Influence of P-glycoprotein on brain uptake of [18F]MPPF in rats

https://doi.org/10.1016/S0014-2999(00)00752-4Get rights and content

Abstract

The aim of this study was to determine if the brain uptake of 4-(2′-methoxyphenyl)-1-[2′-(N-2″-pyridinyl)-p-[18F]fluorobenzamido]ethylpiperazine ([18F]MPPF), a radioligand for the imaging of 5-HT1A receptors, is influenced by the action of P-glycoprotein.

Anesthetized male Wistar rats were injected i.v. with the 5-HT1A receptor antagonist [18F]MPPF (2 MBq, S.A.>110 TBq/mmol) after treatment with saline (controls) or with the 5-HT1A receptor antagonist 1-(2′-methoxyphenyl)-4-[4-(2-phtalimido)butyl]piperazine (NAN-190) (2.5 mg/kg i.v.). After 60 min, the animals were sacrificed and 13 areas of the brain were dissected for ex vivo gamma counting. The regional distribution of radioactivity was also assessed in brain slices using a storage phosphor system. Modulation of P-glycoprotein was achieved by injection of cyclosporin A (50 mg/kg) 30 min prior to injection of [18F]MPPF.

The distribution of 18F-derived radioactivity corresponded to regional 5-HT1A receptor density as known from autoradiography. Modulation of P-glycoprotein with cyclosporin A caused a 5- to 10-fold increase in the uptake of [18F]MPPF. Tissue/cerebellum ratios in the brain correlated with receptor densities determined by in vitro autoradiography. Measurements of plasma radioactivity showed that the increased brain uptake of [18F]MPPF is partially due to a rise in ligand delivery after treatment with cyclosporin A (area under the curve, AUC, increased by a factor of 1.8). Biodistribution experiments in wild type and mdr1a(−/−) knockout mice confirmed that [18F]MPPF is a substrate for P-glycoprotein.

Introduction

Positron emission tomography (PET) and biodistribution studies in rats, cats, and monkeys have demonstrated that the regional distribution of 4-(2′-methoxyphenyl)-1-[2′-(N-2″-pyridinyl)-p-[18F]fluorobenzamido]ethylpiperazine ([18F]MPPF) in the brain corresponds to 5-HT1A receptor localization Kung et al., 1996, Shiue et al., 1997, Le Bars et al., 1998, Ginovart et al., 2000. Recently, our group presented the first human data for [18F]MPPF Passchier et al., 1999, Passchier et al., 2000a, Passchier et al., 2000b. Again, the radioactivity distribution in human CNS was in good agreement with known receptor localization and with previous PET studies, which employed [carbonyl-11C]WAY 100635 Farde et al., 1998, Gunn et al., 1998, Ito et al., 1999. Shortly afterwards, these results were confirmed by researchers from Liège (Plenevaux et al., 1999), indicating that [18F]MPPF is a useful radioligand for clinical application and may be employed to measure the 5-HT1A receptor occupancy of new drugs during phase 1 and 2 clinical trials.

Comparison of data obtained for [18F]MPPF in rats with data for [carbonyl-11C]WAY 100635, showed that MPPF displays a six- to seven-fold lower uptake in the brain (data not shown). This difference must arise from the p-fluoro-benzyl group in MPPF since the compounds are otherwise identical (Scheme 1).

It is known that P-glycoprotein, an ATP-driven transmembrane efflux pump, which is present with high density in brain endothelium, has a high affinity for lipophilic molecules of moderate weight and size that contain cationic centers and planar aromatic domains Abbott and Romero, 1996, Dellinger et al., 1992, Pawagi et al., 1994, Pearce et al., 1990, Schinkel et al., 1996, Van Asperen et al., 1996, Van Asperen et al., 1997. Active transport back into the blood could result in a lower brain penetration of [18F]MPPF than might be expected from its lipophilicity. The efflux action of P-glycoprotein can be inhibited by so-called modulators (e.g. cyclosporin A, verapamil, and PSC833) Begley, 1992, Hendrikse et al., 1998, Hughes et al., 1998, Schinkel et al., 1995, Van Asperen et al., 1996. These compounds reduce P-glycoprotein functionality and thus prevent a radioligand from being expelled from the endothelial membrane back into the blood.

In rodents, two P-glycoproteins are present encoded by the mdr1a and mdr1b genes. Brain P-glycoprotein is expressed by the mdr1a gene Borst and Schinkel, 1996, Schinkel et al., 1995, Schinkel et al., 1996, Van Asperen et al., 1996. The generation of mice with homozygously disrupted mdr1a genes [mdr1a(−/−) knockout mice] has made it possible to test if the brain uptake of a drug is affected by the action of P-glycoprotein Schinkel et al., 1995, Schinkel et al., 1996, Van Asperen et al., 1996.

