in vivo quantitative autoradiographic analysis of brain

32
JPET#136390 In Vivo Quantitative Autoradiographic Analysis of Brain Muscarinic Receptor Occupancy by Antimuscarinic Agents for Overactive Bladder Treatment Shuji Maruyama, Hideo Tsukada, Shingo Nishiyama, Takeharu Kakiuchi, Dai Fukumoto, Naoto Oku and Shizuo Yamada Department of Pharmacokinetics and Pharmacodynamics, Medical Biochemistry and Global Center of Excellence (COE) Program, School of Pharmaceutical Sciences, University of Shizuoka (SM, NO, SY), Shizuoka; Central Research Laboratory., Hamamatsu Photonics K. K. (HT, SN, TK, DF), Hamamatsu, Shizuoka, Japan JPET Fast Forward. Published on March 7, 2008 as DOI:10.1124/jpet.108.136390 Copyright 2008 by the American Society for Pharmacology and Experimental Therapeutics. This article has not been copyedited and formatted. The final version may differ from this version. JPET Fast Forward. Published on March 7, 2008 as DOI: 10.1124/jpet.108.136390 at ASPET Journals on December 3, 2021 jpet.aspetjournals.org Downloaded from

Upload: others

Post on 04-Dec-2021

2 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: In Vivo Quantitative Autoradiographic Analysis of Brain

JPET#136390

1

In Vivo Quantitative Autoradiographic Analysis of Brain Muscarinic

Receptor Occupancy by Antimuscarinic Agents for Overactive Bladder

Treatment

Shuji Maruyama, Hideo Tsukada, Shingo Nishiyama, Takeharu Kakiuchi, Dai Fukumoto, Naoto

Oku and Shizuo Yamada

Department of Pharmacokinetics and Pharmacodynamics, Medical Biochemistry and

Global Center of Excellence (COE) Program, School of Pharmaceutical Sciences,

University of Shizuoka (SM, NO, SY), Shizuoka; Central Research Laboratory.,

Hamamatsu Photonics K. K. (HT, SN, TK, DF), Hamamatsu, Shizuoka, Japan

JPET Fast Forward. Published on March 7, 2008 as DOI:10.1124/jpet.108.136390

Copyright 2008 by the American Society for Pharmacology and Experimental Therapeutics.

This article has not been copyedited and formatted. The final version may differ from this version.JPET Fast Forward. Published on March 7, 2008 as DOI: 10.1124/jpet.108.136390

at ASPE

T Journals on D

ecember 3, 2021

jpet.aspetjournals.orgD

ownloaded from

Page 2: In Vivo Quantitative Autoradiographic Analysis of Brain

JPET#136390

2

A running title: brain muscarinic receptor occupancy by antimuscarinic agents

Correspondence to : Shizuo Yamada, Ph.D., Department of Pharmacokinetics and

Pharmacodynamics and Global Center of Excellence (COE) Program, School of

pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka

422-8526, Japan. (Tel: +81-54-264-5631, Fax: +81-54-264-5635. E-mail:

[email protected] )

Number of text page: 19 pages (including Abstract, Introduction, Materials and Methods, Results,

Discussion, References)

Number of Tables: 3

Number of Figures: 5

Number of References: 39

Number of words in Abstract: 250 words

Number of words in Introduction: 535 words

Number of words in Discussion: 1470 words

ABBREVIATIONS : OAB, overactive bladder; [11C](+)3-MPB,

(+)N-[11C]methyl-3-piperidyl benzilate; CNS, central nervous system; REM, rapid eye

movement; RO, receptor occupancy; qEEG, quantitative electrocephalography; BBB,

blood-brain barrier; QNB, quinuclidinyl benzilate

This article has not been copyedited and formatted. The final version may differ from this version.JPET Fast Forward. Published on March 7, 2008 as DOI: 10.1124/jpet.108.136390

at ASPE

T Journals on D

ecember 3, 2021

jpet.aspetjournals.orgD

ownloaded from

Page 3: In Vivo Quantitative Autoradiographic Analysis of Brain

JPET#136390

3

ABSTRACT

We evaluated the effects of five clinically used antimuscarinic agents for overactive bladder

(OAB) treatment on in vivo muscarinic receptor binding in rat brain by quantitative

autoradiography. There was a dose-related decrease in in vivo specific

(+)N-[11C]methyl-3-piperidyl benzilate ([11C](+)3-MPB) binding in each brain region of rats

10 min after i.v. injection of oxybutynin, propiverine, solifenacin and tolterodine. Rank order

of the i.v. dose (RO50) for 50% receptor occupancy of antimuscarinic agents in rat brain

regions was propiverine > solifenacin > tolterodine, oxybutynin. There was a good linear

relationship between in vivo (pRO50 values in the rat hippocampus) and in vitro (pKi values in

human M1 receptors) receptor binding activities of propiverine, solifenacin and tolterodine.

The observed RO50 value of oxybutynin was about five times smaller than the predicted in

vitro Ki value. The dose ratios of antimuscarinic agents for the brain receptor occupancy

(RO50) to the inhibition of carbachol- and volume-induced increase in intravesical pressure

(ID50), which reflects in vivo selectivity for the urinary bladder over the brain, were greater for

solifenacin, tolterodine and propiverine than oxybutynin. Darifenacin displayed only slight

decrease in specific [11C](+)3-MPB binding in the rat brain regions and it was not dose-related.

In conclusion, in vivo quantitative autoradiographic analysis of brain muscarinic receptor

occupancy may provide fundamental basis for managing CNS side effects in antimuscarinic

therapy for OAB. It is suggested that in the treatment of OAB, CNS side effects can be

avoided by antimuscarinic agents with high selectivity for the urinary bladder over the brain.

This article has not been copyedited and formatted. The final version may differ from this version.JPET Fast Forward. Published on March 7, 2008 as DOI: 10.1124/jpet.108.136390

at ASPE

T Journals on D

ecember 3, 2021

jpet.aspetjournals.orgD

ownloaded from

Page 4: In Vivo Quantitative Autoradiographic Analysis of Brain

JPET#136390

4

Introduction

Antimuscarinic agents are widely used as the first line therapy for urgency, frequency with

or without urge incontinence, all symptoms of the disorder termed overactive bladder (OAB).

Antimuscarinic treatment is frequently associated with side-effects that limit its clinical use

(Andersson et al., 2004). Because the muscarinic receptor mediates the excitatory and

inhibitory actions of acetylcholine in the central and peripheral nervous systems, various

systemic adverse effects may occur by the administration of antimuscarinic agents for OAB.

In fact, dry mouth occurs most commonly and thereby decreases the quality of life of patients.

Therefore numerous studies involving anitmuscarinic agents to treat OAB have focused to the

selectivity for the urinary bladder over the salivary gland. The incidence of central nervous

system (CNS) side effects by antimuscarinic agents is generally lower than that of dry mouth,

but CNS side-effects may be of great concern in elderly patients because of an increase of

blood-brain barrier (BBB) permeability with aging (Pakulski et al., 2000; Ouslander, 2004). In

fact, short-term and chronic administration of oxybutynin in elderly subjects resulted in a

non-degenerative mild cognitive dysfunction (Katz et al., 1998; Ancelin et al., 2006).

