Sunteți pe pagina 1din 8

Eur J Nucl Med Mol Imaging (2009) 36:446–453

DOI 10.1007/s00259-008-0968-x

ORIGINAL ARTICLE

Effect of cyclosporin A administration on the biodistribution


and multipinhole μSPECT imaging of [123I]R91150
in rodent brain
P. Blanckaert & I. Burvenich & S. Staelens &
S. De Bruyne & L. Moerman & L. Wyffels & F. De Vos

Received: 3 March 2008 / Accepted: 21 September 2008 / Published online: 5 November 2008
# Springer-Verlag 2008

Abstract observed in NMRI mice. Also, a dose-response relationship


Purpose P-glycoprotein (Pgp) is an efflux protein found was established between the dose of cyclosporin A and the
amongst other locations in the blood–brain barrier. It is brain uptake of [123I]R91150 in mice. Compared to the con-
important to investigate the effect of Pgp modulation on trol group, a five-fold increase in [123I]R91150 radioactivity
clinically used brain tracers, because brain uptake of the tracer concentration was observed in the brain of Sprague-Dawley
can be altered by blocking of the Pgp efflux transporter. The rats after cyclosporin A treatment (50 mg/kg). Radioactivity
function of Pgp can be blocked with cyclosporin A. concentration in the frontal cortex increased from 0.24±
Methods We investigated the effect of cyclosporin A admin- 0.0092 to 1.58±0.097% injected dose per gram of tissue after
istration on the biodistribution of [123I]R91150 in rodents, treatment with cyclosporin A (at the 1-h time-point). Blood
and the effect of Pgp blocking on the quality of multipinhole radioactivity concentrations did not increase to the same
μSPECT imaging with [123I]R91150. The influence of extent. The cortical activity was displaced by administration
increasing doses of cyclosporin A on the brain uptake of of ketanserin. A metabolite study confirmed that there was no
[123I]R91150 was investigated in NMRI mice. A biodistri- increased metabolism of [123I]R91150 due to cyclosporin A.
bution study with [123I]R91150 was performed in male The visual quality of multipinhole μSPECT images with
Sprague-Dawley rats pretreated with cyclosporin A and not [123I]R91150 in Sprague-Dawley rats improved markedly
pretreated. Brain uptake of [123I]R91150 after cyclosporin A after cyclosporin A pretreatment.
injection was compared to the brain uptake in untreated Conclusion From the results obtained in the biodistribution
animals, and a displacement study with ketanserin was studies, it can be concluded that [123I]R91150 is a substrate
performed in both groups. A multipinhole μSPECT brain for Pgp in rodents. A relationship between the administered
imaging study was also performed using a Milabs U-SPECT-II dose of cyclosporin A and the increase in [123I]R91150
camera in male Sprague-Dawley rats. To exclude the effect of brain radioactivity concentration was established. The
possible metabolites, a metabolite study was also performed. overall quality of our multipinhole μSPECT images with
Results At the highest cyclosporin A dose (50 mg/kg), a [123I]R91150 in rats improved markedly after pretreatment
sevenfold increase in brain radioactivity concentration was of the animals with cyclosporin A.

P. Blanckaert (*) : I. Burvenich : S. De Bruyne : L. Moerman : Keywords Cyclosporin A . P-glycoprotein .


L. Wyffels : F. De Vos [123I]R91150 . 5-HT2A receptors
Laboratory for Radiopharmacy, Faculty of Pharmaceutical Sciences,
Ghent University,
Harelbekestraat 72,
9000 Gent, Belgium Introduction
e-mail: peter.blanckaert@hotmail.com
The serotonin 2A receptor (5-HT2AR) is a well-known G-
S. Staelens
protein-coupled receptor that plays a role in the regulation
MEDISIP, Faculty of Engineering, Ghent University - IBBT,
De Pintelaan 185 (blok B), of a number of central nervous system processes and
9000 Gent, Belgium dysfunctions [1] such as mood, appetite, anxiety and
Eur J Nucl Med Mol Imaging (2009) 36:446–453 447

