drug metabolism and pharmacokinetics · in drug discovery, the cerebrospinal fluid (csf) drug...

10
Regular article Investigation of utility of cerebrospinal uid drug concentration as a surrogate for interstitial uid concentration using microdialysis coupled with cisternal cerebrospinal uid sampling in wild-type and Mdr1a(/) rats Yoko Nagaya a, * , Yoshitane Nozaki a , Osamu Takenaka b , Ryuji Watari a , Kazutomi Kusano a , Tsutomu Yoshimura a , Hiroyuki Kusuhara c a Drug Metabolism and Pharmacokinetics Japan, Eisai Product Creation Systems, Eisai Co., Ltd., 5-1-3 Tokodai, Tsukuba-shi, Ibaraki, 300-2635, Japan b Modeling & Simulation, Clinical Pharmacology, Eisai Co., Ltd., 4-6-10 Koishikawa, Bunkyo-ku, Tokyo 112-8088, Japan c Laboratory of Molecular Pharmacokinetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-003, Japan article info Article history: Received 3 August 2015 Received in revised form 22 October 2015 Accepted 23 October 2015 Available online 5 November 2015 Keywords: P-glycoprotein Central nervous system Brain Cerebrospinal uid Interstitial uid Microdialysis Rats Drug discovery abstract In drug discovery, the cerebrospinal uid (CSF) drug concentration (C CSF ) has been used as a surrogate for the interstitial uid (ISF) concentration (C ISF ). However, the C CSF -to-C ISF gradient suggested for P-glyco- protein (P-gp) substrates in rodents causes uncertainty in C ISF estimations and subsequent pharmacokinetic-pharmacodynamic analyses. To evaluate the utility of C CSF as a surrogate for C ISF , this study directly compared the C CSF with the C ISF of 12 compounds, including P-gp substrates, under steady- state conditions in wild-type and Mdr1a(/) rats using microdialysis coupled with cisternal CSF sampling. In wild-type rats, the ISF-to-unbound plasma (K p,uu,ISF ) and CSF-to-unbound plasma (K p,uu,CSF ) concentration ratios of the P-gp substrates, except for metoclopramide, were lower than those of the non-P-gp substrates, and the K p,uu,CSF values were within or close to 3-fold of the K p,uu,ISF values for all the compounds examined. The K p,uu,CSF values of the selected P-gp substrates increased in Mdr1a(/) rats with a similar magnitude to the K p,uu,ISF values, resulting in the K p,uu,CSF -to-K p,uu,ISF ratios being un- changed. These results suggested that P-gp-mediated active efux at the bloodebrain barrier is a major determinant not only for C ISF , but also for C CSF , and that C CSF can be used as a surrogate for C ISF even for P- gp substrates in rats. Copyright © 2015 The Authors. Published by Elsevier Ltd on behalf of The Japanese Society for the Study of Xenobiotics. This is an open access article under the CC BY-NC-ND license (http://creativecommons.org/licenses/by- nc-nd/4.0/). 1. Introduction In drug discovery, understanding the relationship between the pharmacokinetics and the pharmacodynamics of synthesized compounds in animal pharmacological models is crucial for estimating the in vivo efcacious concentrations or exposure levels. Based on the free drug hypothesis[1], the unbound drug con- centration in plasma (C u,plasma ) is frequently used for such at- tempts; however, the C u,plasma at pharmacologically effective dose levels is not necessarily consistent with the in vitro effective con- centration, particularly for central nervous system (CNS)-acting compounds, because the C u,plasma can overestimate unbound drug concentrations in the brain (C u,brain ) because of the restrictive role of the bloodebrain barrier (BBB). The BBB has highly developed tight junctions and expresses active efux transporters such as P- glycoprotein (P-gp/ABCB1) [2], breast cancer resistance protein (BCRP/ABCG2) [3], and multidrug resistance-associated protein 4 (MRP4/ABCC4) [4], thereby limiting drug CNS penetration. Com- pounds with poor CNS penetration need higher doses to elicit their pharmacological effects in the CNS, resulting in higher systemic exposure and a narrower therapeutic index against peripheral toxicities. Therefore, establishing methodologies to estimate the List of nonstandard abbreviations: AUC, area under the plasma concen- trationetime curve; BBB, bloodebrain barrier; BCRP, breast cancer resistance pro- tein; BCSFB, blood-cerebrospinal uid barrier; CNS, central nervous system; CSF, cerebrospinal uid; E2074, 2-[(2R)-2-Fluoro-3-{(3r)-[(3-uorobenzyl)oxy]-8- azabicyclo[3.2.1]oct-8-yl}propyl]-4,5-dimethyl-2,4-dihydro-3H-1,2,4-triazol-3-one; ISF, interstitial uid; KO, knock out; MDR, multidrug resistance; MRP, multidrug resistance-associated protein; PBS, phosphate buffered saline; P-gp, P-glycoprotein; WT, wild type. * Corresponding author. Drug Metabolism and Pharmacokinetics Japan, Tsukuba Research Laboratories, Eisai Co., Ltd., 5-1-3, Tokodai, Tsukuba-shi, Ibaraki, 300- 2635, Japan. E-mail address: [email protected] (Y. Nagaya). Contents lists available at ScienceDirect Drug Metabolism and Pharmacokinetics journal homepage: http://www.journals.elsevier.com/drug-metabolism-and- pharmacokinetics http://dx.doi.org/10.1016/j.dmpk.2015.10.003 1347-4367/Copyright © 2015 The Authors. Published by Elsevier Ltd on behalf of The Japanese Society for the Study of Xenobiotics. This is an open access article under the CC BY-NC-ND license (http://creativecommons.org/licenses/by-nc-nd/4.0/). Drug Metabolism and Pharmacokinetics 31 (2016) 57e66

