Blanckaert 2008

download Blanckaert 2008

of 7

Transcript of Blanckaert 2008

  • 8/7/2019 Blanckaert 2008

    1/7

    In vivo evaluation in rodents of [123

    I]-3-I-CO as a potentialSPECT tracer for the serotonin 5-HT2A receptor

    Peter B.M. Blanckaert, Ingrid Burvenich, Leonie Wyffels,Sylvie De Bruyne, Lieselotte Moerman, Filip De Vos

    Laboratory for Radiopharmacy, Ghent University, B-9000 Ghent, Belgium

    Received 22 May 2008; received in revised form 5 September 2008; accepted 8 September 2008

    Abstract

    Introduction: [123I]-(4-fluorophenyl)[1-(3-iodophenethyl)piperidin-4-yl]methanone ([123I]-3-I-CO) is a potential single photon emission

    computed tomography tracer with high affinity for the serotonin 5-HT2A receptor (Ki=0.51 nM) and good selectivity over other receptor (sub)

    types. To determine the potential of the radioligand as a 5-HT2A tracer, regional brain biodistribution and displacement studies will be

    performed. The influence of P-glycoprotein blocking on the brain uptake of the radioligand will also be investigated.

    Methods: A regional brain biodistribution study and a displacement study with ketanserin were performed with [123I]-3-I-CO. Also, the

    influence of cyclosporin A (50 mg/kg) on the brain distribution of the radioligand was investigated. For the displacement study, ketanserin

    (1 mg/kg) was administered 30 min after injection of [123I]-3-I-CO.

    Results: The initial brain uptake of [123I]-3-I-CO was quite high, but a rapid wash-out of radioactivity was observed. Cortex-to-cerebellum

    binding index ratios were low (1.1 1.7), indicating considerable aspecific binding and a low specific signal of the radioligand. Tracer

    uptake was reduced to the levels in cerebellum (a 60% reduction) after ketanserin displacement. Administration of cyclosporin A resulted in a

    doubling of the brain radioactivity concentration.

    Conclusions: Although [123I]-3-I-CO showed adequate brain uptake and could be displaced by ketanserin, high aspecific binding to brain

    tissue was responsible for very low cortex-to-cerebellum binding index ratios, possibly limiting the potential of the radioligand as a serotonin

    5-HT2A receptor tracer. We also demonstrated that [123I]-3-I-CO is probably a weak substrate for the P-glycoprotein efflux transporter.

    2008 Elsevier Inc. All rights reserved.

    Keywords: Serotonin; P-glycoprotein; 5-HT2A receptor; Brain tracer

    1. Introduction

    Serotonin is a well-known neurotransmitter, regulating

    important functions such as sleep, mood and appetite [1].

    It was demonstrated that serotonergic neurotransmission

    malfunctioning is responsible for several psychiatric

    conditions, for example depression. A subtype of the

    serotonin receptor family, the serotonin 5-HT2A receptor,has been implicated in pathologies such as schizophrenia,

    depression, anorexia and anxiety, both in humans [26]

    and in animals [79]. Imaging of the 5-HT2A receptor with

    SPECT is a valuable tool to aid psychiatrists in the

    diagnosis of these pathologies. Several radioligands have

    already been used to study the 5-HT2A receptor in the

    brain, mostly PET tracers. [11C]-MDL100907 is the most

    frequently used tracer for imaging of the 5-HT2A receptor

    with PET [10]. [18F]-Altanserin [11] and [18F]-setoperone

    [12] have also been used. Currently, only one SPECT

    tracer ([123I]-R91150), has been used clinically for imaging

    of the 5-HT2A receptor [13,14]. Since this radioligand

    shows high aspecific binding in vitro, we decided todevelop a new potential SPECT tracer for imaging of the

    5-HT2A receptor.

    [123I]-(4-fluorophenyl)[1-(3-iodophenethyl)piperidin-4-

    yl]methanone ([123I]-3-I-CO) [15] was synthesized from the

    corresponding tributylstannylprecursor (1) using chlora-

    mine-T in acetic acid (Fig. 1).

    The radioligand is an antagonist with high affinity for the

    5-HT2A receptor (Ki=0.51 nM) and selectivity of at least a

    factor 20 over other receptor (sub)types, including the

    Available online at www.sciencedirect.com

    Nuclear Medicine and Biology 35 (2008) 861867www.elsevier.com/locate/nucmedbio

    Corresponding author. Tel.: +32 9 264 8065; fax: +32 9 264 8071.