anorexia. High levels of 5-HT2AR are found in the brain, NMRI mice, and to determine whether the increased brain
primarily in cortical areas (neocortex, entorhinal and pyri- uptake of the tracer was due to a decreased brain efflux. A
form cortex, claustrum), caudate nucleus, nucleus accum- regional brain biodistribution study in Sprague-Dawley rats
bens, olfactory tubercle and hippocampus [2]. Low levels was performed to assess the impact of Pgp modulation on
are found in the cerebellum, among other locations. A the distribution and brain penetration of the tracer. Also, a
number of tracers has been used for PET and SPECT imag- displacement study (with and without cyclosporin A
ing of the 5-HT2AR, for example [11C]MDL100907 [3–5], administration) in Sprague-Dawley rats was performed to
[123I]R91150 (radioiodinated 4-amino-N-1-[3-(4-fluoro- evaluate the specificity of tracer binding. A metabolite
phenoxy)propyl]-4-methyl-4-piperidinyl 5-iodo-2-methoxy- assay was conducted to exclude possible increased tracer
benzamide) [6, 7] and [18F]altanserin [3, 8–11]. metabolism under the influence of cyclosporin A. Finally, a
Several papers have been published recently concerning multipinhole μSPECT imaging study with [123I]R91150
the importance of P-glycoprotein (Pgp) function/modula- was performed using a Milabs U-SPECT-II camera in
tion in PET or SPECT studies [12–15]. Pgp and multidrug Sprague-Dawley rats pretreated with cyclosporin A, to
resistance-associated protein (MRP) are drug efflux pumps asses the influence of Pgp modulation on the quality of the
that play a role in so-called multidrug resistance (MDR), obtained SPECT images.
together with decreased cellular topoisomerase II and
increased cellular glutathione [16]. Several drugs are
substrates for Pgp and MRP, such as different chemother- Materials and methods
apeutic drugs (vincristine, paclitaxel), cardiovascular drugs
(verapamil, digoxin), immunosuppressive drugs (cyclo- Synthesis and analysis of [123I]R91150
sporin A) [17] and antiepileptic drugs [18]. The function
of Pgp can be blocked by MDR modulators such as calcium [123I]R91150 was synthesized according to the original
channel blockers [19] and cyclosporin A. Pgp is expressed method [28] with modification. Radioiodination was per-
in different tissues such as the blood capillaries of the brain formed by direct electrophilic substitution on the precursor
and the adrenal cortex, and in different tumour types such (4-amino-N-[1-[3-(4-fluorophenoxy)-propyl]-4-methyl-4-
as colon cancer and renal cell carcinoma [20]. Due to the piperidinyl]-2-methoxybenzamide). The 5-position of the
function of Pgp in the blood–brain barrier as a drug efflux methoxybenzamide group was iodinated. The precursor
pump, the impact of possible Pgp modulation on PET and was kindly donated by Janssen Pharmaceutics (J&J, Beerse,
SPECT brain scans may be significant: if a tracer is a Pgp Belgium).
substrate, increased or decreased Pgp expression will have Precursor (0.5–1 mg) was dissolved in 350 μl glacial
an impact on tracer brain uptake [21]. Genetic variations in acetic acid in a V-vial, and carrier solution with no added
Pgp function can lead to successful scans in one subject [123I] (NaI in 0.05 M NaOH, Amersham) was added. Cold
and failed scans in another. This effect also needs to be sodium iodide was added as carrier (0.1 nmol cold NaI per
taken into account when performing PET or SPECT 37 MBq of 123I). At 0, 10, 20 and 25 min, 35 μl of a 30%
brain scans in patients treated with possible modulators solution of H2O2 (Sigma-Aldrich Belgium) was added to
of Pgp. the vial. At 30 min the reaction was quenched by adding
Several tracers have already been evaluated for Pgp 1 ml of sodium sulphite solution (126 mg/ml in 3 M NaOH
modulation, including amongst others [11C]verapamil [22– solution). The reaction mixture was transferred to a 2-ml
24], [99mTc]sestamibi [17, 25], [131I]MIBG [26] and [11C]- HPLC loop and passed through a short column (filled with
(R)-(−)-RWAY [21]; all tracers except [131I]MIBG are Pgp Lichroprep RP-8; Merck, Germany) with 1 mM NaOH. The
substrates. [123I]R91150 is an antagonist with very high column was rinsed with 1 mM NaOH and the tracer was
affinity for the 5-HT2AR (Kd 0.11±0.01 nM). Selectivity for purified by injecting onto a HPLC column (Lichrospher RP
other neurotransmitter receptor binding sites (5-HT1A, 5- Select B, 250×4 mm; Merck, Germany). A sterile sodium
HT1B, 5-HT1D, 5-HT2C, 5-HT3, α1 and α2 adrenoreceptors, acetate/ethanol solution (2.28 g NaOAc·3H2O in 60 ml
histamine H1 and dopamine D2 receptors) amounts to at H2O + 40 ml EtOH) was used as eluent. The fraction
least a factor of 50 [27]. [123I]R91150 has been shown to corresponding to [123I]R91150 (TR 18–20 min, 3.2 ml) was
be suitable for imaging of the 5-HT2AR in human brain [6, collected in a sterile vial after filtration through a 0.22-μm
7, 27]. filter (FP 13/0.2 RC-S; Schleicher&Schuell, pretreated with
In this study we investigated the influence of Pgp HPLC eluent) and diluted with water for injection (9.6 ml).
blocking with cyclosporin A on the biodistribution and Quality control of the tracer was performed by injecting an
brain penetration of [123I]R91150. First a dose-response aliquot onto an analytic HPLC column (Waters Lichrospher
study was performed to assess the impact of increasing RP-8, 125×4 mm, 5 μm) using methanol/acetonitrile/water
cyclosporin A dose on the brain uptake of [123I]R91150 in (190:260:550) as eluent.
448 Eur J Nucl Med Mol Imaging (2009) 36:446–453