Moreover, clinical studies have demonstrated the increased cognitive sensitivity to

scopolamine (Flicker et al., 1992 ; Molchan et al., 1992) and a reduced density of brain

muscarinic receptors in the elderly (Norbury et al., 2004).

Tolterodine seems to be less associated with cognitive impairment than oxybutynin as it is

less lipophilic and, therefore, less likely to cross BBB (Chapple, 2000; Clemett and Jarvis,

2001). However, Diefenbach et al. (2005) showed that oxybutynin and tolterodine

administration resulted in statistically significant reductions in rapid eye movement (REM)

sleep and slight increase in REM latency. Newly developed antimuscarinic agents such as

solifenacin and darifenacin compared with oxybutynin may exert relatively fewer CNS effects

This article has not been copyedited and formatted. The final version may differ from this version.JPET Fast Forward. Published on March 7, 2008 as DOI: 10.1124/jpet.108.136390

at ASPE

T Journals on D

ecember 3, 2021

jpet.aspetjournals.orgD

ownloaded from

Page 5: In Vivo Quantitative Autoradiographic Analysis of Brain

JPET#136390

5

than oxybutynin. Darifenacin did not affect cognitive function in elderly subjects (Lipton et al.,

2005; Kay et al., 2005, 2006). Solifenacin compared with oxybutynin has shown less effect on

the acquisition and consolidation of memory by passive avoidance test in rats (Suzuki et al.,

2007). To our knowledge, the CNS effect of propiverine was little reported previously.

Antimuscarinic agents are considered to exert CNS side effects by binding to brain

muscarinic receptors. Oki et al. (2007) have recently shown that oral administration of

oxybutynin but not tolterodine and darifenacin bound significantly to muscarinic receptors in

the mouse brain. However, these authors examined the effects of only single and low dose of

each agent on brain muscarinic receptor binding. Oxybutynin, propiverine and tolterodine form

active metabolites after oral administration and thus, these metabolites may contribute to the

efficacy and tolerability in vivo (Michel and Hegde, 2006). Because metabolites are generally

more hydrophilic than the parent agents, however, the CNS pharmacological effect may be mainly

due to the parent agents. In fact, the brain concentration of active metabolite (desethyloxybutynin)

after oral oxybutynin(0.25-2.54 µmol/kg) was less than 1% when compared with that of the parent

compound (Maruyama et al., unpublished observation). To clarify the risk of CNS side-effects in

OAB treatment by antimuscarinic agents, therefore, we have quantitatively compared brain

muscarinic receptor binding in rats after the i.v. injection of oxybutynin, propiverine, solifenacin,

tolterodine and darifenacin (Fig. 1A) at four different doses.

This article has not been copyedited and formatted. The final version may differ from this version.JPET Fast Forward. Published on March 7, 2008 as DOI: 10.1124/jpet.108.136390

at ASPE

T Journals on D

ecember 3, 2021

jpet.aspetjournals.orgD

ownloaded from

Page 6: In Vivo Quantitative Autoradiographic Analysis of Brain

JPET#136390

6

Materials and Methods

Animals. Male Sprague-Dawley rats (270 - 320 g) at the age of 8 weeks were purchased

from Shizuoka Laboratory Co. Ltd. (Shizuoka, Japan). They were housed in the laboratory

with free access to food and water, and maintained on a 12-hr dark/light cycle in a room with

controlled temperature (24 ± 1°C) and humidity (55 ± 5%). This study was conducted in

accordance with the guide for care and use of laboratory animals as adopted by the US

National Institutes of Health.

Materials. (+)N-[11C]methyl-3-piperidyl benzilate ([11C](+)3-MPB) (Fig. 1B) was

synthesized by N-methylation of nor-compound (+)3-piperidyl benzilate with [11C]methyl

iodide (Takahashi et al., 1999: Tsukada et al., 2001). Oxybutynin hydrochloride was

purchased from Sigma Aldrich (St. Louis, MO, USA). Propiverine hydrochloride, solifenacin

succinate, tolterodine L-tartrate and darifenacin hydrobromide were donated from Taiho

Pharmaceutical Co., Ltd. (Tokushima, Japan), Astellas Pharma Inc. (Tokyo, Japan),

Pharmacia Co. Ltd. (Tokyo, Japan) and Pfizer Co. Ltd. (Tokyo, Japan), respectively. All other

chemicals were purchased from commercial sources. Darifenacin was dissolved in

dimethylsulfoxide and diluted in physiological saline. [11C](+)3-MPB and the other compound

were dissolved and diluted in physiological saline. The injection volumes of [11C](+)3-MPB

and antimuscarinic agents were 0.5 and 0.25 mL, respectively.

Animal treatment and autoradiography. Rats intraperitoneally anesthetized with chloral

hydrate (400 mg/kg) received an i.v. injection of saline, oxybutynin (0.08 - 2.54 µmol/kg),

propiverine (0.74 - 24.8 µmol/kg), solifenacin (0.62 - 20.8 µmol/kg), tolterodine (0.21 - 6.31

µmol/kg) or darifenacin (0.20 - 5.91 µmol/kg). At 10 min after the injection, [11C](+)3-MPB

This article has not been copyedited and formatted. The final version may differ from this version.JPET Fast Forward. Published on March 7, 2008 as DOI: 10.1124/jpet.108.136390

at ASPE

T Journals on D

ecember 3, 2021

jpet.aspetjournals.orgD

ownloaded from

Page 7: In Vivo Quantitative Autoradiographic Analysis of Brain

JPET#136390

7

(150 MBq) was intravenously injected. At 30 min after the injection of [11C](+)3-MPB, rats

were sacrificed and the brain tissues were rapidly removed for the autoradiography. The brains

were cut into 2 mm thick coronal sections using a brain matrix (Muromachi Co. Ltd., Kyoto,

Japan) at room temperature. The sections were placed on an imaging plate for 10 min, and the

plate was scanned with a bio-imaging analyzer of type BAS 1500 (FUJIX Fuji Photo Film

Co.Ltd., Tokyo, Japan). Regions of interest were placed on the cerebral cortex, corpus striatum,

hippocampus, amygdala, thalamus, hypothalamus, pons and cerebellum by using a Macintosh

computer (Image Reader version 1.2, Fuji film). The value of pmol/g wet tissue was calculated

from the value of photo-stimulated luminescence unit/mm2. For the quantification, individual

calibration standards were prepared for each set of brain sections exposed to the same imaging

plate. The standard (10 µl) of [11C](+)3-MPB solution at the known concentration was placed

on an Advantec filter paper No.2 (Toyo Roshi Kaisha, Ltd., Tokyo, Japan) and exposed

simultaneously with the brain sections. From the radioactivity of standard sample, the amount

of substance expressed in Bq was calcuated. The total counts over the standard, measured by

the phosphor-imaging system, allowed the calculation of a calibration factor in counts per Bq.

The signal measured for region of interest in a structure of a brain slice was given as average

counts per pixel. From the pixel size, the value to counts/mm2 was calculated and, using the

calibration factor, it was converted to MBq/mm2 (and MBq/g tissue). Before the calculation,

the average counts per pixel of the background area close to the brain slices were subtracted

from the average counts of the region of interest. The value of pmol/g tissue was estimated

from specific activity of [11C](+)3-MPB. The data were corrected for decay.