    E-mail addresses: [email protected] ,

    [email protected] (P.B.M. Blanckaert).

    0969-8051/$ see front matter 2008 Elsevier Inc. All rights reserved.

    doi:10.1016/j.nucmedbio.2008.09.004

    mailto:[email protected]:[email protected]://dx.doi.org/10.1016/j.nucmedbio.2008.09.004http://dx.doi.org/10.1016/j.nucmedbio.2008.09.004mailto:[email protected]:[email protected]
  • 8/7/2019 Blanckaert 2008

    2/7

    5-HT2C receptor[15]. The potential of the ligand as a single

    photon emission computed tomography (SPECT) tracer for

    the serotonin 5-HT2A receptor tracer was determined by

    performing a regional brain biodistribution study with [123I]-

    3-I-CO in SpragueDawley rats. To demonstrate specific

    binding of the radioligand to the 5-HT2A receptor, a

    displacement study using ketanserin as the displacing agent

    was performed. Also, a metabolite analysis was executed to

    exclude the presence of radiolabelled metabolites in brain.

    Precursor synthesis, radiosynthesis and biodistribution

    studies in mice have already been published elsewhere [16].

    P-glycoprotein is an adenosine triphosphate-driven efflux

    protein located amongst others in the blood-brain barrier. It is

    also over expressed in different tumour types [17]. It was

    demonstrated recently that blocking of P-glycoprotein

    function with cyclosporin A had a profound effect on the

    brain uptake of several radioligands [1820], resulting in a

    strong increase in brain radioactivity concentration.

    P-glycoprotein modulation could prove useful for

    increasing drug concentrations (a.o. chemotherapeutics)

    and in oncology where P-glycoprotein (Pgp) plays a role in

    multidrug-resistance [21]. Also, several central nervoussystem-active drugs such as phenytoin (antiepileptic),

    clomipramine (antidepressant) and chlorpromazine (neuro-

    leptic) are substrates for Pgp. Possibly, their brain concen-

    tration (and, hence, their therapeutic effect) can be altered by

    modulation of Pgp [17]. Radioligands with demonstrated

    Pgp affinity could be used for monitoring Pgp activity in the

    blood-brain barrier. Also, Pgp efflux can reduce brain uptake

    of radiotracers and thus hamper successful imaging [22]. For

    these reasons, the influence of Pgp blocking with cyclos-

    porin A on the regional brain biodistribution of [123I]-3-I-CO

    in rodents was also investigated.

    2. Materials and methods

    2.1. Chemicals and radiochemicals

    All chemicals and reagents were purchased from Acros

    Organics (Beerse, Belgium) and were used without further

    purification unless described otherwise. Solvents used were

    of high-performance liquid chromatography (HPLC) quality

    and were purchased from Chemlab (Belgium). No carried

    added [123I]-sodium iodide (in 0.05M NaOH) was purchased

    from GE Healthcare. Ketanserin was used as the tartrate salt

    and was obtained from Tocris Cookson (Bristol, UK). It was

    dissolved in 0.9% NaCl solution containing 10% ethanol

    (v/v). Cyclosporin A was obtained from SigmaAldrich and

    was dissolved in a mixture of ethanol and polyethoxylated

    castor oil and diluted with 0.9% NaCl before injection. A

    concentration of 50 mg/kg was used.

    2.2. Chemistry

    Synthesis and radiosynthesis of [123I]-3-I-CO were

    performed as described previously [16]. Briefly, [

    123

    I]-3-I-CO was synthesized starting from the corresponding

    tributylstannylprecursor. The precursor was iodinated

    using chloramine-T in the presence of glacial acetic acid,

    nca [123I]-NaI and ethanol as solvent. Radiosynthesis was

    terminated by the addition of sodiummetabisulphite solu-

    tion. The radioligand was purified on HPLC, using a

    reversed-phase C18 column (Alltech Apollo C18 7250 mm,

    5 m) and a 50/50 mixture of acetonitrile and phosphate

    buffer (0.02 M, pH 7) as the eluent. Solvent was removed

    with a C18 Sep-PAK cartridge, and the radioligand was

    formulated for injection in 0.9% NaCl containing 10%

    ethanol after sterile filtration. A radiochemical yield of

    755% (n=3) was obtained. Radiochemical purity was

    always higher then 95%.