Upload: others

Post on 20-Jun-2020

3 views

Category:

Documents


1 download

TRANSCRIPT

Page 1: Drug Metabolism and Pharmacokinetics · In drug discovery, the cerebrospinal fluid (CSF) drug concentration (CCSF) has been used as a surrogate for the interstitial fluid (ISF)

lable at ScienceDirect

Drug Metabolism and Pharmacokinetics 31 (2016) 57e66

Contents lists avai

Drug Metabolism and Pharmacokineticsjournal homepage: http: / /www.journals .e lsevier .com/drug-metabol ism-and-

pharmacokinet ics

Regular article

Investigation of utility of cerebrospinal fluid drug concentration as a surrogate forinterstitial fluid concentration using microdialysis coupled with cisternalcerebrospinal fluid sampling in wild-type and Mdr1a(�/�) rats

Yoko Nagaya a, *, Yoshitane Nozaki a, Osamu Takenaka b, Ryuji Watari a, Kazutomi Kusano a,Tsutomu Yoshimura a, Hiroyuki Kusuhara c

a Drug Metabolism and Pharmacokinetics Japan, Eisai Product Creation Systems, Eisai Co., Ltd., 5-1-3 Tokodai, Tsukuba-shi, Ibaraki, 300-2635, Japanb Modeling & Simulation, Clinical Pharmacology, Eisai Co., Ltd., 4-6-10 Koishikawa, Bunkyo-ku, Tokyo 112-8088, Japanc Laboratory of Molecular Pharmacokinetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-003, Japan

a r t i c l e i n f o

Article history:Received 3 August 2015Received in revised form22 October 2015Accepted 23 October 2015Available online 5 November 2015

Keywords:P-glycoproteinCentral nervous systemBrainCerebrospinal fluidInterstitial fluidMicrodialysisRatsDrug discovery

List of nonstandard abbreviations: AUC, areatrationetime curve; BBB, bloodebrain barrier; BCRP,tein; BCSFB, blood-cerebrospinal fluid barrier; CNS,cerebrospinal fluid; E2074, 2-[(2R)-2-Fluoro-3-{azabicyclo[3.2.1]oct-8-yl}propyl]-4,5-dimethyl-2,4-dihISF, interstitial fluid; KO, knock out; MDR, multidrugresistance-associated protein; PBS, phosphate bufferedWT, wild type.* Corresponding author. Drug Metabolism and Phar

Research Laboratories, Eisai Co., Ltd., 5-1-3, Tokoda2635, Japan.

