeutic substrates of ABCG2, such as mitoxantrone, doxorubicin, and ABCB1 substrates including colchicine, vincristine, and paclitaxel. Nilotinib also significantly enhances the accumulation of paclitaxel in cell lines overexpressing ABCB1. Similarly, nilotinib significantly increases the intracellular accumulation of mitoxantrone in cells transfected with ABCG2. Furthermore, nilotinib produces a concentration-dependent inhibition of the ABCG2-mediated transport of methotrexate (MTX), as well as E217βG a physiological substrate of ABCG2. Uptake studies in membrane vesicles overexpressing ABCG2 have indicated that nilotinib inhibits ABCG2 similar to other established TKIs as well as fumitremorgin C. Nilotinib is a potent competitive inhibitor of MTX transport by ABCG2 with a Ki value of 0.69 ± 0.083 μM as demonstrated by kinetic analysis of nilotinib. Overall, our results indicate that nilotinib could reverse ABCB1- and ABCG2-mediated MDR by blocking the efflux function of these transporters. These findings may be used to guide the design of present and future clinical trials with nilotinib, elucidating potential pharmacokinetic interactions. Also, these findings may be useful in clinical practice for cancer combination therapy with nilotinib.

Introduction

One of the primary impediments to the successful treatment of cancer is the presence of cells that have acquired the multidrug resistance (MDR) genotype [1], [2]. MDR is often a result of the overexpression of some ATP-dependent transporters that are known as ATP-binding cassette (ABC) transporters. ABC transporters are ubiquitous, in which the most diverse and the largest superfamily of transporters are present on plasma membranes. A majority of the 49 ABC transporters are classified on the basis of two highly conserved nucleotide-binding domains (NBDs), which are located on the cytosolic site [3]. These NBDs have conserved Walker A and B consensus sequences of 90–110 amino acids, where ATP binds and is hydrolyzed via an ATPase enzyme [4]. The energy derived from ATP regulates the transport of various organic molecules, such as vitamins, saccharides, proteins, and several hydrophobic drugs, both intracellularly and extracellularly [5]. It is well established that these ABC transporters, particularly the ABC transporter-subfamily B member 1 (ABCB1/P-gp/MDR1), -subfamily G member 2 (ABCG2/BCRP/MXR/ABCP), and -subfamily C member 1 (ABCC1/MRP1), play a crucial role in the efflux of important chemotherapeutic agents from cells, thereby, inducing MDR [1], [2], [3], [4], [5], [6], [7]. ABCB1 and ABCC1 can also transport various hydrophobic drugs, and ABCC1 can transport some anionic drugs or drug conjugates, including antifolates, certain nucleotides, and also vinca alkaloids [1], [7]. The substrate profile of ABCG2 partially overlaps with that of ABCC1 and ABCB1 tansporters, whose substrates include mitoxantrone (MX), methotrexate (MTX), indolocarbazole topoisomerase I inhibitors, anthracyclines, flavopiridols, daunorubicin, and some camptothecin-derived inhibitors [8]. Also, ABC transporters affect drug disposition by altering absorption, distribution, metabolism, and excretion, as well as their toxicity [9], [10], [11].

In the human genome, more than 500 protein kinases have been identified as molecular switches that catalyze the transition between active and inactive states [12]. Most of these receptors and cytoplasmic protein kinases belong to the serine/threonine and tyrosine kinase families, which play an important role in cellular signaling. Of the 90 genes that code for tyrosine kinases, 32 genes encode non-receptor entities, while the remaining 58 genes encode receptor protein tyrosine kinases (PTKs) [12], [13]. The deregulation of PTKs has been implicated in the development and progression of various solid tumors and hematological malignancies [14]. Recently, a number of tyrosine kinase inhibitors (TKIs) have received FDA approval for the treatment of various types of cancer. Mechanistically, ATP-competitive TKIs directly inhibit autophosphorylation of key tyrosine residues located in the protein kinase activation-loop domain, with subsequent inhibition of substrate phosphorylation and signal transduction. Through binding within a deep cleft between the C-terminal and N-terminal lobes of the conserved kinase domains, TKIs inhibit the access of MgATP to its binding site, thereby preventing the transfer of a phosphate group to a substrate tyrosine residue subsequently abrogating the enzyme's catalytic function in mediating intracellular signaling [15]. There are a significant number of studies indicating that the currently approved TKIs act as anticancer drugs by inhibiting cancer development, proliferation, invasion, metastasis, angiogenesis, and the induction of apoptosis [16], [17].