    2.3. Regional brain biodistribution study in rodents

    All animal experiments were conducted according to the

    regulations of the Belgian law and the Ghent University local

    ethical committee (ECP 05/14). [123I]-3-I-CO (35 MBq)

    was injected through the penile vein in male Sprague

    Dawley rats (n=3 animals per time point, 200250 g

    bodyweight). At predefined time points after injection of

    the radioligand (20 min, 40 min, 1 h, 2 h and 4 h), the

    animals were sacrificed by decapitation under isoflurane

    anaesthesia. Blood was collected from the heart and the brainwas rapidly removed and dissected into different regions: the

    cortex (which was further dissected into the frontal cortex,

    parietal cortex, occipital cortex and temporal cortex),

    cerebellum and a subcortical region. The different brain

    and blood samples were weighed and counted for radio-

    activity with an automated gamma counter [Cobra, Packard

    Canberra, equipped with five 11 NaI(Tl) crystals].

    Aliquots of the injected tracer solution (n=3) were weighed

    and counted for radioactivity to determine the injected

    radioactivity dose received by the animals. Results were

    corrected for decay and tissue radioactivity concentrations

    Fig. 1. Radiosynthesis of [123I]-3-I-CO.

    862 P.B.M. Blanckaert et al. / Nuclear Medicine and Biology 35 (2008) 861867

  • 8/7/2019 Blanckaert 2008

    3/7

    were expressed as a percentage of the injected dose per gram

    of tissue (% ID/g tissue, meanS.D. values, n=3 per

    time point).

    2.4. Ketanserin displacement study

    Ketanserin was used as the tartrate salt and was dissolved

    in 0.9 % NaCl solution containing 10% ethanol at aconcentration of 2 mg/ml. A dosage of 1 mg/kg was used

    for displacement experiments. [123I]-3-I-CO (35 MBq)

    was injected through the penile vein in male Sprague

    Dawley rats (n=3 animals per group). Thirty minutes after

    injection of the radioligand, ketanserin tartrate was injected

    through the penile vein. One hour after injection of the

    radioligand, the animals were sacrificed by decapitation

    under isoflurane anaesthesia.

    Blood was collected and the brain was rapidly dissected.

    All tissues were weighed and counted for radioactivity with

    an automated gamma counter. Results were corrected for

    decay and are expressed as % ID/g tissue (meanS.D.).

    2.5. Influence of Pgp modulation

    Cyclosporin A (50 mg) was dissolved in ethanol (100 l).

    Polyethoxylated castor oil (900 l, Cremophore EL, Bayer,

    Germany) was added and the mixture was shaken for 2 min

    on a vortex mixer. The mixture was transferred to a sterile 10

    ml injection vial, and sterile saline solution (9 ml) was added

    to obtain an injectable solution containing 5 mg/ml

    cyclosporin A. This solution could be stored in a refrigerator

    for a maximum period of about two months. Male Sprague

    Dawley rats (n=3 per group) were anesthetized with

    isoflurane and injected in the penile vein with cyclosporin

    A (50 mg/kg body weight). Thirty minutes after cyclosporinA administration, the rats were anesthetized with isoflurane

    and injected in the penile vein with [123I]-3-I-CO (35

    MBq). The animals were sacrificed by decapitation under

    isoflurane anaesthesia at selected time points after injection

    of the radioligand (20 min, 40 min, 60 min, 2 h and 4 h).

    Blood was collected and the brain was rapidly removed and

    dissected into different regions. Brain and blood samples

    were weighed and counted for radioactivity with an

    automated gamma counter. Results were corrected for

    decay and expressed as % ID/g tissue (meanS.D., n=3).

    The results were compared with the normal brain biodis-

    tribution results.

    2.6. Metabolite analysis in rodents

    Male SpragueDawley rats (weight 200250 g, n=3)

    were injected in the penile vein with [123I]-3-I-CO (1020

    MBq). At 30 min post injection, the animals were sacrificed

    by decapitation under isoflurane anaesthesia. Blood was

    collected and the brain was rapidly dissected. Blood samples

    were centrifuged for 3 min at 3000g, and the pellet was

    discarded. Plasma (200 l) was mixed with acetonitrile (800

    l), and the mixture was vortexed for 30 s, followed by

    centrifugation for 3 min at 3000g. The supernatant (500 l)