Cyclosporin A dose-response study in NMRI mice 10 MBq of [123I]R91150. Male Sprague-Dawley rats
(weight 200–250 g, three per group) were divided into
Cyclosporin A formulated in Cremophor EL (polyoxyethy- two groups: one group was treated with cyclosporin A
lated castor oil) was obtained from Sandoz (Sandimmune (50 mg/kg) 1 h before the injection of [123I]R91150; the
Injection, cyclosporin A 50 mg/ml, Cremophor EL 650 mg/ml; second group received no pretreatment. Ketanserin tartrate
Basel, Switzerland) and was diluted with sterile saline (1 mg/kg) was injected into the penile vein 30 min after
solution. Adult male NMRI mice (three per dosage group, injection of [123I]R91150. The animals were killed by
weight 20–25 g) were injected in the tail vein with various decapitation under isoflurane anaesthesia 1 h after tracer
amounts of cyclosporin A (the doses used were 10, 20, 35 injection. Blood was collected and the brain was rapidly
and 50 mg/kg). For the control group, only the vehicle was removed. The brain was dissected into different regions:
injected. [123I]R91150 (300–500 KBq) was administered cortex (frontal, temporal, parietal and occipital cortex),
via the tail vein 60 min after administration of cyclosporin cerebellum and subcortical region. Blood and brain parts
A, and 1 h after tracer injection the animals were killed by were weighed and counted for radioactivity with an
decapitation under isoflurane anaesthesia. Blood was automated gamma counter (Cobra, Packard Canberra).
collected and the brain was rapidly removed. Blood and The results were corrected for decay and tissue radioactivity
brain were weighed and counted for radioactivity in an concentrations are expressed as a percentage of the injected
automated gamma counter (Cobra, Packard Canberra). dose per gram of tissue (% ID/g). To exclude possible
Three aliquots of the injected tracer solution were weighed interference of Cremophor EL, three animals were injected
and counted for radioactivity to determine the injected with only the vehicle (Cremophor EL, saline and 10%
radioactivity dose received by the animals. The results were ethanol) 1 h before [123I]R91150 injection.
corrected for decay, and tissue radioactivity concentrations
are expressed as a percentage of the injected dose per gram Metabolite determination
of tissue (% ID/g tissue, mean±standard deviation values,
n=3). For determination of possible lipophilic metabolites of
[123I]R91150, three male Sprague-Dawley rats (200–250 g)
Regional brain biodistribution in Sprague-Dawley rats were injected with [123I]R91150 (20 MBq) 1 h after
administration of cyclosporin A (50 mg/kg). The animals
The dose of cyclosporin A used was 50 mg/kg [12]. Adult were killed by decapitation 1 h after tracer injection, blood
male Sprague-Dawley rats (weight 200–250 g) were was collected into heparinized tubes, and the brain was
divided into two groups: one group received an injection quickly removed by dissection. The blood was centrifuged
of cyclosporin A (50 mg/kg) into the penile vein 1 h before for 3 min at 3,000×g and the plasma was separated from the
injection of [123I]R91150. The second group received no pellet. Plasma (500 μl) was mixed with acetonitrile (1 ml).
pretreatment. The animals (three per time point) were killed The mixture was vortexed on ice for 10 s and centrifuged
by decapitation under isoflurane anaesthesia at selected for 3 min at 3,000×g. Brain tissue was homogenized in
time-points (10 min, 30 min, 60 min, 2 h and 4 h) after acetonitrile (2 ml) for 30 s and the mixture was centrifuged
injection of [123I]R91150 (5–10 MBq per animal). Blood for 3 min at 3,000×g. For both brain and plasma, an aliquot
and brain were removed, the brain was rapidly dissected of the acetonitrile fraction was spotted onto a silica thin-
and the brain parts were weighed. The radioactivity of the layer chromatography (TLC) plate (Polygram TLC Sil G/
samples was measured with an automated gamma counter UV254 20×20 cm, 200 μm layer thickness; Machery-Nagel,
(Cobra, Packard Canberra). Germany). An aliquot of authentic [123I]R91150 solution
Tissue radioactivity concentrations are expressed as a was also spotted as a reference. The TLC plate was
percentage of the injected dose per gram of tissue (% ID/g). developed in a dichloromethane/methanol mixture (88/12
The results after cyclosporin A treatment were compared to with 5% triethylamine). The TLC plate was dried at room
the values obtained without pretreatment. The effect of the temperature, exposed to a phosphor imaging screen (plate
isoflurane anaesthesia on the brain uptake of [123I]R91150 type SR, 12.5×25.2 cm; Perkin Elmer Life Sciences, USA)
was investigated earlier, and no effect was observed (data for 30 min and analysed using a Cyclone phosphor imager
not shown). (Perkin Elmer Life Sciences).