Specific binding of [11C](+)3-MPB was defined as the difference of radioactivity between

each brain region and cerebellum because even high dose of atropine failed to lower in vivo

[3H]quinuclidinyl benzilate (QNB) binding in the rat cerebellum (Yamamura et al., 1974).

Receptor occupancy (RO) was calculated from the degree of reduction (%) by antimuscarinic

This article has not been copyedited and formatted. The final version may differ from this version.JPET Fast Forward. Published on March 7, 2008 as DOI: 10.1124/jpet.108.136390

at ASPE

T Journals on D

ecember 3, 2021

jpet.aspetjournals.orgD

ownloaded from

Page 8: In Vivo Quantitative Autoradiographic Analysis of Brain

JPET#136390

8

agents in in vivo specific [11C](+)3-MPB binding in each brain region.

Data analysis. The dose-receptor occupancy curves were fitted to following hyperbolic

function with an assumption of single binding site by GraphPad Prism 4 (GraphPad Software

Inc., San Diego, CA). RO values were calculated by the following equation (1):

………………..(1)

where RO is the receptor occupancy (%), D is the dose (µmol/kg) and γ is the Hill

coefficient. RO50 is the dose at which a half-maximal receptor occupancy is obtained. The

graphs shown in the figures were drawn using the fitting constants.

Because the M1 subtype is the most abundant in hippocampus which plays important roles

in working and reference memory, the binding affinities (inhibition constant: Ki) of

antimuscarinic agents in human recombinant M1 receptor subtypes were used as an index of in

vitro affinities of these agents (Maruyama et al., 2006). A relationship between in vivo (RO50

values in rat hippocampus) and in vitro (Ki values in human M1 receptor) affinities of

antimuscarinic agents were examined by a linear regression analysis using GraphPad Prism 4

(GraphPad Software Inc., San Diego CA). The ratio for brain receptor occupancy (RO50) to the

pharmacological potency in the urinary bladder (ID50) of antimuscarinic agents was calculated.

The ID50 value, the dosage of antimuscarinic agent needed to inhibit 50% of carbachol- and

volume-induced increases in the intraurethral pressure in rats, was cited from previous

literatures (Ikeda et al., 2002; Modiri et al., 2002; Ohtake et al., 2004).

All data are expressed as the mean ± SEM or with 95% confidence interval (CI). The

statistically significant difference was analyzed by Williams’s test. Difference with p<0.05

were considered statistically significant.

[D]γ

[D]γ + [RO50]γ

× 100 RO (%) =

This article has not been copyedited and formatted. The final version may differ from this version.JPET Fast Forward. Published on March 7, 2008 as DOI: 10.1124/jpet.108.136390

at ASPE

T Journals on D

ecember 3, 2021

jpet.aspetjournals.orgD

ownloaded from

Page 9: In Vivo Quantitative Autoradiographic Analysis of Brain

JPET#136390

9

Results

Fig. 2A illustrates typical autoradiographic images in the rat brain at 30 min after the i.v.

injection of [11C](+)3-MPB. The highest radioactivity of [11C](+)3-MPB was detected in the

corpus striatum and the lowest value was seen in the cerebellum. This agrees well with the

previous finding by Yamamura and Snyder (1974) who showed the rank of regional

distribution of muscarinic receptors in the rat brain, being corpus striatum > cerebral cortex >

hippocampus > midbrain and pons, thalamus, hypothalamus >> cerebellar cortex. Even high

dose of atropine failed to lower in vivo [3H]QNB binding in the rat cerebellum (Yamamura et

al., 1974).

The effects of i.v. injection of antimuscarinic agents were examined on specific

[11C](+)3-MPB binding in the cerebral cortex, corpus striatum, hippocampus, amygdala,

thalamus, hypothalamus and pons of rats. As shown in Fig. 2B, there was a dose-dependent

decrease in the radioactivity of [11C](+)3-MPB in the brain of rats 10 min after the i.v.

injection of oxybutynin (0.08 - 2.54 µmol/kg), propiverine (0.74 - 24.8 µmol/kg), solifenacin

(0.62 - 20.8 µmol/kg) and tolterodine (0.21 - 6.31 µmol/kg). In the case of darifenacin (0.20 -

5.91 µmol/kg), slight decrease in the radioactivity was also observed, but it was not the

dose-related. From the data in Fig. 2B, we estimated in vivo specific [11C](+)3-MPB binding in

each brain region after the i.v. injection of antimuscarinic agents (Fig. 3). Following the i.v.

injection of oxybutynin (0.08 - 2.54 µmol/kg), specific [11C](+)3-MPB binding was

significantly decreased in the hippocampus (20.3 - 74.3%), amygdala (33.8 - 76.5%), thalamus

(34.1 - 73.0%) and hypothalamus (33.9 - 75.4%) in a dose-dependent manner. The i.v.

injection of oxybutynin (0.25 - 2.54 µmol/kg) showed a significant decrease of specific

[11C](+)3-MPB in the cerebral cortex (41.4 - 75.8 %), corpus striatum (35.8 - 74.7%) and pons

(41.9 - 73.2%).

Similarly, i.v. injection of propiverine (7.43, 24.8 µmol/kg) showed significant and

This article has not been copyedited and formatted. The final version may differ from this version.JPET Fast Forward. Published on March 7, 2008 as DOI: 10.1124/jpet.108.136390

at ASPE

T Journals on D

ecember 3, 2021

jpet.aspetjournals.orgD

ownloaded from

Page 10: In Vivo Quantitative Autoradiographic Analysis of Brain

JPET#136390

10

dose-dependent decreases in specific [11C](+)3-MPB binding in the cerebral cortex (33.5%,

56.8%), hippocampus (26.7%, 49.6%), amygdala (34.2%, 54.6%), thalamus (40.5%, 65.2%),

hypothalamus (44.0%, 70.6%) and pons (39.7%, 59.7%). In the corpus striatum, significant

decreases of specific [11C](+)3-MPB binding by 24.8 µmol/kg propiverine was 40.2%. This

agent at 0.74 and 2.48 µmol/kg had little significant effect on the brain [11C](+)3-MPB

binding.

Solifenacin at i.v. doses of 2.08 to 20.8 µmol/kg induced significant decreases in specific

[11C](+)3-MPB binding in all brain regions compared with control values, and the decreases in

the cerebral cortex, corpus striatum, hippocampus, amygdala, thalamus, hypothalamus and

pons ranged from 30.6% to 78.3%. This effect at these doses seemed to be not necessarily the

dose-related. The lower dose (0.62 µmol/kg) of solifenacin had little significant effect on the

brain [11C](+)3-MPB binding.

Following i.v. injection of tolterodine (0.21 - 6.31 µmol/kg), specific [11C](+)3-MPB

binding was significantly decreased in the hippocampus (21.0 - 90.2%), amygdala (25.7% -

91.6%), thalamus (31.3 - 90.5%), hypothalamus (35.1 - 95.6%) and pons (43.8 - 90.6%) in a

dose-dependent manner. The i.v. injection of tolterodine (0.63 - 6.31 µmol/kg) showed a

significant decrease of specific [11C](+)3-MPB in the cerebral cortex (48.2 - 91.4%) and

corpus striatum (43.5 - 91.4%). In contrast, darifenacin (0.20 - 5.91µmol/kg) displayed little

significant change in specific [11C] (+) 3-MPB binding.