    was analysed on HPLC. The brain was transferred to a tube,

    and acetonitrile (2 ml) was added. The mixture was cooled

    on ice, homogenized for 30 s using a mixer (Ultra Turrax

    T18 basic, IKA Works, Setting 5) and finally centrifuged at

    3000gfor 3 min. The supernatant (500 l) was analysed on

    HPLC. An Alltech Econosil C18 column (25010 mm; 10

    m particle size) was used. The mobile phase was a 60/40

    mixture of ethanol and buffer (0.02 M phosphate buffer pH

    7.4) at a flow rate of 5 ml/min. A fraction collector was

    placed at the end of the HPLC column, and the eluate was

    collected in fractions of 5 ml (1 min). The fractions were then

    counted for radioactivity with an automated gamma counter.

    Extraction efficiency was always N90%.

    3. Results and discussion

    3.1. Regional brain biodistribution study in rodents

    The results of the regional brain biodistribution study

    with [123

    I]-3-I-CO in Sprague

    Dawley rats are shown inFig. 2. Brain radioactivity concentration values (% ID/g

    tissue) for all brain regions are indicated in Table 1 as

    meanS.D. values (n=3 animals per time point).

    These results demonstrate that [123I]-3-I-CO readily

    enters the brain. Brain radioactivity concentration was

    about 0.25 % ID/g tissue for the whole brain at 1 hour post

    injection. A maximum uptake in the target region (frontal

    cortex) of 0.6740.074% ID/g tissue at 20 min post injection

    was obtained. Uptake in the reference region (cerebellum)

    was considerably lower (a maximum value of 0.380.072 %

    ID/g tissue was obtained at 20 min post injection).

    Initial brain uptake of [123I]-3-I-CO was highest in the

    occipital cortex (0.9420.034% ID/g tissue at 20 min post

    injection) and frontal cortex (0.6740.074% ID/g tissue at

    20 min post injection). Uptake of [123I]-3-I-CO activity in

    the blood was consistently low (maximum value was

    0.0620.014% ID/g tissue at 20 min post injection). At 1 h

    after tracer injection, brain uptake of [123I]-3-I-CO was the

    Fig. 2. Regional brain biodistribution study with [123I]-3-I-CO in Sprague

    Dawley rats. Results are expressed as % ID/g tissue (meanS.D., n=3 per

    time point).

    863P.B.M. Blanckaert et al. / Nuclear Medicine and Biology 35 (2008) 861867

  • 8/7/2019 Blanckaert 2008

    4/7

    highest in the frontal cortex (0.3490.131 % ID/g tissue at

    1 h post injection).

    Fig. 2 demonstrates that radioactivity concentration results

    for the frontal cortex were well above the radioactivity

    concentrations in cerebellum up to 2 h after injection.

    Compared to other brain tracers, [123I]-3-I-CO showed a quite

    rapid washout out of the brain; the other brain regions showedroughly the same radioactivity clearance pattern (Table 1).

    The highest cortex-to-cerebellum ratio was achieved in

    the occipital cortex: a maximum ratio of 2.48 was obtained at

    20 min post injection. Starting at 1 h after radioligand

    injection, the ratio occipital cortex-to-cerebellum stabilised,

    varying between 1.3 and 1.2. A maximal frontal cortex-to-

    cerebellum ratio of 1.77 was reached at 20 min post injection,

    decreasing to 1.66 at 1 h after radioligand injection. The ratio

    stabilised around 1.1 at the later time points.

    The data obtained in the [123I]-3-I-CO rat biodistribution

    study were compared with the biodistribution data of a

    clinically used 5-HT2A tracer, [123I]-R91150. Although brain

    uptake of [123I]-3-I-CO in frontal cortex was high compared

    to [123I]-R91150 (0.3490.131% ID/g and 0.240.01% ID/g,

    respectively, at 1 h after injection), [123I]-3-I-CO also

    showed considerably more aspecific binding to cerebellum

    (0.2110.086% ID/g versus 0.040.002% ID/g for [123I]-

    R91150 at 1 h after injection). Moreover, a maximal frontal

    cortex-to-cerebellum ratio of only 1.7 was obtained through-

    out the study, whereas the biodistribution study with [123I]-

    R91150 revealed ratios varying between 7 and 18.

    From these data, it is clear that [123I]-3-I-CO showed

    considerable aspecific binding to brain tissue, possibly

    limiting its potential as a 5-HT2A brain tracer. Also, the level

    of specific binding of the radioligand was limited comparedto the aspecific binding of [123I]-3-I-CO in cerebellum.