Displacement study in Sprague-Dawley rats Multipinhole μSPECT imaging in Sprague-Dawley rats

Ketanserin tartrate (1 mg/kg, dissolved in water/ethanol 95/ μSPECT images were acquired on a U-SPECT-II camera
5; Tocris, Bristol, UK) was used for displacement studies. (Milabs, Utrecht, The Netherlands) equipped with a
The amount of radioactivity injected was approximately 5– dedicated 75 pinhole high-resolution rat collimator. Male
Eur J Nucl Med Mol Imaging (2009) 36:446–453 449

Sprague-Dawley rats (250–266 g) were anaesthetized with


isoflurane and injected with cyclosporin A (50 mg/kg).
[123I]R91150 (29.6 MBq) was injected into the tail vein
30 min later. After a further 40 min, a U-SPECT-II scan was
performed using the rat collimator (75 pinholes of 1.0 mm)
for a continuous 60 min (12 frames of 5 min) with list-
mode output enabled. As a reference, animals were also
scanned in a similar protocol but without administration of
cyclosporin A. A 20% photopeak window was selected
around 159 keV, while background correction was per-
formed using two additional 10% windows around 133 and
188 keV. The time-frames from 45 min to 75 min after
injection were selected and the data were reconstructed on
0.75-mm voxels by three iterations of 16 OSEM subsets.
The images were 3-D postfiltered by a 15-pixel Gaussian Fig. 1 Effects of cyclosporin A dose on the blood concentration and
kernel with a width of 1.5 mm. Two regions of interest brain uptake of [123I]R91150 in NMRI mice. The results are expressed
(ROIs) were drawn around the frontal cortex and the as means±standard deviation (n=3 per dosage group)
cerebellum using Amide (OpenSource software package)
with 3-D freehand on the coronal slices. The mean value
and standard deviation over all voxels in the respective The greatest effect was observed at the highest dose of
ROIs were calculated for post-analysis. cyclosporin A used (50 mg/kg). After administration of cyclo-
All experiments were conducted following the principles sporin A at this dose, a brain tracer concentration of 7.16±
of laboratory animal care and the Belgian law on the pro- 0.25% ID/g brain tissue was reached 1 h after tracer injection.
tection of animals. Our research protocol was approved by the For comparison, brain uptake of [123I]R91150 without cyclo-
Ghent University Hospital ethics committee (ECP 06-18). sporin A pretreatment averaged at 1.0±0.08% ID/g tissue.
Lower doses of cyclosporin A were associated with lower
Statistical analysis radioactivity concentrations in the brain. A slight increase in
blood radioactivity concentration was observed after admin-
Differences between [123I]R91150 accumulation in the istration of increasing doses of cyclosporin A. At the highest
normal and the cyclosporin A groups were analysed using cyclosporin A dose used, a radioactivity concentration of
nonparametric analysis (Mann-Whitney U-test). Only p 2.68±0.82% ID/g tissue was observed in the blood. Blood
values <0.05 were considered statistically significant. radioactivity concentration without cyclosporin A pretreat-
ment averaged at 0.70±0.18% ID/g tissue.

Results Regional brain biodistribution in Sprague-Dawley rats

[123I]R91150 synthesis The effect of cyclosporin A administration on the biodis-


tribution of [123I]R91150 in male Sprague-Dawley rats was
[123I]R91150 was prepared from the precursor (4-amino-N- investigated. Radioactivity concentrations in the different
[1-[3-(4-fluorophenoxy)-propyl]-4-methyl-4-piperidinyl]-2- brain regions of the cyclosporin A-treated rats are shown in
methoxybenzamide) with an overall radiochemical yield of Fig. 2, and of the rats with no pretreatment are shown in
75.6±8.6% (n=12; mean±standard deviation). Radiochemi- Fig. 3.
cal purity at the time of injection was always >95%. Specific The influence of cyclosporin A administration on the
activity of the tracer was approximately 350 TBq/mmol. regional brain biodistribution of [123I]R91150 in rats can be
clearly seen by comparing Figs. 2 and 3. On average, there
Cyclosporin A dose-response study in NMRI mice was a five- to sixfold increase in [123I]R91150 concentra-
tion in the brain after cyclosporin A treatment. For
The effects of administering increasing doses of cyclo- example, at 1 h after injection, the radioactivity concentra-
sporin A on the blood concentration and brain uptake of tion in the frontal cortex increased from 0.24±0.0092 to
[123I]R91150 were investigated in male NMRI mice. The 1.58±0.097% ID/g tissue after treatment with cyclosporin
results are shown in Fig. 1. A. In the reference tissue, the cerebellum, radioactivity
Administration of cyclosporin A had a dose-dependent concentration increased from 0.038±0.0023% ID/g tissue at
effect on the brain uptake of [123I]R91150 in NMRI mice. 1 h after injection of [123I]R91150 to 0.45±0.017% ID/g
450 Eur J Nucl Med Mol Imaging (2009) 36:446–453