On the basis of the data in Fig. 3, we estimated the dose-receptor occupancy curves in each

brain region after i.v. injection of oxybutynin, proiverine, solifenacin and tolterodine (Fig. 4,

Table 1). As shown in Table 1, the rank order of average RO50 values of antimuscarinic agents

in the rat brain regions was propiverine (10.0 - 40.5 µmol/kg) > solifenacin (3.3 - 10.7

µmol/kg) > tolterodine (0.34 - 0.98 µmol/kg), oxybutynin (0.28 - 0.57 µmol/kg). In the case of

darifenacin, the RO50 value could not be estimated by the lack of dose-dependent inhibition of

This article has not been copyedited and formatted. The final version may differ from this version.JPET Fast Forward. Published on March 7, 2008 as DOI: 10.1124/jpet.108.136390

at ASPE

T Journals on D

ecember 3, 2021

jpet.aspetjournals.orgD

ownloaded from

Page 11: In Vivo Quantitative Autoradiographic Analysis of Brain

JPET#136390

11

specific [11C](+)3-MPB binding. The average RO50 values of oxybutynin seemed to be similar

among each brain region, while RO50 values of propiverine and tolterodine tended to be larger

in the corpus striatum and hippocampus than in the hypothalamus and pons.

Fig. 5 shows a relationship between in vivo (pRO50 values in the rat hippocampus) and in

vitro (pKi values in human M1 receptors, Maruyama et al., 2006) receptor binding affinity of

antimuscarinic agents. The linear regression analysis between propiverine, solifenacin and

tolterodine showed a good fitness (r2 = 0.9999) by the equation of pRO50 = 0.6946 ×pKi –

0.001054. By this equation, the in vivo affinity (pRO50 value) of these agents from in vitro

affinity (pKi value) could be predicted. Table 2 compares the observed and predicted values of

RO50 in the rat hippocampus of antimuscarinic agents with their ratios after the i.v injection.

The predicted values of propiverine, solifenacin and tolterodine corresponded nicely to the

observed values, but the predicted value of oxybutynin was about five times larger than the

observed value.

The selectivity for the urinary bladder over the brain region of antimuscarinic agents in rats

was evaluated by ratios for the brain receptor occupancy (RO50) to the pharmacological

potency in the urinary bladder (ID50). The pharmacological potency (ID50) of each agent in the

urinary bladder was cited from previous literatures of inhibitory effects of carbachol- and

volume-induced increases in the intravesical pressure of rats (Ikeda et al., 2002; Modiri et al.,

2002; Ohtake et al., 2004). The rank order of the ratio (RO50/ID50) in brain regions to the

urinary bladder was solifenacin (8.1 - 46.7), tolterodine (3.6 - 17.9), propiverine (2.2 - 8.9) >

oxybutynin (1.4 - 3.4) (Table 3). Thus, the selectivity for the urinary bladder over the

hippocampus was greater for solifenacin (11.2, 20.0), tolterodine (9.5, 16.3) and propiverine

(5.4) than oxybutynin (2.5 - 3.0). Similar tendency was observed also in other brain regions.

This article has not been copyedited and formatted. The final version may differ from this version.JPET Fast Forward. Published on March 7, 2008 as DOI: 10.1124/jpet.108.136390

at ASPE

T Journals on D

ecember 3, 2021

jpet.aspetjournals.orgD

ownloaded from

Page 12: In Vivo Quantitative Autoradiographic Analysis of Brain

JPET#136390

12

Discussion

The brain muscarinic receptor was measured by quantitative autoradiographic analysis in rats

after i.v. injection of [11C](+)3-MPB. [11C](+)3-MPB may be a superior radioligand for the in

vivo labeling of brain muscarinic receptors because of lower binding affinity and rapid

equilibrium in comparison with other raioligands such as [3H]QNB (Takahashi et al., 1999;

Tsukada et al., 2001). The autoradiographic technique to measure various receptors is well

established with long-lived radiotracers (e.g. 3H and 14C etc), but the major disadvantage is to

take several weeks as the exposure time of film for the data analysis due to the low-energy

emitting radionuclides. In contrast, the merit of use of high-energy β+- emitting radionuclides

such as 11C or 18F is to make the exposure time markedly shorter, being usually only several

minutes or hours. Moreover, the autoradiographic method using β+- emitting radionuclides

could apply to positron emission tomography. The in vivo competition binding assay by the

quantitative autoradiographic method is a powerful way to analyze directly muscarinic

receptor occupancy of antimuscarinic agents in the brain, making possible to quantify the

penetration of antimuscarinic agents into brain tissues.

To clarify the risk of CNS side-effects in OAB treatment, using the quantitative

autoradiographic method, we have comparatively characterized brain muscarinic receptor binding

after the i.v. injection of oxybutynin, propiverine, solifenacin, tolterodine and darifenacin. The i.v.

doses of antimuscarinic agents used in the present study were determined on the basis of

pharmacologically relevant doses to impair learning in rat passive avoidance response (Suzuki et

al., 2007). The i.v. injection of oxybutynin, propiverine, tolterodine and solifenacin

significantly decreased in vivo specific [11C](+)3-MPB binding in each brain region of rats in a

dose-dependent manner. According to the estimated RO50 values, the potency of muscarinic

receptor occupancy by each agent in the rat brain appeared to be greatest in oxybutynin,

This article has not been copyedited and formatted. The final version may differ from this version.JPET Fast Forward. Published on March 7, 2008 as DOI: 10.1124/jpet.108.136390

at ASPE

T Journals on D

ecember 3, 2021

jpet.aspetjournals.orgD

ownloaded from

Page 13: In Vivo Quantitative Autoradiographic Analysis of Brain

JPET#136390

13

followed by tolterodine, solifenacin and proiverine. Further, it was shown that in vivo

potencies (pRO50) of propiverine, solifenacin and tolterodine in occupying in vivo

hippocampal receptors after the i.v. injection correlated well with their in vitro binding

affinities in human M1 muscarinic receptor subtype (Maruyama et al., 2006) (Fig. 5). However,

the observed RO50 value of oxybutynin was about five times smaller than the value predicted

by the linear correlation between propiverine, solifenacin and tolterodine. These data suggest

that oxybutynin crosses more easily through BBB than other antimuscarinic agents examined.

Antimuscarinic agents must first pass the BBB to occupy central muscarinic receptors. The

observed difference among antimuscarinic agents in the potency of brain muscarinic receptor

occupancy may be defined by BBB permeability which is responsible for CNS effects in

patients. The passive penetration of antimuscarinic agents through this physiologic barrier

generally depends on physicochemical factors such as high lipophilicity, low degree of

ionization (neutral charge) and small molecular size (Sheife and Takeda, 2005). The

characteristics of chemical properties of oxybutynin relative to tolterodine - lipophilicity (Log

Ko/w: 4.68 vs 1.83) (Watanabe, 2007; Abrams, 2001) and neutral polarity (pKa: 6.44 vs 9.87)

(Yokoyama et al., 1996; Abrams, 2001) - make it the most likely to cross the BBB (Kay and

Granville, 2005). Påhlman et al. (2001) have revealed that the distribution of radioactivity in

the brain was the lowest among other tissues from the mice received oral [14C]tolterodine. This

may be consistent with the relatively lower in vivo binding activity of brain muscarinic

receptors by tolterodine compared with the pharmacological activity in the bladder. Also,

solifenacin, like tolterodine, may have molecular characteristics that make it unlikely to cross

the BBB.