    3.2. Ketanserin displacement study

    The influence of displacement with ketanserin on the

    regional brain biodistribution of [123I]-3-I-CO in Sprague

    Dawley rats can be seen in Fig. 3. [123I]-3-I-CO radioactivity

    was displaced by ketanserin in the cortical areas of the brain,

    decreasing the radioactivity concentration to the levels

    observed in cerebellum. In frontal cortex, radioactivity

    concentration decreased from 0.3490.131 % ID/g tissue at

    1 h post injection to 0.170.036% ID/g tissue after

    displacement with ketanserin. [123I]-3-I-CO activity also

    decreased in other parts of the cortex (temporal, occipital and

    parietal cortex), although the relative decrease in radio-

    activity concentration was the highest in frontal cortex

    (50% decrease after displacement with ketanserin).

    No significant difference in radioactivity concentration

    was found after ketanserin displacement in blood andcerebellum (cerebellum: 0.2110.086% ID/g tissue at 1 h

    post injection for the normal group and 0.1840.02% ID/g

    tissue after ketanserin displacement; blood: 0.0450.018%

    ID/g tissue for the normal and 0.0420.008% ID/g tissue for

    the ketanserin-treated group). This is in accordance with

    using the cerebellum as a reference region when performing

    PET or SPECT brain scans with 5-HT2A tracers. The results

    of the displacement study of [123I]-3-I-CO with ketanserin

    tartrate indicate that the radioligand binds specifically to the

    central 5-HT2A receptor in vivo in SpragueDawley rats.

    3.3. Influence of Pgp modulation

    The influence of cyclosporin A pretreatment (50 mg/kg

    intravenously, administered 30 min before injection of [123I]-

    3-I-CO) on the regional brain biodistribution results with

    [123I]-3-I-CO in SpragueDawley rats was investigated and

    Table 1

    Regional brain biodistribution with [123I]-3-I-CO in Sprague-Dawley rats

    Time (min)

    20 40 60 120 240

    Blood 0.0620.01 0.0590.001 0.0450.02 0.0430.001 0.0260.001

    Subcortical area 0.430.07 0.3430.08 0.280.13 0.0760.02 0.0420.002

    Temporal cortex 0.500.04 0.370.07 0.300.1 0.0780.03 0.0460.002

    Frontal cortex 0.670.07 0.430.08 0.350.1 0.0850.04 0.0520.004

    Occipital cortex 0.940.03 0.660.2 0.280.09 0.0980.08 0.0740.009

    Parietal cortex 0.380.03 0.310.02 0.230.01 0.0510.007 0.0270.001

    Cerebellum 0.380.07 0.300.06 0.210.09 0.0810.02 0.0460.002

    Results are expressed as % ID/g tissue (meanS.D., n=3 per time point).

    Fig. 3. Ketanserin displacement study with [123I]-3-I-CO in Sprague

    Dawley rats. Ketanserin was administered 30 min after radioligand injection.

    Results are expressed as % ID/g tissue (meanS.D. values).

    864 P.B.M. Blanckaert et al. / Nuclear Medicine and Biology 35 (2008) 861867

  • 8/7/2019 Blanckaert 2008

    5/7

    can be seen in Fig. 4 (for reasons of clarity, only frontalcortex and cerebellum are shown). Brain radioactivity

    concentration values are shown in Table 2.

    On average, radioligand uptake doubled in all brain areas

    after treatment of the animals with cyclosporin A. A

    maximum radioactivity uptake of 0.9050.114% ID/g tissue

    was obtained in frontal cortex at 40 min post injection. At

    1 h after injection, radioactivity concentration in frontal

    cortex was 0.5820.084% ID/g tissue. The radioactivity

    concentration in cerebellum was 0.3550.094% ID/g tissue

    at 1 h after injection of [123I]-3-I-CO. Radioactivity

    concentration in blood remained low and was fairly

    constant over time (0.0370.010% ID/g tissue at 2 h afterinjection, 0.0340.003% ID/g tissue at 4 h after injection).

    It is clear from Fig. 4 that both frontal cortex and

    cerebellum demonstrated an increased uptake of [123I]-3-I-

    CO radioactivity after pre-treatment of the animals with

    cyclosporin A.