increased brain uptake could be the result of a higher blood


radioactivity concentration, resulting in an increased supply
of tracer to the brain. Because the increase in blood
radioactivity concentration was not as pronounced after
cyclosporin A treatment, this theory could be ruled out.
To exclude possible effects of Cremophor EL, a blank
test was performed (injecting only the vehicle, diluted ten
times). No effects of Cremophor EL on [123I]R91150
accumulation in the brain of Sprague-Dawley rats were
found (data not shown). These results are in accordance
with the findings of Hendrikse et al. [23].

Displacement study in Sprague-Dawley rats

The effect of displacement of [123I]R91150 with ketanserin


on the biodistribution of [123I]R91150 in the brain of male
Fig. 2 Regional brain biodistribution of [123I]R91150 in male Sprague- Sprague-Dawley rats was investigated. Radioactivity con-
Dawley rats after cyclosporin A treatment (50 mg/kg). The results are centrations of different brain regions in the different
expressed as means±standard deviation (n=3 per time-point)
treatment groups are shown in Fig. 4.
[123I]R91150 was displaced by ketanserin at the 1-h time-
tissue after treatment of the animals with cyclosporin A. point in target tissues (cortex tissues, especially the frontal
The other brain regions (temporal, parietal and occipital cortex) but not in the cerebellum or blood, which is in
cortex) showed similar increases in [123I]R91150 radioac- accordance with the use of the cerebellum as a reference
tivity concentrations after treatment of the animals with region (i.e. reference tissue model).
cyclosporin A. On the other hand, the large increases in The radioactivity concentration in the frontal cortex of the
brain radioactivity concentrations after cyclosporin A animals with no cyclosporin A pretreatment decreased from
treatment were not found in the blood, indicating that the 0.24±0.0092% ID/g tissue in those without ketanserin dis-
increased brain uptake was not the result of an increased placement to 0.18±0.005% ID/g tissue in those with ketan-
supply of tracer to the brain. serin displacement (i.e. a decrease of 25% after displacement
The increase in [123I]R91150 concentrations in the brain with ketanserin). The radioactivity concentration in the frontal
of male Sprague-Dawley rats is as expected after cyclo- cortex of the animals pretreated with cyclosporin A decreased
sporin A treatment because of the effect of Pgp in from 1.58±0.097% ID/g tissue in those without ketanserin
decreasing efflux of tracer out of the brain (since its displacement to 1.14±0.15% ID/g tissue in those with
function is blocked by cyclosporin A). Theoretically, an ketanserin displacement (i.e. a decrease also of 25% after
displacement with ketanserin). As mentioned above, in the
cerebellum there was no significant difference between the
groups with and without ketanserin, in both the animals with
no pretreatment (0.038±0.002 and 0.0379±0.004% ID/g

Fig. 4 Effect of displacement with ketanserin on the biodistribution of


[123I]R91150 in Sprague-Dawley rats. Animals were pretreated with
Fig. 3 Regional brain biodistribution of [123I]R91150 in male cyclosporin A (50 mg/kg) 30 min before tracer injection. Ketanserin
Sprague-Dawley rats with no pretreatment. The results are expressed tartrate was administered 30 min after [123I]R91150. The results are
as means±standard deviation (n=3 per time-point) expressed as means±standard deviation (n=3 per group). *p<0.05
Eur J Nucl Med Mol Imaging (2009) 36:446–453 451

tissue, respectively) and the animals pretreated with cyclo- In general, a low brain uptake was observed after
sporin A (0.456±0.017 and 0.450±0.038% ID/g tissue, injection of [123I]R91150 in the animals without pretreat-
respectively). Radioactivity concentration in the blood did ment. As evident from Fig. 5 part A, hardly any activity
not change significantly after ketanserin displacement. was visible on the μSPECT scans. The count rate obtained
was too low to perform reliable statistical analysis on the
Metabolite study data from the animals without pretreatment. Radioactivity
uptake in the brain was considerably higher after treatment
Metabolites in blood and brain of Sprague-Dawley rats of the animals with cyclosporin A. Highest uptake of [123I]
were determined to exclude possible interference of cyclo- R91150 was observed in the frontal cortex, and the lowest
sporin A with in vivo metabolism of [123I]R91150. No concentration of radioactivity was found in cerebellum. As
lipophilic metabolites were detected in the blood or brain. can be seen in Fig. 5 part B, radioactivity concentrations
The only radiolabelled product found in brain samples was after cyclosporin A treatment were higher than the values
unmetabolized [123I]R91150, by comparing the retention obtained with no cyclosporin A pretreatment.
times with a [123I]R91150 reference standard. Besides Tracer uptake in the cerebellum was used as a reference
unmetabolized [123I]R91150, a small amount of free radio- for nonspecific binding, based on the reference tissue
iodine was present in the blood samples, probably due to in model. For semiquantification, the binding index was
vivo deiodination of the tracer. estimated as (counts/voxel in frontal cortex ROI)/(counts/
voxel in cerebellum ROI). A binding index value of 3.16
Multipinhole μSPECT imaging study was obtained from the μSPECT scans in the animals
pretreated with cyclosporin A. This result is in accordance
The μSPECT images obtained are shown in Fig. 5. Based to the binding index data calculated from the biodistribution
on anatomical data, two ROIs were drawn on the coronal study with [123I]R91150 in Sprague-Dawley rats (a binding
view around the frontal cortex and the cerebellum (Fig. 5). index of around 4 was obtained).