E-mail address: [email protected] (Y. Nagay

http://dx.doi.org/10.1016/j.dmpk.2015.10.0031347-4367/Copyright © 2015 The Authors. Published bBY-NC-ND license (http://creativecommons.org/licens

a b s t r a c t

In drug discovery, the cerebrospinal fluid (CSF) drug concentration (CCSF) has been used as a surrogate forthe interstitial fluid (ISF) concentration (CISF). However, the CCSF-to-CISF gradient suggested for P-glyco-protein (P-gp) substrates in rodents causes uncertainty in CISF estimations and subsequentpharmacokinetic-pharmacodynamic analyses. To evaluate the utility of CCSF as a surrogate for CISF, thisstudy directly compared the CCSF with the CISF of 12 compounds, including P-gp substrates, under steady-state conditions in wild-type and Mdr1a(�/�) rats using microdialysis coupled with cisternal CSFsampling. In wild-type rats, the ISF-to-unbound plasma (Kp,uu,ISF) and CSF-to-unbound plasma (Kp,uu,CSF)concentration ratios of the P-gp substrates, except for metoclopramide, were lower than those of thenon-P-gp substrates, and the Kp,uu,CSF values were within or close to 3-fold of the Kp,uu,ISF values for all thecompounds examined. The Kp,uu,CSF values of the selected P-gp substrates increased in Mdr1a(�/�) ratswith a similar magnitude to the Kp,uu,ISF values, resulting in the Kp,uu,CSF-to-Kp,uu,ISF ratios being un-changed. These results suggested that P-gp-mediated active efflux at the bloodebrain barrier is a majordeterminant not only for CISF, but also for CCSF, and that CCSF can be used as a surrogate for CISF even for P-gp substrates in rats.

Copyright © 2015 The Authors. Published by Elsevier Ltd on behalf of The Japanese Society for the Study ofXenobiotics. This is an open access article under the CC BY-NC-ND license (http://creativecommons.org/licenses/by-

nc-nd/4.0/).

1. Introduction

In drug discovery, understanding the relationship between thepharmacokinetics and the pharmacodynamics of synthesizedcompounds in animal pharmacological models is crucial for

under the plasma concen-breast cancer resistance pro-central nervous system; CSF,(3r)-[(3-fluorobenzyl)oxy]-8-ydro-3H-1,2,4-triazol-3-one;resistance; MRP, multidrugsaline; P-gp, P-glycoprotein;

macokinetics Japan, Tsukubai, Tsukuba-shi, Ibaraki, 300-

a).

y Elsevier Ltd on behalf of The Japaes/by-nc-nd/4.0/).

estimating the in vivo efficacious concentrations or exposure levels.Based on the “free drug hypothesis” [1], the unbound drug con-centration in plasma (Cu,plasma) is frequently used for such at-tempts; however, the Cu,plasma at pharmacologically effective doselevels is not necessarily consistent with the in vitro effective con-centration, particularly for central nervous system (CNS)-actingcompounds, because the Cu,plasma can overestimate unbound drugconcentrations in the brain (Cu,brain) because of the restrictive roleof the bloodebrain barrier (BBB). The BBB has highly developedtight junctions and expresses active efflux transporters such as P-glycoprotein (P-gp/ABCB1) [2], breast cancer resistance protein(BCRP/ABCG2) [3], and multidrug resistance-associated protein 4(MRP4/ABCC4) [4], thereby limiting drug CNS penetration. Com-pounds with poor CNS penetration need higher doses to elicit theirpharmacological effects in the CNS, resulting in higher systemicexposure and a narrower therapeutic index against peripheraltoxicities. Therefore, establishing methodologies to estimate the

nese Society for the Study of Xenobiotics. This is an open access article under the CC

Page 2: Drug Metabolism and Pharmacokinetics · In drug discovery, the cerebrospinal fluid (CSF) drug concentration (CCSF) has been used as a surrogate for the interstitial fluid (ISF)

Y. Nagaya et al. / Drug Metabolism and Pharmacokinetics 31 (2016) 57e6658

Cu,brain accurately is crucial for a quantitative understanding ofin vivo pharmacological effects based on in vitro effective concen-trations, as well as for optimizing the compound in terms of itsin vivo CNS penetration.

