Personalised Medicine
Enhanced brain accumulation of pazopanib by modulating P-gp and Bcrp1 mediated efflux with canertinib or erlotinib

https://doi.org/10.1016/j.ijpharm.2012.05.038Get rights and content

Abstract

Primary objective of this investigation was to delineate the differential impact of efflux transporters P-glycoprotein (P-gp/Abcb1) and breast cancer resistance protein (Bcrp1/Abcg2) on brain disposition and plasma pharmacokinetics of pazopanib. In addition, this research investigated whether inhibition of these efflux transporters with clinically relevant efflux modulators canertinib or erlotinib could be a viable strategy for improving pazopanib brain delivery. In vitro assays with MDCKII cell monolayers suggested that pazopanib is a high affinity substrate for Bcrp1 and a moderate substrate for P-gp. Co-incubation with specific transport inhibitors restored cell accumulation and completely abolished the directionality of pazopanib flux. Brain and plasma pharmacokinetic studies were conducted in FVB wild type mice in the absence and presence of specific transport inhibitors. Drug levels in plasma and brain were determined using a validated high performance liquid chromatography method using vandetanib as an internal standard. In vivo studies indicated that specific inhibition of either P-gp (by zosuquidar or LY335979) or Bcrp1 (by Ko143) alone did not significantly alter pazopanib brain accumulation. However, dual P-gp/Bcrp1 inhibition by elacridar (GF120918), significantly enhanced pazopanib brain penetration by ∼5-fold without altering its plasma concentrations. Thus, even though Bcrp1 showed higher affinity towards pazopanib in vitro, in vivo at the mouse BBB both P-gp and Bcrp1 act in concert to limit brain accumulation of pazopanib. Furthermore, erlotinib and canertinib as clinically relevant efflux modulators efficiently abrogated directionality in pazopanib efflux in vitro and their co-administration resulted in 2–2.5-fold increase in pazopanib brain accumulation in vivo. Further pre-clinical and clinical investigations are warranted as erlotinib or canertinib may have a synergistic pharmacological effect in addition to their primary role of pazopanib efflux modulation as a combination regimen for the treatment of recurrent brain tumors.

Introduction

Recurrent brain tumors still remain one of the most lethal forms of solid tumors with poor prognosis. Antiangiogenic therapy is at the forefront of current clinical practice for management of highly vascularized brain tumors. The role of epidermal growth factor receptor (EGFR), platelet derived growth factor receptor (PDGFR) and vascular endothelial growth factor receptors (VEGFR) in angiogenesis and cell proliferation is very well established (Hermanson et al., 1992, Plate et al., 1992). Pazopanib (GW786034, Votrient®) is an orally active, second generation tyrosine kinase inhibitor (TKI) that targets VEGFR-1, -2, and -3, PDGFR-α, PDGFR-β, and c-Kit. It has been recently approved by the United States Food and Drug Administration (FDA) for the treatment of metastatic renal cell carcinoma (Ward and Stadler, 2012). Recently in a Phase II study, pazopanib was evaluated for its efficacy in recurrent glioblastoma (Iwamoto et al., 2010). Although pazopanib showed in situ biological activity, but as monotherapy was unable to prolong the progression free survival (PFS) in patients.

It is now well established that ATP binding cassette (ABC) efflux transporters restrict the entry of chemotherapeutic agents across BBB (Miller, 2010). Apical localization and overlapping substrate specificity of P-glycoprotein (P-gp) and breast cancer resistance protein (BCRP) regulate central nervous system penetration and toxicity. Breast cancer resistance protein in addition to P-gp has shown significantly higher affinity toward small molecule TKIs (Agarwal et al., 2011, Breedveld et al., 2005); overexpression of P-gp and BCRP at tumor cell surface further compromises brain penetration of anti-tumor agents (Gottesman et al., 2002). No reports yet exist relating to the failure of pazopanib in glioblastoma treatment. We hypothesize that active efflux at the BBB could be partially responsible for sub-therapeutic concentrations of pazopanib attained in tumor tissue.