Fig. 5 Multipinhole μSPECT


images with [123I]R91150 in
Sprague-Dawley rats. Animals
were scanned around 1 h after
injection of [123I]R91150
(29.6 MBq): A without pretreat-
ment; B after pretreatment with
cyclosporin A (50 mg/kg).
ROI’s were drawn around front-
al cortex and cerebellum. The
lower part shows high resolu-
tion [123I]R91150 μSPECT
images after cyclosporin A
treatment (transversal, coronal
and sagittal views)
452 Eur J Nucl Med Mol Imaging (2009) 36:446–453

The lower part of Fig. 5 again shows the results of a different radiotracers [22]. Similar to the results obtained
[123I]R91150 scan after administration of cyclosporin A, by Liow et al. [21] with the 5-HT1A tracer [11C]RWAY
but now optimized for optimal spatial resolution (the data (who observed a five-fold increase in brain tracer concen-
were reconstructed on 0.375 mm voxels by three iterations tration after cyclosporin A treatment), brain uptake of [123I]
of 16 OSEM subsets). The images were 3-D postfiltered by R91150 increased four- to sixfold. Our results are also in
a 15 pixel Gaussian kernel with a width of 1.125 mm. agreement with the findings of Hendrikse et al. [23], where
a dose-dependent relationship was observed between the
brain uptake in mice of [11C]verapamil and the dose of
Discussion cyclosporin A; a tenfold increase in brain radioactivity
concentration was found after treatment of the animals with
The human mdr1 gene (that encodes Pgp) has several single cyclosporin A (50 mg/kg).
nucleotide polymorphisms that can change the pharmaco- In the multipinhole μSPECT imaging study, the effect of
kinetics of drugs that are substrates for this transporter [21]. cyclosporin A administration could be seen as an increase
This can lead to successful scans in one patient, and failed in brain uptake of [123I]R91150 (Fig. 5, part B), while the
scans in another, and is very important in patients treated brain was hardly visible when only the tracer [123I]R91150
with Pgp modulators such as cyclosporin A. Therefore it is was administered (Fig. 5, part A), in contrast with, for
necessary in the development of new tracers to determine if example, human trials where the tracer does show ade-
they are modulated by Pgp or not. It is also of interest to quate brain uptake [6, 7]. After pretreatment with cyclo-
know whether the tracers presently used clinically are sporin A, cortical tissues could be clearly distinguished
modulated by Pgp. In this regard, several clinically used (Fig. 5, part B).
tracers have already been examined as Pgp substrates [13, Due to possible peripheral effects of cyclosporin A (for
20, 21, 25, 26, 29]. example CYP3A4 inhibition), we found it necessary to
The Sprague-Dawley rat biodistribution studies with perform a metabolite analysis after cyclosporin A treatment.
[123I]R91150 and the rat μSPECT imaging study revealed a Since no radiolabelled metabolites were found, the in-
four- to sixfold increase in brain [123I]R91150 radioactivity creased radioactivity uptake in the brain can only be
concentration after blocking of Pgp with cyclosporin A. attributed to an increased concentration of authentic [123I]
Theoretically, an increased brain uptake may be the result R91150.
of an increased tracer influx to the brain, or a decreased Future work will investigate possible changes in [123I]
tracer efflux out of the brain. Since the drug efflux protein R91150 free plasma fraction under the influence of cyclo-
function of Pgp was blocked by the administration of sporin A, to exclude possible displacement of the tracer
cyclosporin A, the increase in [123I]R91150 brain uptake from plasma proteins, also resulting in an increased tracer
should have been the result of decreased tracer efflux out of influx to the brain.
the brain. This is in accordance with the localization and
function of Pgp as an efflux protein at the blood–brain
barrier [21]. Moreover, a dose-response relationship was Conclusion
found between the doses of cyclosporin A that were
administered and the tracer concentration in the brain of We evaluated the effect of cyclosporin A pretreatment on
NMRI mice. Although blood radioactivity concentrations the [123I]R91150 radioactivity concentrations in the brain of
did increase slightly after treatment of the mice with NMRI mice and Sprague-Dawley rats. A relationship
cyclosporin A, this increase was not as pronounced as the between the administered dose of cyclosporin A and the
resulting increased tracer concentration in the brain, and radioactivity concentration of [123I]R91150 in rodent brain
could have been the result of an influence of cyclosporin A was established. The effect of cyclosporin A administration
on the pharmacokinetics of [123I]R91150 [30]. on the quality of multipinhole μSPECT images with [123I]
The increased [123I]R91150 radioactivity concentration R91150 was also evaluated in Sprague-Dawley rats. From
in rodent brain after cyclosporin A treatment could not be the results obtained it can be concluded that [123I]R91150 is
fully displaced by ketanserin (Fig. 4), indicating that at least an in vivo substrate for Pgp in rodents. Pretreatment of the
part of the increase in [123I]R91150 uptake is due to animals with cyclosporin A resulted in an increased brain
increased aspecific binding in brain. This is also evident uptake of [123I]R91150 and improved the quality of the
from the increased radioactivity concentration after cyclo- multipinhole μSPECT images obtained.
sporin A treatment in the reference area, the cerebellum. Therefore, we suggest further exploration of the use of
Our results are in accordance with those of other cyclosporin A (or other less toxic Pgp blocking agents) for
previously published studies investigating the effect of improving the otherwise limited brain uptake of some
cyclosporin A administration on the brain uptake of radioligands for brain imaging.
Eur J Nucl Med Mol Imaging (2009) 36:446–453 453