As unbound drugs in the brain interstitial fluid (ISF) can directlyinteract with the pharmacological targets in the CNS, measurementor estimation of the drug ISF concentration (CISF) is required.Microdialysis is the only method that can be used to assess CISFdirectly, wherein a microdialysis probe is implanted in a brain re-gion to supply and collect perfusate in equilibrium with unbounddrugs in the ISF [5]. This technique is useful for evaluating the CISF,but it involves technical challenges and has a low throughput,making it unsuitable for drug discovery settings. Instead, alterna-tive approaches have been proposed, such as using the product ofthe total drug concentration in the brain in vivo and the unboundfraction in the brain (fu,brain) determined in vitro using brain ho-mogenates [6] or brain slices [7], or using the cerebrospinal fluid(CSF) drug concentrations (CCSF).

The CCSF has been used as a surrogate for the CISF based on theassumption of a rapid equilibrium between the ISF and CSFcompartments across the ependymal layer. Because of low proteinconcentrations [8], the CCSF can be identical to the unbound drugconcentration in the CSF. One of the advantages of CSF sampling isthat this technique is applicable not only to nonclinical animalspecies, but also to human subjects in clinical studies. Thisadvantage enables a direct comparison of CCSF between animalsand humans [8], which can be used to provide supportive evi-dence to guide the efficacious dose range to be tested in clinicalstudies for CNS-acting drugs. Although the CCSF is considered tobe better than the Cu,plasma for predicting the CISF [6,9,10], theutility of CCSF as a surrogate for CISF is still debatable, and the CCSF-to-CISF gradient has been suggested for substrates of effluxtransporters, such as P-gp and BCRP, in rodents [8,11e13].Therefore, an understanding of the capability and limitations ofCCSF as a surrogate for CISF has been a topic of interest. Recently,we compared the CISF and the CCSF of 12 compounds, including P-gp substrates, in cynomolgus monkeys and found that the CCSFwas within 3-fold of the CISF, even for P-gp substrates [14].However, the impact of P-gp-mediated active efflux across theBBB on the CISF and CCSF could not be directly evaluated in thatmonkey study.

Recently, transporter-deficient rats have been developed [15,16],and these animal models are considered to be useful tools forevaluating the impact of efflux transporters on drug CNS distribu-tion [17]. In the pharmaceutical industry, rats are commonly usedfor toxicology and pharmacokinetic studies. In vivo pharmacolog-ical and behavioral models for CNS disorders have also beendeveloped using rats. The larger size of rats is one of their advan-tages over mice, even for CNS drug discovery, as it allows for sur-gical manipulation to develop pharmacological models and higherresolutions in imaging studies. Therefore, to evaluate the utility ofCCSF as a surrogate for CISF in rats, this study directly compared theCCSF with the CISF of 12 compounds, including P-gp substrates, atsteady-state conditions in wild-type rats using microdialysiscoupled with cisternal CSF sampling. Among the efflux transportersexpressed at the BBB, the present study focused on P-gp because P-gp substrate liability is a major hurdle in CNS drug discovery. Sixselected compounds were then further evaluated in Mdr1a(�/�)rats. Cu,brain was also determined as the product of the total drugconcentration in the brain and fu,brain obtained using the brainhomogenate method, or predicted fu,brain based on a pH partition-ing model. This study demonstrated that the CCSF of the testedcompounds, including that of the P-gp substrates, was within orclose to a practically acceptable threshold of 3-fold the CISF value inrats.

2. Materials and methods

2.1. Chemicals

Antipyrine, carbamazepine, lamotrigine, metoclopramide hy-drochloride, ondansetron hydrochloride dihydrate, propranololhydrochloride, quinidine hydrochloride, and verapamil hydro-chloride were purchased from Sigma Aldrich (St. Louis, MO). Mid-azolam and desloratadine were obtained fromWako Pure ChemicalIndustries (Osaka, Japan) and LKT Laboratories (St Paul, MN),respectively. Risperidone and paliperidone (9-hydroxyrisperidone)were obtained from AK Scientific (Union City, CA). E2074 [18]synthesized at the Eisai Tsukuba Research Laboratories (Ibaraki,Japan) was used. All other reagents and solvents were of analyticalgrade and were commercially available.

2.2. Animals

All the experimental protocols and procedures were approvedby the Institutional Animal Care and Use Committee of Eisai, and allthe animal experiments were conducted at an animal research fa-cility of Eisai accredited by the Center for Accreditation of Labora-tory Animal Care and Use, Japan Health Sciences Foundation. MaleSpragueeDawley wild-type and Mdr1a(�/�) rats (8e9 weeks old)were purchased from Charles River Laboratories Japan (Kanagawa,Japan) and SAGE Labs (Boyertown, PA), respectively. The animalswere maintained at 20 �Ce26 �C and a relative humidity of 40%e70% on a 12-h lightedark cycle with free access to food and water.All surgeries were performed under anesthesia, and all efforts weremade to minimize suffering.