    For example, at 1 h after injection of [123I]-3-I-CO,

    radioactivity concentration in frontal cortex increased

    from 0.3490.131% ID/g tissue for the normal group

    to 0.5820.084% ID/g tissue after treatment with

    cyclosporin A.

    A similar pattern was observed in the cerebellum:

    radioactivity concentration increased from 0.2110.086%

    ID/g tissue at 1 h after injection for the normal group to

    0.3550.094% ID/g tissue at 1 h after injection for the

    cyclosporin A group. The relative increase in [123I]-3-I-CO

    radioactivity concentration was the same for the cerebellum

    and the frontal cortex (a 67% increase). Other brain regions

    followed more or less the same pattern. No significant effect

    of cyclosporin A administration on the cortex-to-cerebellum

    binding index ratios could be established.

    Also, no significant difference in radioactivity concentra-

    tion in blood could be found between the two treatment

    groups. This could indicate that the increase in brain [123I]-3-

    I-CO radioactivity concentration after treatment with

    cyclosporin A is not the result of an increased influx of

    tracer to the brain but, rather, the consequence of a decreased

    tracer efflux out of the brain as a result of the Pgp blocking

    effect of cyclosporin A.

    However, studies with Pgp modulation and other brain

    tracers revealed a much larger increase in brain radioactivityconcentration after cyclosporin A treatment (e.g., [123I]-

    R91150 radioactivity concentration throughout the brain

    increased five- to sixfold after treatment of the animals with

    cyclosporin A). This is in contrast with the [123I]-3-I-CO

    data, where only a twofold increase was observed.

    From the above results, it can be concluded that [123I]-

    3-I-CO is a substrate for efflux by Pgp but it is definitely a

    much weaker substrate than for example [123I]-R91150.

    3.4. Metabolite analysis in rodents

    The metabolism pattern of [123I]-3-I-CO in Sprague

    Dawley rats was roughly the same as the metabolism

    pattern obtained in NMRI mice [16]. In blood, only two

    radiolabelled components were present: one component

    (90%) had the same HPLC retention time as authentic

    [123I]-3-I-CO, and the other component (510%) was

    identified as free radioiodide (data not shown). The same

    pattern was observed in the brain, only with lower

    concentrations of free radioiodide (35%). No lipophilic

    radiolabelled metabolites were found that could possibly

    interfere with later [123I]-3-I-CO imaging studies. The small

    Fig. 4. Regional brain biodistribution study with [123I]-3-I-CO (with and

    without cyclosporin A pretreatment). MeanS.D. values are shown (n=3 per

    time point).

    Table 2

    Regional brain biodistribution with [123I]-3-I-CO in SpragueDawley rats after treatment with cyclosporin A

    Time (min)

    20 40 60 120 240

    Blood 0.070.01 0.0470.01 0.0560.01 0.040.01 0.0340.003

    Subcortical area 0.650.1 0.670.06 0.350.08 0.180.1 0.0750.01

    Temporal cortex 0.560.26 0.810.23 0.320.06 0.140.09 0.0630.003

    Frontal cortex 0.610.17 0.910.1 0.580.08 0.220.09 0.070.005

    Parietal cortex 0.910.26 0.850.18 0.580.08 0.180.07 0.0710.01

    Occipital cortex 0.610.16 0.720.12 0.360.06 0.170.07 0.0550.009

    Cerebellum 0.630.11 0.650.09 0.350.09 0.160.06 0.0720.008

    The dosage of cyclosporin A was 50 mg/kg. Results are expressed as % ID/g tissue (meanS.D., n=3 per time point).

    865P.B.M. Blanckaert et al. / Nuclear Medicine and Biology 35 (2008) 861867

  • 8/7/2019 Blanckaert 2008

    6/7

    amount of free radioiodide found in blood should have no

    negative effect.

    4. Conclusion

    The regional brain biodistribution study in Sprague

    Dawley rats with [123I]-3-I-CO demonstrated that the

    radioligand readily crossed the bloodbrain barrier, resulting

    in concentration of the radioligand in areas of the brain

    containing high concentrations of 5-HT2A receptors. Highest

    brain radioactivity concentrations were found in the 5-HT2A-

    rich areas of the brain (cortex, mostly frontal cortex and

    occipital cortex). A maximum radioactivity concentration of

    0.6740.074% ID/g tissue was found in frontal cortex at 20

    min post injection. Lowest radioactivity concentrations were

    found in areas of the brain containing low concentrations of

    5-HT2A receptors (cerebellum). Uptake of [123I]-3-I-CO

    radioactivity in the blood was consistently low (maximum

    value was 0.0620.014% ID/g tissue at 20 min postinjection). A maximal frontal cortex-to-cerebellum binding

    index ratio of 1.77 was obtained at 20 min post injection.