RO50 values of antimuscarinic agents (Table 1) were similar to i.v. doses inhibiting

learning/memory behavior in rats (Suzuki et al., 2007). Thus, these values could be regarded

as the index of CNS pharmacological effects following the blockade of brain muscarinic

This article has not been copyedited and formatted. The final version may differ from this version.JPET Fast Forward. Published on March 7, 2008 as DOI: 10.1124/jpet.108.136390

at ASPE

T Journals on D

ecember 3, 2021

jpet.aspetjournals.orgD

ownloaded from

Page 14: In Vivo Quantitative Autoradiographic Analysis of Brain

JPET#136390

14

receptors. Subsequently, we evaluated the selectivity for the urinary bladder over the brain of

antimuscarinic agents by using pharmacological data in the rat urinary bladder (Ohtake et al.,

2004; Ikeda et al., 2002; Modiri et al., 2002). It was shown that antimuscarinic agents

inhibited significantly carbachol- and volume-induced increase in the intravesical pressure in

rats. The dose ratios (RO50/ID50) of antimuscarinic agents for the brain receptor occupancy to

the inhibitory potency of increases in intravesical pressure were considered to reflect in vivo

pharmacological selectivity for the urinary bladder over the brain. This ratio was relatively

large in solifenacin (8.1 - 46.7), tolterodine (3.6 - 17.9) and propiverine (2.2 - 8.9), compared

with oxybutynin (1.4 - 3.4) (Table 3). Thus, the selectivity for the urinary bladder over the

brain was relatively low for oxybutynin, suggesting a high feasibility of CNS side effects at

pharmacological doses to treat OAB. This finding seems to be in a reasonable agreement with

previous observations of oxybutynin shown by pharmacological (Sugiyama et al., 1999) and

ex vivo receptor binding (Oki et al., 2001) studies in rats, and also by clinical studies (Katz et

al., 1998; Pietzko et al., 1994; Todorova et al., 2001; Ancelin et al., 2006). In fact, CNS side

effect by oxybutynin occurred in patients with OAB and in older subjects receiving this drug

(Scheife and Takeda, 2005; Kay et al., 2006). It was shown that only higher dose (24.8

µmol/kg i.v.) of propiverine induced CNS effects in rats (Suzuki et al., 2007) and bound to

muscarinic receptors in rat brain (Oki et al., 2001). Thus, propiverine may exhibit lower

incidence of CNS effects than that of oxybutynin in patients with OAB.

The selectivity for the urinary bladder over the brain of solifenacin and tolterodine was

clearly higher than that of oxybutynin in rats. In clinical study, the incidence of CNS side

effect of tolterodine was shown to be lower than that of oxybutynin and comparable to that

with placebo (Chapple, 2000; Clemett and Jarvis, 2001; Scheife and Takeda, 2005). Thus,

these data suggest that solifenacin and tolterodine have advantages in the treatment of OAB

owing to lower CNS effect. Todorova et al. (2001) comparatively evaluated electrophysiologic

This article has not been copyedited and formatted. The final version may differ from this version.JPET Fast Forward. Published on March 7, 2008 as DOI: 10.1124/jpet.108.136390

at ASPE

T Journals on D

ecember 3, 2021

jpet.aspetjournals.orgD

ownloaded from

Page 15: In Vivo Quantitative Autoradiographic Analysis of Brain

JPET#136390

15

effects of antimuscarinic agents on the CNS in healthy male volunteers by using quantitative

electrocephalography (qEEG). They found that oxybutynin significantly altered qEEG activity,

while tolterodine and trospium induce only a slight effect on qEEG activity. Similar

electrophysiological results in healthy males with oxybutynin have been shown by Pietzko et

al. (1994).

Darifenacin at pharmacologically effective doses did not significantly reduce in vivo

specific [11C](+) 3-MPB binding so that the RO50 value could not be estimated. These data

suggest that darifenacin induces the lowest incidence of CNS effects among antimuscarinic

agents examined. This result agrees with our previous ex vivo findings (Oki et al., 2007) and

previous clinical observations (Lipton et al., 2005; Kay et al., 2005, 2006), showing that the

treatment with darifenacin may have little effect on the cognitive function in the elderly

subjects. Several lines of evidence suggest that darifenacin is unlikely to cross the BBB.

Darifenacin is a substrate for P-glycoprotein (Skerjanec, 2006), an active-transporter system

that carry back across the BBB.

Additionally, muscarinic receptor subtype selectivity of antimuscarinic agents may be

implicated in the appearance of CNS effect. All of five muscarinic receptor subtypes are

expressed in the brain (Flynn et al., 1997; Oki et al., 2005). The M1 receptor was abundant in

the cortex and hippocampus. In the striatum, the M1 and M4 receptors were distributed. By

contrast, the M2 receptor was predominantly localized in the brainstem and cerebellum. The

M3 receptor displays lower density in the brain when compared to M1, M2 and M4 receptor

expressions. The cognitive dysfunction by antimuscarinic agents may be mediated mainly by

the M1 and M2 receptors in the CNS (Kay et al., 2005). Oxybutynin shows selectivity for the

M1, M3 and M4 receptors, while tolterodine and propiverine are relatively nonselective to

muscarinic receptor subtypes (Maruyama et al., 2006; Ohtake et al., 2007). Solifenacin and

darifenacin show higher selectivity to the M3 receptor than M1, M2 and M4 receptors. Thus, M1

This article has not been copyedited and formatted. The final version may differ from this version.JPET Fast Forward. Published on March 7, 2008 as DOI: 10.1124/jpet.108.136390

at ASPE

T Journals on D

ecember 3, 2021

jpet.aspetjournals.orgD

ownloaded from

Page 16: In Vivo Quantitative Autoradiographic Analysis of Brain

JPET#136390

16

selectivity in addition to high BBB permeability of oxybutynin may be more apt to cause CNS

side effects. Such muscarinic receptor subtype selectivity may be partly associated with an

observed difference among antimuscarinic agents in the in vivo potency of brain receptor

occupancy (Fig. 4, Table 1). Taken together, the present study has basically confirmed ex vivo

binding data in mouse brain obtained by single oral dose of oxybutynin, tolterodine and

darifenacin (Oki et al., 2007), and greatly expanded their findings by using four different i.v.

doses of clinically used five antimuscarinic agents under in vivo condition.

In conclusion, analysis of brain muscarinic receptor occupancy in vivo may provide

fundamental basis for antimuscarinic agent-induced CNS side effects in OAB treatment. The

use in OAB patients would require careful dose adjustment to avoid the risk of side effects

associated with brain receptor binding. Relatively newer generation of antimuscarinic agents

which have high selectivity of the urinary bladder rather than the brain could be advantageous

to treat OAB.