2.3. Microdialysis

In vivo microdialysis was performed in awake, freely moving,jugular vein and carotid artery-cannulated wild-type andMdr1a(�/�) rats. One to two days before dosing, C-I-4 micro-dialysis probes (Eicom, Kyoto, Japan) with 5-mm (for hippocam-pus) and 3-mm (for frontal cortex) regenerated cellulosemembranes (50-kDa molecular weight cutoff) were stereotaxicallyimplanted under pentobarbital anesthesia (50 mg/kg, i.p.) into thefrontal cortex (3.2 mm anterior, 0.8 mm lateral from bregma, and4.5 mm ventral from dura) and hippocampus (5 mm posterior,5 mm lateral from bregma, and 8 mm ventral from dura) [19],respectively, and were fixed to the skull using dental cement. Onthe day of the experiment, the probe was perfused with RingerHEPES buffer at a flow rate of 1.5 mL/min. One to 2 h after the pre-perfusion, a cocktail of two or three compounds was infused viathe jugular vein following an intravenous loading dose. The infu-sion rate and loading dose are given in Figs. 1 and 2. In this study,cassette dosing was used to minimize the study period and thenumber of animals that were used. Since some compounds testedin this study were reported to be P-gp inhibitors (e.g., verapamiland quinidine), the dose levels were optimized to maintain theCu,plasma equal to or less than the lower limit of the reported half-maximal inhibitory concentrations for P-gp [20] at steady state,thereby minimizing the risk of P-gp-mediated drug-drug in-teractions at the BBB [21].

The microdialysis dialysate was serially collected from thefrontal cortex and hippocampus at 30-min intervals up to 4 hpostdosing. Blood was also serially collected at 1-h intervals up to4 h postdosing via the carotid artery cannula to prepare the plasmasamples.

After the 4-h sampling, CSF was immediately collected bycisternal puncture under isoflurane anesthesia (3% inhalation),and the animals were then euthanized by exsanguination under

Page 3: Drug Metabolism and Pharmacokinetics · In drug discovery, the cerebrospinal fluid (CSF) drug concentration (CCSF) has been used as a surrogate for the interstitial fluid (ISF)

Fig. 1. Concentration-time profiles of antipyrine (A), carbamazepine (B), lamotrigine (C), midazolam (D), metoclopramide (E), ondansetron (F), E2074 (G), risperidone (H), verapamil(I), quinidine (J), paliperidone (K), and desloratadine (L) in unbound plasma (Cu,plasma, open circles), ISF in brain cortex (CISF,Cx, closed triangles), and ISF in hippocampus (CISF,HIP,open triangles) in wild-type rats after a constant intravenous infusion with an intravenous loading dose. The infusion rate (mg/h/kg) and loading dose (mg/kg) were as follows: 0.10and 0.22 for antipyrine, 0.20 and 0.21 for carbamazepine, 0.050 and 0.89 for lamotrigine, 0.40 and 0.40 for midazolam, 1.0 and 0.80 for metoclopramide, 1.0 and 0.46 forondansetron, 0.40 and 0.33 for E2074, 1.0 and 0.70 for risperidone, 1.0 and 0.90 for verapamil, 5.0 and 8.0 for quinidine, 1.0 and 0.80 for paliperidone, and 5.0 and 5.0 fordesloratadine. Blood and dialysate samples were serially collected for up to 4 h. The ISF concentrations were obtained from dialysate concentrations corrected for microdialysisprobe recoveries (Supplemental Table 1). Each point represents the mean ± SEM of three to five animals.

Y. Nagaya et al. / Drug Metabolism and Pharmacokinetics 31 (2016) 57e66 59

anesthesia to collect blood and brain tissues (frontal cortex andhippocampus contralateral to the microdialysis probe-implantedside). The brain tissue samples were homogenized in distilledwater to prepare 20% homogenates (w/v) immediately beforeuse.