    Starting 1 h after injection, the cortex-to-cerebellum binding

    index ratio stabilised around 1.2. This value is very low

    compared to other serotonin 5-HT2A brain SPECT tracers (in

    studies with [123I]-R91150, the binding index ratio varied

    between 7 and 18).

    This low binding index ratio was caused by the

    considerable amount of aspecific binding of [123I]-3-I-CO

    to brain tissues, limiting the specific signal of the

    radioligand. No radiolabelled metabolites were detected in

    blood or brain.

    Specific binding of the radioligand to the 5-HT2A receptor

    was demonstrated in the displacement study with ketanserin.

    [123I]-3-I-CO radioactivity was displaced in the 5-HT2A rich

    areas in the brain (cortical areas), whereas no displacement

    was observed in the brain areas lacking 5-HT2A receptors

    (cerebellum). No significant difference was observed in

    blood radioactivity concentrations after ketanserin treatment.

    The biodistribution study with [123I]-3-I-CO after cyclos-

    porin A treatment of SpragueDawley rats demonstrated that

    the radioligand is a substrate for Pgp in vivo in rodents.

    Blocking of Pgp function with cyclosporin A doubled the

    [123I]-3-I-CO radioactivity concentration in brain. However,

    other authors reported a five- to sixfold increase in brainradioactivity concentration after cyclosporin A treatment

    [18,23]. Contrary to these results, the increase in brain [123I]-

    3-I-CO uptake after cyclosporin A treatment was rather

    limited, thus leading us to conclude that [123I]-3-I-CO is only

    a weak substrate for Pgp efflux in vivo.

    The potential of [123I]-3-I-CO as a possible radiotracer for

    the serotonin 5-HT2A receptor is probably limited by a fairly

    rapid washout of radioactivity out of the brain. The

    radioligand demonstrated considerable aspecific binding to

    brain tissues, resulting in cortex-to-cerebellum binding index

    ratios that are probably too low for successful in vivo

    imaging. We can also conclude that [123I]-3-I-CO is most

    probably a weak substrate for the Pgp efflux transporter.

    References

    [1] Barnes NM, Sharp T. A review of central 5-HT receptors and their

    function. Neuropharmacology 1999;38:1083152.[2] Audenaert K, Van Laere K, Dumont F, Slegers G, et al. Decreased

    frontal serotonin 5-HT2a receptor binding index in deliberate self-harm

    patients. Eur J Nucl Med 2001;28:17582.

    [3] Audenaert K, Van Laere K, Dumont F, Vervaet M, et al. Decreased 5-

    HT2a receptor binding in patients with anorexia nervosa. J Nucl Med

    2003;44:1639.

    [4] Hashimoto T, Kitamura N, Kajimoto Y, Shirai Y, et al. Differential

    changes in serotonin 5-Ht(1A) and 5-Ht(2) receptor-binding in patients

    with chronic-schizophrenia. Psychopharmacology 1993;112:S359.

    [5] Meyer JH, Kapur S, Houle S, DaSilva J, et al. Prefrontal cortex 5-HT2

    receptors in depression: an [F-18]setoperone PET imaging study. Am J

    Psychiatry 1999;156:102934.

    [6] Mintun MA, Sheline YI, Moerlein SM, Vlassenko AG, et al.

    Decreased hippocampal 5-HT2A receptor binding in major depressive

    disorder: in vivo measurement with [18F]altanserin positron emissiontomography. Biol Psychiatry 2004;55:21724.

    [7] Peremans K, Audenaert K, Blanckaert P, Jacobs F, et al. Effects of

    aging on brain perfusion and serotonin-2A receptor binding in the

    normal canine brain measured with single photon emission tomo-

    graphy. Prog Neuropsychopharmacol Biol Psychiatry 2002;26:

    1393404.

    [8] Peremans K, Audenaert K, Coopman F, Blanckaert P, et al. Estimates

    of regional cerebral blood flow and 5-HT2A receptor density in

    impulsive, aggressive dogs with Tc-99m-ECD and I-123-5-I-R91150.