References with a radiolabelled antibody. Eur J Nucl Med Mol Imaging


2006;33:1266–72.
16. Gottesman MM, Pastan I. Biochemistry of multidrug-resistance
1. Heinrich T, Böttche H, Prücher H, Gottschlich R, Ackermann K-A, mediated by the multidrug transporter. Annu Rev Biochem 1993;
van Amsterdam C. 1-(1-Phenethylpiperidin-4-yl)-1-phenylethanols 62:385–427.
as potent and highly selective 5-ht2A antagonists. Chem Med 17. Hendrikse NH, Frans EJ, Van der Graaf WTA, Vaalburg W, De
Chem 2006;1:245–55. Vries EGE. Visualization of multidrug resistance in vivo. Eur J
2. Barnes NM, Sharp T. A review of central 5-HT receptors and their Nucl Med 1999;26:283–93.
function. Neuropharmacology 1999;38:1083–152. 18. Weiss J, Kerpen CJ, Lindenmaier H, Dormann SM, Haefeli WE.
3. Kristiansen H, Elfving B, Plenge P, Pinborg LH, Gillings N, Interaction of antiepileptic drugs with human P-glycoprotein in
Knudsen GM. Binding characteristics of the 5-HT2A receptor vitro. J Pharmacol Exp Ther 2003;307:262–7.
antagonists altanserin and MDL 100907. Synapse 2005;58:249– 19. Herzog CE, Tsokos M, Bates SE, Fojo AT. Increased Mdr-1/P-
57. Glycoprotein expression after treatment of human colon-carcino-
4. Lundkvist C, Halldin C, Ginovart N, Nyberg S, Swahn CG, Carr ma cells with P-glycoprotein antagonists. J Biol Chem 1993;268:
AA, et al. [C-11]MDL 100907, a radioligand for selective imaging 2946–52.
of 5-HT2A receptors with positron emission tomography. Life Sci
20. van Asperen J, Schinkel AH, Beijnen JH, Nooijen WJ, Borst P,
1996;58:L187–92.
van Tellingen O. Altered pharmacokinetics of vinblastine in
5. Mathis CA, Mahmood K, Huang Y, Simpson NR, Gerdes JM,
Mdr1a P-glycoprotein-deficient mice. J Natl Cancer Inst 1996;88:
Price JC. Synthesis and preliminary in vivo evaluation of [C-11]
994–9.
MDL 100907: a potent and selective radioligand for the 5-HT2A
21. Liow JS, Lu SY, McCarron JA, Hong JS, Musachio JL, Pike VW,
receptor system. Med Chem Res 1996;6:1–10.
et al. Effect of a P-glycoprotein inhibitor, cyclosporin A, on the
6. Catafau AM, Danus M, Bullich S, Llop J, Perich J, Cunningham
disposition in rodent brain and blood of the 5-HT1A receptor
VJ, et al. Characterization of the SPECT 5-HT2A receptor ligand
radioligand, [C-11](R)-(-)-RWAY. Synapse 2007;61:96–105.
I-123-R91150 in healthy volunteers: Part 1 – Pseudoequilibrium
22. Elsinga PH, Hendrikse NH, Bart J, van Waarde A, Vaalburg W.
interval and quantification methods. J Nucl Med 2006;47:919–
Positron emission tomography studies on binding of central
28.
nervous system drugs and P-glycoprotein function in the rodent
7. Catafau AM, Danus M, Bullich S, Nucci G, Llop J, Abanades S,
brain. Mol Imaging Biol 2005;7:37–44.
et al. Characterization of the SPECT 5-HT2A receptor ligand I-
123-R91150 in healthy volunteers: part 2 – ketanserin displace- 23. Hendrikse NH, Schinkel AH, De Vries EGE, Fluks E, Van der
ment. J Nucl Med 2006;47:929–37. Graaf WTA, Willemsen ATM, et al. Complete in vivo reversal of
8. Marner L, Knudsen GM, Madsen K, Haugbol S, Vogel A, Holm P-glycoprotein pump function in the blood-brain barrier visualized