A viable strategy to improve brain penetration of compounds that are substrates of efflux proteins is to inhibit the efflux activity at the BBB (Breedveld et al., 2006). Such inhibitors may significantly enhance the brain distribution of substrate drug molecules to therapeutically relevant concentrations in the target tumor tissue located behind an intact BBB. Inhibition of P-gp at BBB using selective transport inhibitors has yielded encouraging results in preclinical studies. Kemper et al. (2004) reported enhanced brain penetration (∼5.6-fold) of paclitaxel in mice upon co-administration of a specific P-gp inhibitor zosuquidar trihydrochloride (LY335979). Similar results were reported by Hubensack et al. (2008). More recently, co-operative role of efflux proteins in limiting the brain penetration of several antitumor agents has been shown to be reversed by simultaneous chemical inhibition of P-gp and Bcrp1 using elacridar at mouse BBB (Agarwal et al., 2010, Tang et al., 2012, Chen et al., 2009, Yamasaki et al., 2011). Although much of the data regarding improved brain penetration upon efflux modulation are available from pre-clinical studies, first clinical study was reported by Sasongko et al. (2005) wherein, enhanced brain penetration of verapamil; a probe P-gp substrate, was demonstrated using PET imaging upon co-administration of a potent and specific P-gp inhibitor; cyclosporine A. In another report, Wagner et al. (2009) published similar findings upon co-administration of tariquidar as a P-gp inhibitor using PET imaging.

Thus far, many attempts have been made to overcome multidrug resistance in oncology leading to development of numerous transport inhibitors, with limited clinical success (Tamaki et al., 2011). Although potent, specific efflux modulators may cause systemic toxicity at doses required to modulate efflux activity (Katragadda et al., 2005). Hence, a dual advantage could be achieved if the co-administered molecule has a synergistic pharmacological effect in addition to its primary role of efflux modulation. Erlotinib and canertinib are currently under investigation for the treatment of recurrent brain tumors (Raizer et al., 2010, Slichenmyer et al., 2001). Both these compounds act by inhibiting the tyrosine kinase domain of the EGFR; known to play a pivotal role in tumor cell proliferation. Furthermore, it has been reported that EGFR is overexpressed in 60% of brain tumors (Smith et al., 2001). In addition both erlotinib and canertinib are know to reverse MDR1 and BCRP mediated resistance to paclitaxel and mitoxantrone or topotecan, respectively in cancer cells (Erlichman et al., 2001, Shi et al., 2007). Hence, we selected canertinib and erlotinib as potential candidates for modulating P-gp and BCRP mediated efflux of pazopanib.

The primary aim of this investigation was to demonstrate the interaction of pazopanib with efflux proteins P-gp and BCRP at the BBB and to elucidate their role in pazopanib brain disposition. Furthermore, from the clinical viewpoint, we wanted to investigate if co-administration of erlotinib and canertinib, could prove to be a viable strategy for modulating P-gp and BCRP mediated active efflux of pazopanib at the BBB.

Section snippets

Chemicals

Pazopanib, erlotinib, canertinib and vandetanib were purchased from LC Laboratories (Woburn, MA). Elacridar [GF120918, N-[4-[2-(6,7-dimethoxy-3,4-dihydro-1H-isoquinolin-2-yl)ethyl]-5-methoxy-9-oxo-10H-acridine-4-carboxamide]] was a generous gift from GlaxoSmithKline Ltd. (Research Triangle Park, NC). Ko143 (a fumitremorgin C analog) was procured from Enzo Life Sciences (Plymouth Meeting, PA) and zosuquidar [LY335979] was kindly provided by Dr. Branimir Sikic at Stanford University and Kanisa

Cellular uptake of pazopanib in MDCKII cells

Intracellular accumulation of pazopanib (0.1 μM) was studied in MDCK-WT, MDR1 and Bcrp1 overexpressing cells. Pazopanib accumulation was approximately 5% of WT cells in Bcrp1 overexpressing variant, suggesting the involvement of Bcrp1 in its efflux (Fig. 1B). Pazopanib accumulation in MDR1 overexpressing cells was 60% of WT cells, indicating a moderate effect of P-gp mediated efflux (Fig. 1A). Pre-treatment with specific inhibitors (200 nM Ko143 for Bcrp1, and 1 μM zosuquidar for MDR1) restored