    Eur J Nucl Med Mol Imaging 2003;30:153846.

    [9] Peremans K, Audenaert K, Hoybergs Y, Otte A, et al. The effect of

    citalopram hydrobromide on 5-HT2A receptors in the impulsive-

    aggressive dog, as measured with I-123-5-I-R91150 SPECT. Eur J

    Nucl Med Mol Imaging 2005;32:70816.

    [10] Halldin C, Lundkvist C, Ginovart N, Nyberg S, et al. [C-11]MDL

    100907, a radioligand for selective imaging of 5-HT2A receptors with

    PET. J Nucl Med 1996;37:424.

    [11] Marner L, Knudsen GM, Madsen K, Haugbol S, et al. Longitudinal

    assessment of cerebral 5-HT2A receptors in normal volunteers. An

    [18F]-altanserin PET study. Neuroimage 2006;31:T101.

    [12] Ngan ETC, Yatham LN, Ruth TJ, Liddle PF. Decreased serotonin 2A

    receptor densities in neuroleptic-naive patients with schizophrenia: a

    PETstudy using [F-18]setoperone. Am J Psychiatry 2000;157:10168.

    [13] Catafau AM, Danus M, Bullich S, Llop J, et al. Characterization of the

    SPECT 5-HT2A receptor ligand I-123-R91150 in healthy volunteers:

    Part 1 pseudoequilibrium interval and quantification methods.

    J Nucl Med 2006;47:91928.

    [14] Catafau AM, Danus M, Bullich S, Nucci G, et al. Characterization of

    the SPECT 5-HT2A receptor ligand I-123-R91150 in healthy

    volunteers: Part 2 ketanserin displacement. J Nucl Med 2006;47:

    92937.

    [15] Fu X, Tan PZ, Kula NS, Baldessarini R, et al. Synthesis, receptor

    potency, and selectivity of halogenated diphenylpiperidines as

    serotonin 5-HT2A ligands for PET or SPECT brain imaging. J Med

    Chem 2002;45:231924.

    [16] Blanckaert P, Burvenich I, Devos F, Slegers G. Synthesis and in vivo

    evaluation in mice of (I-123)-(4-fluorophenyl)(1-(3-iodophenethyl)

    piperidin-4-yl) methanone as a potential SPECT-tracer for the

    serotonin 5-HT2A receptor. J Label Compounds Radiopharm 2007;

    50:1838.

    [17] Hendrikse NH, Franssen EJ, Van der Graaf WTA, Vaalburg W, et al.

    Visualization of multidrug resistance in vivo. Eur J Nucl Med 1999;26:

    28393.

    866 P.B.M. Blanckaert et al. / Nuclear Medicine and Biology 35 (2008) 861867

  • 8/7/2019 Blanckaert 2008

    7/7

    [18] Elsinga PH, Hendrikse NH, Bart J, van Waarde A, et al. Positron

    emission tomography studies on binding of central nervous system

    drugs and P-glycoprotein function in the rodent brain. Mol Imaging

    Biol 2005;7:3744.

    [19] Ishiwata K, Kawamura K, Yanai K, Hendrikse NH. In vivo evaluation

    of P-glycoprotein modulation of 8 PET radioligands used clinically.

    J Nucl Med 2007;48:817.

    [20] Kiyono Y, Yamashita T, Doi H, Kuge Y, et al. Is MIBG a

    substrate of P-glycoprotein? Eur J Nucl Med Mol Imaging 2007;34:44852.

    [21] Marian T, Szabo G, Goda K, Nagy H, et al. In vivo and in vitro

    multitracer analyses of P-glycoprotein expression-related multidrug

    resistance. Eur J Nucl Med Mol Imaging 2003;30:114754.

    [22] de Vries EFJ, Kortekaas R, van Waarde A, Dijkstra D, et al. Synthesis

    and evaluation of dopamine D-3 receptor antagonist C-11-GR218231

    as PET tracer for P-glycoprotein. J Nucl Med 2005;46:138492.

    [23] Liow JS, Lu SY, McCarron JA, Hong JS, et al. Effect of a

    P-glycoprotein inhibitor, cyclosporin A, on the disposition in rodent

    brain and blood of the 5-HT1A receptor radioligand, [C-11](R)-(

    )-RWAY. Synapse 2007;61:96105.

    867P.B.M. Blanckaert et al. / Nuclear Medicine and Biology 35 (2008) 861867