Evaluation of [11C]laniquidar as a tracer of P-glycoprotein: radiosynthesis and biodistribution in rats

https://doi.org/10.1016/j.nucmedbio.2009.03.004Get rights and content

Abstract

At present, P-glycoprotein (P-gp) function can be studied using positron emission tomography (PET) together with a labelled P-gp substrate such as (R)-[11C]verapamil. Such a tracer is, however, less suitable for investigating P-gp (over)expression. Laniquidar is a third-generation P-gp inhibitor, which has been used in clinic trials for modulating multidrug resistance transporters. The purpose of the present study was to develop the radiosynthesis of [11C]laniquidar and to assess its suitability as a tracer of P-gp expression.

The radiosynthesis of [11C]laniquidar was performed by methylation of the carboxylic acid precursor with [11C]CH3I. The product was purified by HPLC and reformulated over a tC18 Seppak, yielding a sterile solution of [11C]laniquidar in saline. For evaluating [11C]laniquidar, rats were injected with 20 MBq [11C]laniquidar via a tail vein and sacrificed at 5, 15, 30 and 60 min after injection. Several tissues and distinct brain regions were dissected and counted for radioactivity. In addition, uptake of [11C]laniquidar in rats pretreated with cyclosporine A and valspodar (PSC 833) was determined at 30 min after injection. Finally, the metabolic profile of [11C]laniquidar in plasma was determined.

[11C]Laniquidar could be synthesized in moderate yields with high specific activity. Uptake in brain was low, but significantly increased after administration of cyclosporine A. Valspodar did not have any effect on cerebral uptake of [11C]laniquidar. In vivo rate of metabolism was relatively low. Further kinetic studies are needed to investigate the antagonistic behaviour of [11C]laniquidar at tracer level.

Introduction

The blood–brain barrier (BBB) is the main barrier between blood and brain. Its purpose is to maintain homeostasis in the central nervous system (CNS). Furthermore, it protects the CNS from endo- and exogenous toxins that are circulating in blood [1], [2].

Only small lipophilic compounds can enter the brain by passive diffusion via the cell membrane. In addition, very small hydrophilic molecules can penetrate the brain via tight junctions between the cells [3]. All other molecules have to pass the BBB via active transport systems, such as active influx and efflux transporters. Several transporters have been identified. The most important, according to our current knowledge, and best characterized efflux transporter is P-glycoprotein (P-gp) [4].

Changes of P-gp expression in the BBB may play a role in several brain disorders and therefore it is of interest to measure its functionality with positron emission tomography (PET) [5], [6]. In recent years, several PET tracers have been developed and characterised in vitro and in vivo for imaging P-gp. Amongst others, these are [94mTc] complexes, [64Cu] complexes, [68Ga] complexes, [11C]loperamide, [11C]desmethyl loperamide and [11C]verapamil [7], [8], [9], [10], [11], [12], [13], [14].

[11C]Verapamil is the best validated P-gp PET tracer and has been already used in humans [15], [16], [17]. Initially, the racemic mixture was used, but more recently enatiomerically pure (R)-[11C]verapamil was developed enabling quantification [18] and used in PET studies [19], [20], [21], [22], [23].

There are, however, several limitations associated with the use of (R)-[11C]verapamil, especially its extensive peripheral metabolism, in which radioactive metabolites are formed that can penetrate the BBB and also are substrates for P-gp. In addition, polar radiolabelled metabolites are formed, which might result in a non-P-gp-mediated signal [24]. Moreover, effects of medication (antiepileptic drugs) on peripheral metabolism of (R)-[11C]verapamil are found in epilepsy patients [21]. From these results, it was concluded that an average arterial plasma input function derived from healthy volunteers cannot be used for the analysis of patient studies.

Overexpression of multidrug transporters (MDR-1) at the BBB reduces drug penetration into cerebral tissue. This has been shown to be relevant in patients with epilepsy and brain tumours [6], [25]. In case of overexpression of P-gp, cerebral uptake of (R)-[11C]verapamil will also be reduced, resulting in poor counting statistics and, consequently, in reduced precision of PET measurements. To measure overexpression of P-gp, a better strategy would be to use a specific inhibitor instead of a substrate of P-gp, such as laniquidar. Laniquidar is a third-generation inhibitor, which has been used in Phase I and II clinical trials [26], [27] as well as in extensive animal studies [28]. Laniquidar is a noncompetitive inhibitor, actively binds with high affinity to the P-gp transporter (IC50 value of 0.51 μM) and is less active as a substrate [29]. Therefore it is likely that it will not actively be transported out of the brain, but will remain in cerebral tissue. As P-gp contains several binding sites, its overexpression should lead to increased uptake of [11C]laniquidar. Other potential advantages of [11C]laniquidar are its high selectivity and lack of interactions with cytochrome P450 isoenzymes and therefore insensitive to liver metabolism, which in theory should lead to relatively low levels of labelled metabolites of [11C]laniquidar [30].