This article has not been copyedited and formatted. The final version may differ from this version.JPET Fast Forward. Published on March 7, 2008 as DOI: 10.1124/jpet.108.136390

at ASPE

T Journals on D

ecember 3, 2021

jpet.aspetjournals.orgD

ownloaded from

Page 17: In Vivo Quantitative Autoradiographic Analysis of Brain

JPET#136390

17

References

Abrams P (2001) Evidence for the efficacy and safety of tolterodine in the treatment of

overactive bladder. Expert Opin Pharmacother 2:1685-1701.

Ancelin ML, Artero S, Portet F, Dupuy AM, Touchon J and Ritchie K (2006)

Non-degenerative mild cognitive impairment in elderly people and use of anticholinergic

drugs: longitudinal cohort study. BMJ 332:455-459.

Andersson KE (2004) Antimuscarinics for treatment of overactive bladder. Lancet Neurol

3:46-53.

Chapple CR (2000) Muscarinic receptor antagonists in the treatment of overactive bladder.

Urol 55(Suppl. 5A):33-46.

Clemett D and Jarvis B (2001) Tolterodine: a review of its use in the treatment of overactive

bladder. Drugs Aging 18:277-304.

Diefenbach K, Arold G, Wollny A, Schwantes U, Haselmann J and Roots I (2005) Effects on

sleep of anticholinergics used for overactive bladder treatment in healthy volunteers aged≥

50 years. BJU Int 95:346-349.

Flicker C, Ferris SH and Serby M (1992) Hypersensitivity to scopolamine in the elderly.

Psychopharmacol 107:437-441.

Flynn DD, Reever CM and Ferrari-DiLeo G (1997) Pharmacological strategies to selectivity

label and localize muscarinic receptor subtypes. Drug Dev Rev 40:104-116.

Ikeda K, Kobayashi S, Suzuki M, Miyata K, Takeuchi M, Yamada T and Honda K (2002) M3

receptor antagonism by the novel antimuscarinic agent solifenacin in the urinary bladder

and salivary gland. Naunyn-Schmiedeberg’s Arch Pharmacol 366:97-103.

Katz IR, Sands LP, Bilker W, DiFilippo S, Boyce A and D’Angelo K (1998) Identification of

medications that cause cognitive impairment in older people: the case of oxybutynin

This article has not been copyedited and formatted. The final version may differ from this version.JPET Fast Forward. Published on March 7, 2008 as DOI: 10.1124/jpet.108.136390

at ASPE

T Journals on D

ecember 3, 2021

jpet.aspetjournals.orgD

ownloaded from

Page 18: In Vivo Quantitative Autoradiographic Analysis of Brain

JPET#136390

18

chloride. J Am Geriatr Soc 46:8-13.

Kay GG and Granville LJ (2005) Antimuscarinic agents: Implications and concerns in the

management of overactive bladder in the elderly. Clin Ther 27:127-138.

Kay GG, Abou-Donia MB, Messer WS Jr, Murphy DG, Tsao JW and Ouslander JG (2005).

Antimuscarinic drugs for overactive bladder and their potential effects on cognitive function

in older patients. J Am Geriatr Soc 53:2195-2201.

Kay G, Crook T, Rekeda L, Lima R, Ebinger U, Arguinzoniz M and Steel M (2006).

Differential effects of the antimuscarinic agents darifenacin and oxybutynin ER on memory

in older subjects. Eur Urol 50:317-326.

Lipton RB, Kolodner K and Wesnes K (2005). Assessment of cognitive function of the elderly

population: effects of darifenacin. J Urol 173:493-498

Maruyama S, Oki T, Otsuka A, Shinbo H, Ozono S, Kageyama S, Mikami Y, Araki I, Takeda

M, Masuyama K and Yamada S (2006). Human muscarinic receptor binding characteristics

of antimuscarinic agents to treat overactive bladder. J Urol 175:365-369.

Michel MC and Hegde SS (2006) Treatment of the overactive bladder syndrome with

muscarinic receptor antagonists-a matter of metabolites? Naunyn-Shcmiedeberg’s Arch

Pharmacol 374:79-85.

Modiri AR, Alberts P and Gillberg PG (2002) Effect of muscarinic antagonists on micturition

pressure measured by cystometry in normal, conscious rats. Urol 59:963-968.

Molchan SE, Martinez RA, Hill JL, Weingartner HJ, Thompson K, Vitiello B and Sunderland

T (1992) Increased cognitive sensitivity to scopolamine with age and a perspective on the

scopolamine model. Brain Res Brain Res Rev 17:215-226.

Norbury R, Travis MJ, Erlandsson K, Waddington W, Owens J, Ell PJ and Murphy DG (2004)

SPET imaging of central muscarinic receptors with (R,R)[123I]-I-QNB: methodological

considerations. Nucl Med Biol 31:583-590.

This article has not been copyedited and formatted. The final version may differ from this version.JPET Fast Forward. Published on March 7, 2008 as DOI: 10.1124/jpet.108.136390

at ASPE

T Journals on D

ecember 3, 2021

jpet.aspetjournals.orgD

ownloaded from

Page 19: In Vivo Quantitative Autoradiographic Analysis of Brain

JPET#136390

19

Ohtake A, Ukai M, Hatanaka T, Kobayashi S, Ikeda K, Sato S, Miyata K and Sasamata M,

(2004). In vitro and in vivo tissue selectivity profile of solifenacin succinate (YM905) for

urinary bladder over salivary gland in rats. Eur J Pharmacol 492: 243-250.

Ohtake A, Saitoh C, Yuyama H, Ukai M, Okutsu H, Noguchi Y, Hatanaka T, Suzuki M, Sato

S, Sasamata M and Miyata K (2007) Pharmacological characterization of a new

antimuscarinic agent, solifenacin succinate, in comparison with other antimuscarinic agents.

Biol Pharm Bull 30:54-58.

Oki T, Kageyama A, Takagi Y, Uchida S and Yamada S (2007) Comparative evaluation of

central muscarinic receptor binding activity by oxybutynin, tolterodine and darifenacin used

to treat overactive bladder. J Urol 177:766-770.

Oki T, Takagi Y, Inagaki S, Taketo MM, Manabe T, Matsui M, and Yamada S (2005)

Quantitative analysis of binding parameters of [3H]N-methylscopolamine in central nervous

system of muscarinic acetylcholine receptor knockout mice. Brain Res Mol Brain Res 133:

6-11.

Oki T, Yamada S, Tohma A, and Kimura R (2001) Muscarinic receptor binding characteristics

in rat tissues after oral administration of oxybutynin and propiverine. Biol Pharm Bull 24:

491-495.

Ouslander JG (2004) Management of overactive bladder. N Engl J Med 350: 786-799.

Påhlman I, d'Argy R and Nilvebrant L (2001). Tissue distribution of tolterodine, a muscarinic

receptor antagonist, and transfer into fetus and milk in mice. Arzneimittelforschung 51:

125-133.

Pakulski C, Drobnik L and Millo B (2000) Age and sex as factors modifying the function of

the blood-cerebrospinal fluid barrier. Med Sci Monit 6:314-318.