If uptake of the radiopharmaceutical in the brain is indeed limited by the action of P-glycoprotein, inhibition of this pump by a modulator could result in improved counting statistics within the CNS. A positive result will demonstrate the importance of taking the action of P-glycoprotein into account during the development of radioligands for CNS receptors and for 5-HT1A receptor antagonists in particular.

The aim of the present study was to investigate if the introduction of the extra aromatic ring in MPPF results in a decreased brain uptake due to the efflux action of P-glycoprotein. To that purpose, biodistribution experiments were performed in rats with modulated P-glycoprotein functionality and in mdr1a(−/−) knockout mice.

Section snippets

Materials and methods

All solvents were of analytical grade and were obtained from Merck (Darmstadt, Germany) or Rathburn (Walkerburn, Scotland). Chemicals were purchased from Aldrich (Milwaukee, USA) and ACROS Chimica (Geel, Belgium). NAN-190 Hydrobromide and (R)-(+)-8-Hydroxy-DPAT were from Sigma (St. Louis, USA) Cyclosporin A (Sandimmune) was a product of Sandoz (Basle, Switzerland). Ketamine (Ketalar®) was purchased from Parke-Davis (Hoofddorp, The Netherlands) and xylazine (Rompun®) from Bayer (Leverkusen,

[18F]MPPF uptake and distribution in rat brain

We measured a logP of 2.14±0.06 (n=5) for [18F]MPPF. On the basis of this value, a good uptake of the radioligand in the brain could be expected. However, measured uptake values in biodistribution experiments were quite low. The uptake of [18F]MPPF in the brain of saline- and NAN-190 pretreated Wistar rats is presented in Fig. 1. Standard uptake values were in the range of 0.05–0.25. A significant reduction caused by NAN-190 (2.5 mg/kg) was observed in hippocampus, amygdala/piriform-,

Discussion

Biodistribution experiments showed a five- to seven-fold lower brain uptake for the 5-HT1A receptor antagonist MPPF compared to the prototype ligand WAY 100635 (Fig. 1A) Plenevaux et al., 1997, Shiue et al., 1997, Pike et al., 1994. This was unexpected since the lipophilicity of MPPF is sufficient for rapid diffusion through the blood–brain barrier (logP=2.14). Since biodistribution is a rather crude method that does not provide information on ligand uptake in small (greater than a few

References (35)

  • H.L. Pearce et al.

    Structural characteristics of compounds that modulate P-glycoprotein-associated multidrug resistance

    Adv. Enzyme Regul.

    (1990)
  • L. Rydelek-Fitzgerald et al.

    NAN-190: agonist and antagonist interactions with brain 5-HT1A receptors

    Brain Res.

    (1990)
  • J. Van Asperen et al.

    The functional role of P-glycoprotein in the blood–brain barrier

    J. Pharm. Sci.

    (1997)
  • L.E. Arvidsson et al.

    8-Hydroxy-2-(di-n-propylamino)tetralin, a new centrally acting 5-hydroxytryptamine receptor agonist

    J. Med. Chem.

    (1981)
  • M. Dellinger et al.

    Structural requirements of simple organic cations for recognition by multidrug-resistant cells

    Cancer Res.

    (1992)
  • L. Farde et al.

    Quantitative analyses of carbonyl-carbon-11-WAY-100635 binding to central 5-hydroxytryptamine-1A receptors in man

    J. Nucl. Med.

    (1998)
  • N. Ginovart et al.

    In vivo characterization of p-[(18)F]MPPF, a fluoro analog of WAY-100635 for visualization of 5-HT(1a) receptors

    Synapse

    (2000)
  • Cited by (60)

    • <sup>18</sup>F-FCWAY, a serotonin 1A receptor radioligand, is a substrate for efflux transport at the human blood-brain barrier

      2016, NeuroImage
      Citation Excerpt :

      The authors interpreted the results to mean that 5-HT1A receptors were decreased in the temporal lobe of patients, but the findings could also reflect increased removal of the radioligand from brain. In fact, other 5-HT1A radioligands with a structure similar to that of FCWAY (Liow et al. 2007; Passchier et al. 2000) have been reported to be substrates for permeability-glycoprotein (P-gp), one of the two most prevalent efflux transporters at the blood–brain barrier. In addition, most patients in that study were drug-resistant.

    • 5-HT2A receptor SPECT imaging with [<sup>123</sup>I]R91150 under P-gp inhibition with tariquidar: More is better?

      2016, Nuclear Medicine and Biology
      Citation Excerpt :

      This has led to the development of strategies to overcome the impact of P-gp activity on PET and SPECT preclinical and clinical imaging with pharmacological inhibition of P-gp. In this context, previous studies have demonstrated the potential of two P-gp inhibitors, cyclosporine A (CsA) [2,6–8] and tariquidar (TQD) [5,9,10] to augment brain uptake of radiotracers and signal-to-noise ratio. However, translating the aforementioned effects to an amelioration of quantitative measures is not straightforward.

    View all citing articles on Scopus
    View full text