2.4. Microdialysis probe recovery

The microdialysis probe recovery of the test compounds wasdetermined as described earlier [14], and the details are given inSupplemental Table 1. Based on the probe recoveries, the concen-tration of a test compound in ISF (CISF) from the frontal cortex(CISF,Cx) or hippocampus (CISF,HIP) was calculated using the in vivodialysate concentration (Cdialysate) as follows:

CISF;Cx or CISF;HIP ¼ CdialysateRecovery

Our pilot study confirmed that the recovery values for antipy-rine, lamotrigine, E2074, quinidine, risperidone, and verapamil inwild-type rats were consistent with those in Mdr1a(�/�) rats forboth the brain cortex and hippocampus regions. Thus, the recoveryvalues of the test compounds obtained from wild-type rats wereused in this study.

2.5. Drug unbound fractions in rat plasma and brain tissues

The unbound fractions of the test compounds in rat plasma(fu,plasma) and brain (fu,brain) for cortex (fu,brain,Cx) and hippocampus

Page 4: Drug Metabolism and Pharmacokinetics · In drug discovery, the cerebrospinal fluid (CSF) drug concentration (CCSF) has been used as a surrogate for the interstitial fluid (ISF)

Fig. 2. Concentration-time profiles of antipyrine (A), lamotrigine (B), E2074 (C), risperidone (D), verapamil (E), and quinidine (F) in unbound plasma (Cu,plasma, open circles), ISF inbrain cortex (CISF,Cx, closed triangles), and ISF in hippocampus (CISF,HIP, open triangles) in Mdr1a(�/�) rats after a constant intravenous infusion with an intravenous loading dose.The infusion rate (mg/h/kg) and loading dose (mg/kg) were as follows: 0.10 and 0.22 for antipyrine, 0.050 and 0.89 for lamotrigine, 0.20 and 0.17 for E2074, 0.50 and 0.35 forrisperidone, 1.0 and 0.90 for verapamil, and 3.0 and 5.0 for quinidine. Blood and dialysate samples were serially collected from Mdr1a(�/�) rats for up to 4 h. The ISF concentrationswere obtained from dialysate concentrations corrected for microdialysis probe recoveries (Supplemental Table 1). Each point represents the mean ± SEM of three to four animals.

Y. Nagaya et al. / Drug Metabolism and Pharmacokinetics 31 (2016) 57e6660

(fu,brain,HIP) were determined by equilibrium dialysis as describedpreviously [14]; the details are given in Table 1. The fu,plasma wascalculated as the receiver (phosphate buffered saline, PBS) side-to-donor (plasma) side concentration ratio. The fu,brain,Cx and fu,brain,HIPwere calculated as follows [11]:

fu;brain;Cx or fu;brain;HIP ¼ 1=D�1�fu0 � 1

�þ 1=D

where D and fu0 represent the dilution factor (5-fold) used to pre-pare 20% tissue homogenates and the unbound fraction (deter-mined as the ratio of the drug concentration in PBS to that in 20%brain tissue homogenate), respectively. According to our pre-liminary experiments, no remarkable difference in the whole brainhomogenate binding of selected P-gp substrates was observed be-tween wild-type and Mdr1a(�/�) rats, for which the fu,brain values(mean of 3 determinations) of quinidine, risperidone, and verap-amil were 0.0286, 0.0893, and 0.0176 in wild-type rats and 0.0342,0.0931, and 0.0280 in Mdr1a(�/�) rats, respectively. Thus, thefu,brain,Cx and fu,brain,HIP values obtained from wild-type rats wereused in this study.

2.6. Analytical procedure

Samples obtained from the microdialysis probe recovery, theprotein-binding assay, and the in vivo studies were mixed withacetonitrile containing an internal standard (300 nM propranolol),

followed by centrifugation. The resulting supernatant was filtrated,and an aliquot of the filtrate was subjected to high-performanceliquid chromatography with tandem mass spectrometry analysis.The analytical conditions were reported previously [14] except forthose for paliperidone, which was subjected to electrospray ioni-zation in positive ion mode and was monitored at 427.4 > 207.4.