Pietzko A, Dimpfel W, Schwantes U and Topfmeier P (1994) Influences of trospium chloride

and oxybutynin on quantitative EEG in healthy volunteers. Eur J Clin Pharmacol

This article has not been copyedited and formatted. The final version may differ from this version.JPET Fast Forward. Published on March 7, 2008 as DOI: 10.1124/jpet.108.136390

at ASPE

T Journals on D

ecember 3, 2021

jpet.aspetjournals.orgD

ownloaded from

Page 20: In Vivo Quantitative Autoradiographic Analysis of Brain

JPET#136390

20

47:337-343.

Scheife R and Takeda M (2005) Central nervous system safety of anticholinergic drugs for the

treatment of overactive bladder in the elderly. Clin Ther 27:144-153.

Skerjanec A (2006) The clinical pharmacokinetics of darifenacin. Clin Pharmacokinet

45:325-350.

Sugiyama T, Park YC and Kurita T (1999) Oxybutynin disrupts learning and memory in the

rat passive avoidance response. Urol Res 27: 393-395.

Suzuki M, Noguchi Y, Okutsu H, Ohtake A and Sasamata M (2007) Effect of antimuscarinic

drugs used for overactive bladder on learning in a rat passive avoidance response test. Eur J

Pharmacol 557:154-158.

Takahashi K, Murakami M, Miura S, Iida H, Kanno I and Uemura K (1999) Synthesis and

autoradiographic localization of muscarinic cholinergic antagonist

(+)N-[11C]methyl-3-piperidyl benzilate as a potent radioligand for positron emission

tomography. Appl Radiat Isot 50:521-525.

Todorova A, Vonderheid-Guth B and Dimpfel W (2001) Effects of tolterodine, trospium

chloride, and oxybutynin on the central nervous system. J Clin Pharmacol 41:636-644.

Tsukada H, Takahashi K, Miura S, Nishiyama S, Kakiuchi T, Ohba H, Sato K, Hatazawa J and

Okudera T (2001) Evaluation of novel PET ligands (+)N-[11C]methyl-3-piperidyl benzilate

([11C](+)3-MPB) and its stereoisomer [11C](-)3-MPB for muscarinic cholinergic receptors in

the conscious monkey brain: a PET study in comparison with [11C]4-MPB. Synapse

39:182-192.

Watanabe T (2007) The present conditions and the prospects of a skin application drug. Drug

Delivery System 22:450-457

Yamamura HI, Kuhar MJ and Snyder SH (1974) In vivo identification of muscarinic

cholinergic receptor binding in rat brain. Brain Res. 80:170-176

This article has not been copyedited and formatted. The final version may differ from this version.JPET Fast Forward. Published on March 7, 2008 as DOI: 10.1124/jpet.108.136390

at ASPE

T Journals on D

ecember 3, 2021

jpet.aspetjournals.orgD

ownloaded from

Page 21: In Vivo Quantitative Autoradiographic Analysis of Brain

JPET#136390

21

Yamamura HI and Snyder SH (1974) Muscarinic cholinergic binding in rat brain. Proc. Nat.

Acad. Sci. USA. 71:1725-1729

Yokoyama O, Ishiura Y, Nakamura Y, and Ohkawa M (1996) Urodynamic effects of

intravesical oxybutynin chloride in conscious rats. J Urol 155:768-771.

This article has not been copyedited and formatted. The final version may differ from this version.JPET Fast Forward. Published on March 7, 2008 as DOI: 10.1124/jpet.108.136390

at ASPE

T Journals on D

ecember 3, 2021

jpet.aspetjournals.orgD

ownloaded from

Page 22: In Vivo Quantitative Autoradiographic Analysis of Brain

JPET#136390

22

Footnotes

This study was financially supported in part by Shizuoka Triangle Cluster Initiative, Shizuoka,

and by CREST, Japan Science and Technology Agency (JST), Saitama, Japan.

This article has not been copyedited and formatted. The final version may differ from this version.JPET Fast Forward. Published on March 7, 2008 as DOI: 10.1124/jpet.108.136390

at ASPE

T Journals on D

ecember 3, 2021

jpet.aspetjournals.orgD

ownloaded from

Page 23: In Vivo Quantitative Autoradiographic Analysis of Brain

JPET#136390

23

LEGENDS FOR FIGURES

Fig. 1. Chemical structures of oxybutynin, propiverine, tolterodine, solifenacin and darifenacin

(A), and [11C](+)3-MPB (B).

Fig. 2. (A) Representative autoradiographic images (distribution of radioactivity) in the brain

regions of rats received i.v. injection of [11C](+)3-MPB. At 30 min after i.v. injection of

[11C](+)3-MPB (150 MBq) in rats, brain tissues were removed and cut every 2 mm as a center

of thalamus and analyzed for the autoradiography. (B) Effects of i.v. injection of different

doses of oxybutynin, propiverine, solifenacin, tolterodine and darifenacin on autoradiographic

images of [11C](+)3-MPB in rat brain. Rats received i.v. injection of different doses of each

antimuscarinic agent, and at 10 min later, they received i.v. injection of [11C](+)3-MPB (150

MBq). At 30 min later, brain tissues were removed.

Fig. 3. Effects of i.v. injection of oxybutynin, propiverine, solifenacin, tolterodine and

darifenacin on in vivo specific [11C](+)3-MPB binding in rat brain regions (cerebral cortex,

corpus striatum, hippocampus, amygdala, thalamus, hypothalamus and pons). Rats received i.v.

injection of [11C](+)3-MPB (150 MBq) 10 min after i.v. injection of oxybutynin, propiverine,

solifenacin, tolterodine and darifenacin. Specific [11C](+)3-MPB binding was determined as

described in Materials and Methods. Each column represents the mean ± SEM of three rats.

*,** Significantly different from the control values (vehicle), *p<0.05, **p<0.01.

Fig. 4. Dose-muscarinic receptor occupancy curves in each brain region of rats after i.v.

injection of oxybutynin (●), propiverine (▲), solifenacin (○) and tolterodine (△). Rats

received i.v. injection of [11C](+)3-MPB (150 MBq) 10min after i.v. injection of oxybutynin,

propiverine, solifenacin and tolterodine. The muscarinic receptor occupancy was determined

from the degree of reduction (%) by antimuscarinic agents of in vivo specific [11C](+)3-MPB

This article has not been copyedited and formatted. The final version may differ from this version.JPET Fast Forward. Published on March 7, 2008 as DOI: 10.1124/jpet.108.136390

at ASPE

T Journals on D

ecember 3, 2021

jpet.aspetjournals.orgD

ownloaded from

Page 24: In Vivo Quantitative Autoradiographic Analysis of Brain

JPET#136390

24

binding in each brain region as described in Materials and Methods. Each point represents the

mean ± SEM of three rats.

Fig. 5. Relationship between in vivo hippocampal receptor occupancy (average pRO50) after

i.v. injection of oxybutynin, propiverine, solifenacin and tolterodine (Table 1) and their in

vitro binding affinities (pKi values) of human recombinant M1 receptors. The in vitro binding

affinities data (pKi values) of human recombinant M1 receptors were obtained from Maruyama

et al. (2006). The solid line represents a linear regression of propiverine, solifenacin and

tolterodine. The multiple correlation coefficient was 0.9999 and the linear equation was pRO50

= 0.6946×pKi - 0.001054. The broken lines represent 95% confidence limits of the line. The

data represent the mean and 95% confidence interval.