The purpose of the present study was to develop a synthesis for [11C]laniquidar and to evaluate [11C]laniquidar as a potential P-gp tracer both in control rats and in rats pretreated with P-gp modulators cyclosporine-A and valspodar (PSC 833).

Section snippets

General

Laniquidar used in this study was kindly donated by Johnson & Johnson Pharmaceutical Research and Development (Beerse, Belgium). Valspodar (PSC833) was a gift from Novartis (Basel, Switzerland). All chemicals used were purchased from Sigma-Aldrich. [11C]CO2 was produced using an IBA Cyclone 18/9 cyclotron of the BV Cyclotron VU. [11C]CH3I was synthesized as reported previously [31], and all syntheses were performed using home-made synthesis modules [32]. HPLC columns for purification, quality

Chemistry

The precursor, R102207, was synthesized with a yield of 65%. HPLC analyses with UV detection showed no chemical impurities (see Fig. 2).

The methylation reaction was optimized by investigating the effects of several parameters, like amount of precursor, solvent, solvent volume, temperature, reaction time and amount of base, on radiochemical yield (see Table 1).

With 2 mg of R102207 dissolved in 350 μl DMSO, a reaction time of 2 min at 60°C and 1.2 μl tetrabutylammoniumhydroxide (TBAOH) (60% in

Discussion

This study reports the first radiosynthesis and biodistribution in rats of the highly selective P-gp inhibitor [11C]laniquidar.

The precursor, R102207, was synthesized in fair yield by hydrolysis of the methyl ester of laniquidar with sodium hydroxide. To be able to dissolve laniquidar, 50% THF in water was used as solvent. Special attention was paid to analysing the product obtained, because laniquidar itself should not be present as a contaminant, as this would decrease specific activity

Conclusion

[11C]Laniquidar, a P-gp inhibitor, was synthesized in moderate yields, sufficient for initial biodistribution studies. Although in vivo rate of metabolism of [11C]laniquidar was relatively low, cerebral uptake was lower than expected. Cerebral uptake significantly increased after administration of cyclosporine A, but was not affected by the highly specific inhibitor valspodar. Further kinetic studies are needed to investigate the transport for P-gp of [11C]laniquidar at a tracer level.

Acknowledgments

Johnson & Johnson and Novartis are acknowledged for the donation of laniquidar and valspodar, respectively. The BV Cyclotron VU is acknowledged for providing [11C]CO2. Dr. Koos Herscheid is gratefully acknowledged for his assistance with the animal study.

References (40)

  • de LangeE.C. et al.

    Considerations in the use of cerebrospinal fluid pharmacokinetics to predict brain target concentrations in the clinical setting: implications of the barriers between blood and brain

    Clin Pharmacokinet

    (2002)
  • PardridgeW.M.

    CNS drug design based on principles of blood–brain barrier transport

    J Neurochem

    (1998)
  • TaylorE.M.

    The impact of efflux transporters in the brain on the development of drugs for CNS disorders

    Clin Pharmacokinet

    (2002)
  • KuhnkeD. et al.

    MDR1-P-Glycoprotein (ABCB1) mediates transport of Alzheimer's amyloid-beta peptides—implications for the mechanisms of Abeta clearance at the blood–brain barrier

    Brain Pathol

    (2007)
  • MarchiN. et al.

    Significance of MDR1 and multiple drug resistance in refractory human epileptic brain

    BMC Med

    (2004)
  • BigottH.M. et al.

    Production, processing and uses of 94mTC

    J Lab Compd Radiopharm

    (2001)
  • LewisJ.S. et al.

    Copper bis(diphosphine) complexes: radiopharmaceuticals for the detection of multi-drug resistance in tumours by PET

    Eur J Nucl Med

    (2000)
  • HendrikseN.H. et al.

    In vivo measurement of [11C]verapamil kinetics in human tissues

    Eur J Clin Pharmacol

    (2001)
  • LazarovaN. et al.

    Synthesis and evaluation of [N-methyl-11C]N-desmethyl-loperamide as a new and improved PET radiotracer for imaging P-gp function

    J Med Chem

    (2008)
  • ZoghbiS.S. et al.

    [11C]Loperamide and its N-desmethyl radiometabolite are avid substrates for brain permeability-glycoprotein efflux

    J Nucl Med

    (2008)
  • Cited by (65)

    • Natural products as multidrug resistance modulators in cancer

      2019, European Journal of Medicinal Chemistry
      Citation Excerpt :

      QSAR approach has opted in the development of third generation drugs to overcome the limitations of both first and second generation modulators [61]. Elacridar (11), laniquidar (12) [62], zosuquidar (13) [63], dofequidar (14), mitotane (15) [64], annamycin (16), and tariquidar (17) [65], are the members of third generation modulators category. In contrast to first and second generation modulators, these are more specific and potent with diminutive pharmacokinetic interactions [66–68].

    View all citing articles on Scopus

    The presented work was made possible by a financial contribution from the European Community's Seventh Framework Programme, project EURIPIDES, HEALTH-F5-2007-201380.

    View full text