2.7. Calculations

The brain-to-plasma total drug concentration ratio (Kp,brain) forthe brain cortex (Kp,brain,Cx) or hippocampus (Kp,brain,HIP) was ob-tained by dividing the total concentration in the brain cortex(Cbrain,Cx) or hippocampus (Cbrain,HIP) by the plasma total concen-tration (Cplasma), respectively. The plasma unbound concentration(Cu,plasma) was a product of Cplasma and fu,plasma. The unbound drugconcentrations in the brain cortex (Cu,Cx,homogenate) and hippo-campus (Cu,HIP,homogenate), as determined using the brain tissuehomogenate method, were obtained from Cbrain,Cx and Cbrain,HIPmultiplied by fu,brain,Cx and fu,brain,HIP, respectively. The ISF-to-plasma unbound concentration ratio (Kp,uu,ISF) for the brain cortex(Kp,uu,ISF,Cx) or hippocampus (Kp,uu,ISF,HIP), the CSF-to-plasma un-bound concentration ratio (Kp,uu,CSF), and the ratio of the brainunbound concentration determined using the brain homogenatemethod to the plasma unbound concentration (Kp,uu,brain) for thebrain cortex (Kp,uu,brain,Cx) or hippocampus (Kp,uu,brain,HIP) werecalculated as follows:

Page 5: Drug Metabolism and Pharmacokinetics · In drug discovery, the cerebrospinal fluid (CSF) drug concentration (CCSF) has been used as a surrogate for the interstitial fluid (ISF)

Table 1Unbound fractions of test compounds in rat plasma, brain cortex and hippocampustissues.

Test Compounds Unbound fraction

fu,plasma fu,brain,Cx fu,brain,HIP

Antipyrine 0.942 ± 0.049 0.715 ± 0.048 0.691 ± 0.178Carbamazepine 0.389 ± 0.013 0.158 ± 0.001 0.131 ± 0.015Lamotrigine 0.527 ± 0.018 0.176 ± 0.008 0.145 ± 0.010Midazolam 0.0696 ± 0.0003 0.0130 ± 0.0019 0.0160 ± 0.0027Metoclopramide 0.818 ± 0.016 0.359 ± 0.037 0.397 ± 0.068Ondansetron 0.421 ± 0.045 0.143 ± 0.013 0.139 ± 0.010E2074 0.675 ± 0.009 0.0975 ± 0.0199 0.106 ± 0.012Risperidone 0.126 ± 0.015 0.0492 ± 0.0068 0.0516 ± 0.0079Verapamil 0.194 ± 0.025 0.0317 ± 0.0093 0.0305 ± 0.0087Quinidine 0.527 ± 0.039 0.0284 ± 0.0081 0.0213 ± 0.0046Pailperidone 0.211 ± 0.025 0.154 ± 0.018 0.132 ± 0.015Desloratadine 0.193 ± 0.005 0.00892 ± 0.00108 0.00911 ± 0.00046

The unbound fractions of test compounds in rat plasma (fu,plasma), brain cortex(fu,brain,Cx) and hippocampus (fu,brain,HIP) were determined using equilibrium dial-ysis. The brain cortex and hippocampus tissues dissected from the brains of naïvemale wild-type rats were homogenized in distilled water to prepare 20% homoge-nate (w/v). Blank plasma freshly prepared from rat blood was used. The concen-trations of the compounds spiked into the plasma were as follows: 0.1 mg/mL forcarbamazepine, midazolam, metoclopramide, and E2074; 0.2 mg/mL for antipyrineand ondansetron; 0.3 mg/mL for verapamil and paliperidone; 0.5 mg/mL for lamo-trigine and risperidone; and 1 mg/mL for quinidine and desloratadine. The concen-trations of the compounds spiked into the brain cortex and hippocampushomogenates were as follows: 0.1 mg/mL for carbamazepine, midazolam, E2074,risperidone, verapamil, and paliperidone; 0.2 mg/mL for antipyrine, metoclopra-mide, and ondansetron; 1 mg/mL for lamotrigine and quinidine; and 2 mg/mL fordesloratadine. One hundred and 50 mL each of the matrices and PBS (pH7.4) wereadded to the donor and the receiver sides of the dialysis chamber, respectively, andthe dialysis apparatus HTD 96b (HTDialysis, LLC., Gales Ferry, CT) was incubated at37 �C for 24 h. Each value represents the mean ± SEM of three to six animals.