This article has not been copyedited and formatted. The final version may differ from this version.JPET Fast Forward. Published on March 7, 2008 as DOI: 10.1124/jpet.108.136390

at ASPE

T Journals on D

ecember 3, 2021

jpet.aspetjournals.orgD

ownloaded from

Page 25: In Vivo Quantitative Autoradiographic Analysis of Brain

JPET#136390

25

TABLE 1 Brain muscarinic receptor occupancy (RO50 values) of antimuscarinic agents estimated from in

vivo competitive inhibition of specific [11C](+)3-MPB binding in rat brain regions

Average RO50 values (µmol/kg, i.v.)

Brain regions Oxybutynin

(95% CI)

Propiverine (95% CI)

Solifenacin (95% CI)

Tolterodine (95% CI)

Cerebral cortex

0.43 (0.31 - 0.60)

17.2 (10.1 - 28.2)

5.9 (4.0 - 8.6)

0.82 (0.57 - 1.2)

Corpus striatum

0.54 (0.38 - 0.77)

40.5 (16.3 - 101)

7.0 (4.2 - 11.4)

0.98 (0.68 - 1.4)

Hippocampus 0.50

(0.35 - 0.73) 24.7

(13.3 - 45.8) 4.6

(2.9 - 7.3) 0.89

(0.63 - 1.3)

Amygdala 0.39

(0.23 - 0.66) 18.1

(10.0 - 32.9) 3.3

(2.0 - 5.5) 0.61

(0.42 - 0.89)

Thalamus 0.34

(0.21 - 0.55) 12.3

(8.0 - 18.8) 8.2

(4.1 - 16.4) 0.62

(0.45 - 0.85)

Hypothalamus 0.28

(0.18 - 0.44) 10.0

(6.8 - 14.8) 6.2

(3.6 - 10.4) 0.40

(0.34 - 0.47)

Pons 0.57

(0.34 - 0.95) 14.5

(8.7 - 24.0) 10.7

(5.4 - 21.3) 0.34

(0.20 - 0.57)

Rats received i.v. injection of [11C](+)3-MPB (150 MBq) 10 min after i.v. injection of

oxybutynin (0.08 - 2.54 µmol/kg), propiverine (0.74 - 24.8 µmol/kg), solifenacin (0.62 - 20.8

µmol/kg), tolterodine (0.21 - 6.31 µmol/kg) and darifenacin (0.20 - 5.91 µmol/kg). Specific

[11C](+)3-MPB binding was determined and 50% muscarinic receptor occupancy (RO50) was

estimated as described in Materials and Methods.

This article has not been copyedited and formatted. The final version may differ from this version.JPET Fast Forward. Published on March 7, 2008 as DOI: 10.1124/jpet.108.136390

at ASPE

T Journals on D

ecember 3, 2021

jpet.aspetjournals.orgD

ownloaded from

Page 26: In Vivo Quantitative Autoradiographic Analysis of Brain

JPET#136390

26

TABLE 2

Comparison of observed and predicted (from Fig. 5) RO50 values of 50% muscarinic receptor

occupancy in the rat hippocampus

RO50 values (µmol/kg, i.v.)

Oxybutynin Propiverine Solifenacin Tolterodine

Observed values 0.50 24.7 4.6 0.89

Predicted values 2.5 24.4 4.7 0.88

Ratios (Predicted/Observed)

5.0 0.99 1.0 0.99

This article has not been copyedited and formatted. The final version may differ from this version.JPET Fast Forward. Published on March 7, 2008 as DOI: 10.1124/jpet.108.136390

at ASPE

T Journals on D

ecember 3, 2021

jpet.aspetjournals.orgD

ownloaded from

Page 27: In Vivo Quantitative Autoradiographic Analysis of Brain

JPET#136390

27

TABLE 3

The selectivity of antimuscarinic agents for the urinary bladder over the brain

Ratios (RO50/ID50)

Brain regions Oxybutynina,b,c Propiverineb Solifenacina,c Tolterodineb,c

Cerebral cortex 2.1 - 2.5 3.8 14.4, 25.8 8.8, 15.1

Corpus striatum 2.7 - 3.2 8.9 17.0, 30.4 10.4, 17.9

Hippocampus 2.5 - 3.0 5.4 11.2, 20.0 9.5, 16.3

Amygdala 1.9 - 2.3 4.0 8.1, 14.6 6.5, 11.1

Thalamus 1.7 - 2.0 2.7 19.9, 35.7 6.6, 11.3

Hypothalamus 1.4 - 1.7 2.2 15.0, 26.9 4.3, 7.3

Pons 2.9 - 3.4 3.2 26.1, 46.7 3.6, 6.2

RO50 values for antimuscarinic agents were derived from Table 1.

a,b,c Pharmacological potency (ID50) of each agent in the urinary bladder was derived from

inhibitory effects of carbachol- and volume-induced increases in the inravesical pressure in

rats by aIkeda et al. (2002), bModiri et al. (2002) and cOhtake et al. (2004)

This article has not been copyedited and formatted. The final version may differ from this version.JPET Fast Forward. Published on March 7, 2008 as DOI: 10.1124/jpet.108.136390

at ASPE

T Journals on D

ecember 3, 2021

jpet.aspetjournals.orgD

ownloaded from

Page 28: In Vivo Quantitative Autoradiographic Analysis of Brain

This article has not been copyedited and formatted. The final version may differ from this version.JPET Fast Forward. Published on March 7, 2008 as DOI: 10.1124/jpet.108.136390

at ASPE

T Journals on D

ecember 3, 2021

jpet.aspetjournals.orgD

ownloaded from

Page 29: In Vivo Quantitative Autoradiographic Analysis of Brain

This article has not been copyedited and formatted. The final version may differ from this version.JPET Fast Forward. Published on March 7, 2008 as DOI: 10.1124/jpet.108.136390

at ASPE

T Journals on D

ecember 3, 2021

jpet.aspetjournals.orgD

ownloaded from

Page 30: In Vivo Quantitative Autoradiographic Analysis of Brain

This article has not been copyedited and formatted. The final version may differ from this version.JPET Fast Forward. Published on March 7, 2008 as DOI: 10.1124/jpet.108.136390

at ASPE

T Journals on D

ecember 3, 2021

jpet.aspetjournals.orgD

ownloaded from

Page 31: In Vivo Quantitative Autoradiographic Analysis of Brain

This article has not been copyedited and formatted. The final version may differ from this version.JPET Fast Forward. Published on March 7, 2008 as DOI: 10.1124/jpet.108.136390

at ASPE

T Journals on D

ecember 3, 2021

jpet.aspetjournals.orgD

ownloaded from

Page 32: In Vivo Quantitative Autoradiographic Analysis of Brain

This article has not been copyedited and formatted. The final version may differ from this version.JPET Fast Forward. Published on March 7, 2008 as DOI: 10.1124/jpet.108.136390

at ASPE

T Journals on D

ecember 3, 2021

jpet.aspetjournals.orgD

ownloaded from