Currently, no drug has been clinically approved for the reversal of MDR due to detrimental pharmacokinetic interactions, including toxicity issues. Recently, it has been reported that certain clinically used TKIs interact with ABCB1 and ABCG2 transporters [18], [19], [20], [21], [22], [23], [24], [25]. Since hydrophobic TKIs target the intracellular PTK domain, it was hypothesized that some transmembrane transporters, such as ABC transporters, might also modulate the pharmacological activity of specific TKIs [18]. Interestingly, in vitro studies have shown that submicromolar concentrations of several 4-anilinoquinazoline-derived TKIs, such as canertinib, EKI-485, gefitinib, tyrphostin AG1478, erlotinib, lapatinib, and a phenylamino-pyrimidine derivative imatinib (Fig. 1) can selectively modulate MDR protein–ATPase activity, inhibit MDR-dependent active drug efflux, and significantly affect the drug resistance patterns in cells with MDR mediated by ABCB1 and ABCG2 transporters [18], [19], [20], [21], [22], [23], [24], [25]. In vivo studies have suggested that the co-administration of gefitinib enhances oral bioavailability and hence antitumor activity of irinotecan [26]. Gefitinib also decreases the clearance and increases the oral absorption of topotecan by modulating ABCB1 and ABCG2 function in mice [27].

Previously, we have shown that the response to paclitaxel is augmented by lapatinib in ABCB1-overexpressing KBv200 cell xenografts in nude mice [24]. Imatinib has been reported to inhibit ABCG2, enhancing the efficacy of photodynamic therapy by increasing the intracellular accumulation of photosensitizers [28]. In addition, imatinib can significantly reverse topotecan and SN-38 resistance in vitro[29]. The inhibition of ABCB1 and ABCG2 improves the pharmacokinetic delivery of imatinib in brain tumors of mice [30].

Nilotinib (AMN107, Tasigna®) is a phenyl-amino-pyrimidine derivative (Fig. 1), which was designed based on the crystal structure of imatinib–Abl complex. Nilotinib is a potent, relatively selective inhibitor of the tyrosine kinase activities of BCR-Abl, platelet-derived growth factors (PDGFR) and mast/stem-cell growth factor (c-KIT) [31]. A previous study suggested that nilotinib has a high affinity for ABCG2 and that nilotinib is likely to be a substrate of ABCG2 [32]. In addition, a recent study has suggested that nilotinib may also be a substrate of ABCB1 [33]. However, there are no published data indicating if nilotinib can be used to modulate and reverse ABCG2- and ABCB1-mediated MDR. Therefore, in this study, we have sought to determine if nilotinib could modulate ABCB1- and ABCG2-mediated resistance.

Section snippets

Materials

[3H]-MTX (23 Ci/mmol), [3H]-paclitaxel (37.9 Ci/mmol), and [3H]-MX (4 Ci/mmol) were purchased from Moravek Biochemicals, Inc. (Brea, CA). Monoclonal antibodies BXP-34 (against ABCG2) and C-219 (against ABCB1) were acquired from Signet Laboratories, Inc. (Dedham, MA). Anti-actin monoclonal antibody (sc-8432) was obtained from Santa Cruz Biotechnology, Inc. (Santa Cruz, CA). Nilotinib was obtained as a gift from Novartis pharmaceuticals (Basel, Switzerland). Fumitremorgin C (FTC) was synthesized by

Nilotinib significantly enhances the sensitivity of ABCB1- and ABCG2-overexpressing cells to antineoplastic drugs and does not effect the expression of both ABCB1 and ABCG2

Prior to determining the effects of nilotinib to reverse MDR, we examined its effect on the cell viability. The IC50 values of nilotinib alone in ABCG2- and ABCB1-overexpressing cells were ∼10 and ∼30 μM, respectively (data not shown). However, to avoid toxicity in subsequent experiments, the highest concentration of nilotinib used in the reversal experiments was 2.5 μM, a concentration that caused <10% inhibition of growth in all the cell lines used in this study.