S, et al. Longitudinal assessment of cerebral 5-HT2A receptors in with positron emission tomography. Br J Pharmacol 1998;124:
normal volunteers. An [18F]-altanserin PET study. Neuroimage 1413–18.
2006;31:T101. 24. Lee YJ, Maeda J, Kusuhara H, Okauchi T, Inaji M, Nagai Y, et al.
9. Mintun MA, Sheline YI, Moerlein SM, Vlassenko AG, Huang Y, In vivo evaluation of P-glycoprotein function at the blood-brain
Snyder AZ. Decreased hippocampal 5-HT2A receptor binding in barrier in nonhuman primates using [C-11]verapamil. J Pharmacol
major depressive disorder: in vivo measurement with [18F] Exp Ther 2006;316:647–53.
altanserin positron emission tomography. Biol Psychiatry 2004;55: 25. Joseph B, Bhargava KK, Malhi H, Schilsky ML, Jain D, Palestro
217–24. CJ, et al. Sestamibi is a substrate for MDR1 and MDR2 P-
10. Rosier A, Dupont P, Peuskens J, Bormans G, Vandenberghe R, glycoprotein genes. Eur J Nucl Med Mol Imaging 2003;30:1024–
Maes M, et al. Visualisation of loss of 5-HT2A receptors with age 31.
in healthy volunteers using [F-18]altanserin and positron emission 26. Kiyono Y, Yamashita T, Doi H, Kuge Y, Katsura T, Inui KI, et al.
tomographic imaging. Psychiatry Res 1996;68:11–22. Is MIBG a substrate of P-glycoprotein? Eur J Nucl Med Mol
11. Sheline YI, Mintun MA, Moerlein SM, Vlassenko AG, Xue F, Imaging 2007;34:448–52.
Snyder AZ. Decreased hippocampal 5-HT2A receptor binding in 27. Baeken C, D’haenen H, Flamen P, Mertens J, Terriere D, Chavatte
medication naive major depression measured with [18F] altanserin K, et al. I-123-5-I-R91150, a new single-photon emission
positron emission tomography. Biol Psychiatry 2003;53:10S. tomography ligand for 5-HT2A receptors: influence of age and
12. de Vries EFJ, Kortekaas R, van Waarde A, Dijkstra D, Elsinga PH, gender in healthy subjects. Eur J Nucl Med 1998;25:1617–22.
Vaalburg W. Synthesis and evaluation of dopamine D-3 receptor 28. Mertens J, Terriere D, Sipido V, Gommeren W, Janssen PMF,
antagonist C-11-GR218231 as PET tracer for P-glycoprotein. J Leysen JE. Radiosynthesis of a new radioiodinated ligand for
Nucl Med 2005;46:1384–92. serotonin-5Ht(2)-receptors, a promising tracer for gamma-
13. Ishiwata K, Kawamura K, Yanai K, Hendrikse NH. In vivo emission tomography. J Labelled Compd Radiopharm 1994;
evaluation of P-glycoprotein modulation of 8 PET radioligands 34:795–806.
used clinically. J Nucl Med 2007;48:81–7. 29. Takano A, Kusuhara H, Suhara T, Ieiri I, Morimoto T, Lee YJ, et
14. Syvanen S, Blomquist G, Sprycha M, Hoglund AU, Roman M, al. Evaluation of in vivo P-glycoprotein function at the blood-
Eriksson O, et al. Duration and degree of cyclosporin induced P- brain barrier among MDR1 gene polymorphisms by using C-11-
glycoprotein inhibition in the rat blood-brain barrier can be verapamil. J Nucl Med 2006;47:1427–33.
studied with PET. Neuroimage 2006;32:1134–41. 30. Katoh M, Suzuyama N, Takeuch T, Yoshitomi S, Asahi S, Yokoi
15. van Eerd JEM, De Geus-Oei LF, Oyen WJG, Corstens FHM, T. Kinetic analyses for species differences in P-glycoprotein-
Boerman OC. Scintigraphic imaging of P-glycoprotein expression mediated drug transport. J Pharm Sci 2006;95:2673–83.

S-ar putea să vă placă și