Discussion

Inhibition of VEGFR and PDGFR tyrosine kinases has been the new target for treatment of solid brain tumors. In a recently concluded Phase II study, pazopanib failed to demonstrate any significant improvement of the PFS in brain tumor patients over the control arm (placebo) (Iwamoto et al., 2010). Role of active efflux at the BBB has now been very well established. Such efflux transporters may cause multidrug resistance to numerous anti-tumor agents. It has been shown previously by many research

Acknowledgement

This work was supported by National Institutes of Health grant 1R01 AI071199.

References (33)

  • C. Erlichman et al.

    The HER tyrosine kinase inhibitor CI1033 enhances cytotoxicity of 7-ethyl-10-hydroxycamptothecin and topotecan by inhibiting breast cancer resistance protein-mediated drug efflux

    Cancer Res.

    (2001)
  • M.M. Gottesman et al.

    Multidrug resistance in cancer: role of ATP-dependent transporters

    Nat. Rev. Cancer

    (2002)
  • M. Hermanson et al.

    Platelet-derived growth factor and its receptors in human glioma tissue: expression of messenger RNA and protein suggests the presence of autocrine and paracrine loops

    Cancer Res.

    (1992)
  • M. Hubensack et al.

    Effect of the ABCB1 modulators elacridar and tariquidar on the distribution of paclitaxel in nudemice

    J. Cancer Res. Clin. Oncol.

    (2008)
  • F.M. Iwamoto et al.

    Phase II trial of pazopanib (GW786034), an oral multi-targeted angiogenesis inhibitor, for adults with recurrent glioblastoma (North American Brain Tumor Consortium Study 06-02)

    Neuro Oncol.

    (2010)
  • S. Katragadda et al.

    Role of efflux pumps and metabolising enzymes in drug delivery

    Expert Opin. Drug Deliv.

    (2005)
  • Cited by (58)

    • Global management of brain metastasis from renal cell carcinoma

      2022, Critical Reviews in Oncology/Hematology
    • Current approaches and prospective drug targeting to brain

      2021, Journal of Drug Delivery Science and Technology
    • Chemotherapy for the Management of Cerebral Metastases

      2020, Neurosurgery Clinics of North America
    • Nilotinib Alters the Efflux Transporter-Mediated Pharmacokinetics of Afatinib in Mice

      2019, Journal of Pharmaceutical Sciences
      Citation Excerpt :

      Hence, we hypothesize that combination TKI therapy could potentially reverse efflux transporter-mediated TKI resistance. Currently, investigations on efflux transporter-mediated TKI-TKI interactions are fairly limited.28,29 Therefore, the objectives of the present study were to identify a potent TKI efflux transporter inhibitor and to develop a combination therapy that will enhance the accumulation of afatinib, the model TKI substrate, in vitro and in vivo.

    • Effect of 5,7-dimethoxyflavone on Bcrp1-mediated transport of sorafenib in vitro and in vivo in mice

      2018, European Journal of Pharmaceutical Sciences
      Citation Excerpt :

      Most of the TKIs available on the market have been found to be substrates of these two efflux transporters (Brozik et al., 2011). To overcome efflux transporter-mediated TKI resistance, co-administration of TKIs with efflux transporter inhibitors represents a very promising treatment strategy (Agarwal and Elmquist, 2012; Hegedus et al., 2009; Kodaira et al., 2010; Minocha et al., 2012; Ozvegy-Laczka et al., 2005; Shi et al., 2009b; Sun et al., 2014; Tiwari et al., 2009; Zhang et al., 2016). However, although many compounds have been identified as efflux transporter inhibitors in vitro, most of them failed to show an in vivo effect because the concentrations tested in vitro were not reachable in vivo.

    View all citing articles on Scopus
    View full text