Subsequently, we determined

Discussion

Previously, it has been shown that intracellular accumulation of imatinib in the tumor cells was significantly reduced on chronic exposure due to the induction of ABCB1 and ABCG2 transporters, thus leading to pharmacokinetic resistance by decreasing the oral bioavailability of imatinib [40], [41]. In fact, it was seen that the distribution of imatinib in the brain is limited by ABCB1 mediated efflux [30], and that ABCB1 function is responsible for CML resistance on chronic treatment with

Acknowledgements

We are thankful to Novartis for providing us nilotinib. We thank Drs. Michael M. Gottesman (NCI, NIH, Bethesda, USA) for KB-3-1 cells, Shin-ichi Akiyama (Kagoshima University, Japan) for KB-C2 and KB-CV60 cell lines, Susan E. Bates and Robert W. Robey (NIH, USA) for FTC and ABCG2-overexpressing cell lines; Angela Aliberti, Tong Shen (St. John's University) and Yang-min Chen (Mimi) (Montgomery High School, New Jersey) for editorial assistance.

References (53)

  • E.H. Stover et al.

    The small molecule tyrosine kinase inhibitor AMN107 inhibits TEL-PDGFRbeta and FIP1L1-PDGFRalpha in vitro and in vivo

    Blood

    (2005)
  • K.V. Gleixner et al.

    PKC412 inhibits in vitro growth of neoplastic human mast cells expressing the D816V-mutated variant of KIT: comparison with AMN107, imatinib, and cladribine (2CdA) and evaluation of cooperative drug effects

    Blood

    (2006)
  • M.M. Gottesman

    Mechanisms of cancer drug resistance

    Annu Rev Med

    (2002)
  • P. Borst et al.

    Mammalian ABC transporters in health and disease

    Annu Rev Biochem

    (2002)
  • M. Dean et al.

    Evolution of the ATP-binding cassette (ABC) transporter superfamily in vertebrates

    Annu Rev Genomics Hum Genet

    (2005)
  • M.M. Gottesman et al.

    Overview: ABC transporters and human disease

    J Bioenerg Biomembr

    (2001)
  • M.M. Gottesman et al.

    Multidrug resistance in cancer: role of ATP-dependent transporters

    Nat Rev Cancer

    (2002)
  • G.D. Kruh et al.

    The MRP family of drug efflux pumps

    Oncogene

    (2003)
  • Q. Mao et al.

    Role of the breast cancer resistance protein (ABCG2) in drug transport

    AAPS J

    (2005)
  • J.W. Polli et al.

    The role of efflux and uptake transporters in [N-{3-chloro-4-[(3-fluorobenzyl)oxy]phenyl}-6-[5-({[2-(methylsulfonyl)ethyl]amino}methyl)-2-furyl]-4-quinazolinamine (GW572016, lapatinib) disposition and drug interactions

    Drug Metab Dispos

    (2008)
  • S. Shukla et al.

    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

    (2008)
  • S. Shukla et al.

    Sunitinib (Sutent(R), SU11248), a small-molecule receptor tyrosine kinase inhibitor, blocks function of the ABC transporters, P-glycoprotein (ABCB1) and ABCG2

    Drug Metab Dispos

    (2008)
  • G. Manning et al.

    The protein kinase complement of the human genome

    Science

    (2002)
  • D.R. Robinson et al.

    The protein tyrosine kinase family of the human genome

    Oncogene

    (2000)
  • A. Quintas-Cardama et al.

    Flying under the radar: the new wave of BCR-ABL inhibitors

    Nat Rev Drug Discov

    (2007)
  • C. Mamot et al.

    Targeting the epidermal growth factor receptor (EGFR)—a new therapeutic option in oncology?

    Swiss Med Wkly

    (2006)
  • Cited by (188)

    • Updated chemical scaffolds of ABCG2 inhibitors and their structure-inhibition relationships for future development

      2022, European Journal of Medicinal Chemistry
      Citation Excerpt :

      Tyrosine kinases (TKs) are a diverse group of enzymes that catalyzes the transfer of a phosphate group from ATP to target proteins [81,82]. The researchers discovered certain TK inhibitors (compounds 35–42) that inhibit ABCG2 [83–90]. CM082 (35, Fig. 12) is an angiogenesis inhibitor that inhibits both vascular endothelial growth factor receptor (VEGFR) and platelet-derived growth factor receptors (PDGFR).

    View all citing articles on Scopus
    1

    Grant support: This work was supported by funds from St. John's University Tenure Track Faculty Start-Up Funding No. C-0531 (Z.S. Chen) and St. John's University Seed Grant No. 579-1110 (Z.S. Chen).

    View full text