Y. Nagaya et al. / Drug Metabolism and Pharmacokinetics 31 (2016) 57e66 61

Kp;uu;ISF;Cx or Kp;uu;ISF;HIP ¼ CISF;Cx or CISF;HIPCu;plasma

Kp;uu;CSF ¼CCSF

Cu;plasma

Kp;uu;brain;Cx or Kp;uu;brain;HIP¼Cu;Cx;homogenate or Cu;HIP;homogenate

Cu;plasma

3. Results

3.1. ISF, CSF, and brain distribution of test compounds in wild-typerats

Eleven compounds that were characterized as non-P-gp sub-strates (antipyrine, carbamazepine, lamotrigine, and midazolam;corrected efflux ratio, CER< 2), weak P-gp substrates (metoclo-pramide, ondansetron, and E2074; 2 � CER< 3), or good P-gpsubstrates (risperidone, verapamil, desloratadine, and quinidine;3 � CER) in our previous study [14] were selected. In addition,paliperidone was also included as a good P-gp substrate [22] in thepresent study.

The time profiles for the Cu,plasma, CISF,Cx, and CISF,HIP of the testcompounds in wild-type rats are shown in Fig. 1. Cu,plasma wascalculated as the product of the total plasma concentrations andfu,plasma values (Table 1). As the plasma and ISF concentrations of allthe test compounds reached steady state within 4 h after the startof infusion (Fig. 1), the Kp,uu,ISF,Cx and Kp,uu,ISF,HIP values (Table 2)were determined at 4 h postdosing using the Cu,plasma, CISF,Cx, andCISF,HIP values (Supplemental Table 2). The Kp,uu,ISF,Cx values ranged

from 0.0180 (desloratadine) to 1.38 (antipyrine), which generallyagreed with the corresponding Kp,uu,ISF,HIP values ranging from0.0617 (paliperidone) to 1.12 (antipyrine).

Cisternal CSF and brain cortex and hippocampus tissues as wellas plasma were collected immediately after the 4-h ISF sampling inwild-type rats (Supplemental Table 3), at which time the Kp,uu,CSF,Kp,brain,Cx, and Kp,brain,HIP values were determined (Table 2). TheKp,uu,CSF values ranged from 0.0678 (quinidine) to 1.51 (midazolam).The Kp,brain,Cx ranged from 0.227 (paliperidone) to 3.11 (mid-azolam), which agreed with the corresponding Kp,brain,HIP valuesranging from 0.172 (paliperidone) to 3.17 (midazolam).

The unbound fractions of the test compounds in rat brain cortex(fu,brain,Cx) and hippocampus (fu,brain,HIP) tissues were determinedby equilibrium dialysis (Table 1). No remarkable differences be-tween the fu,brain,Cx and the fu,brain,HIP values were observed. Usingthese unbound fractions, the unbound drug concentrations in thebrain cortex and hippocampus were calculated to estimate theKp,uu,brain,Cx and Kp,uu,brain,HIP values, respectively (Table 2). TheKp,uu,brain,Cx values ranged from 0.0220 (quinidine) to 0.805(metoclopramide), which agreed with the Kp,uu,brain,HIP valuesranging from 0.0165 (quinidine) to 0.807 (metoclopramide).

For the compounds for which data were available, the Kp,uu,ISF,Kp,uu,CSF, Kp,brain, and Kp,uu,brain values determined in the presentstudy were close to or within the ranges of the respective valuesreported for wild-type rats (Supplemental Table 4).

3.2. ISF, CSF, and brain distribution of test compounds in Mdr1a(�/�) rats

For antipyrine, lamotrigine, E2074, risperidone, verapamil, andquinidine, the CNS distributions were also investigated inMdr1a(�/�) rats, and the time profiles for Cu,plasma, CISF,Cx, andCISF,HIP are shown in Fig. 2. As the plasma and ISF concentrationsreached steady state within 4 h after the start of infusion, theKp,uu,ISF,Cx and Kp,uu,ISF,HIP values were determined at 4 h postdosing(Table 3). The Kp,uu,ISF,Cx values agreed with the correspondingKp,uu,ISF,HIP values, with smaller variations observed across thecompounds in Mdr1a(�/�) rats (Kp,uu,ISF,Cx, 0.438 to 0.941;Kp,uu,ISF,HIP, 0.559 to 1.26) compared with the variations observed inwild-type rats (Kp,uu,ISF,Cx, 0.0674 to 1.38; Kp,uu,ISF,HIP, 0.111 to 1.12).

Cisternal CSF and brain cortex and hippocampus tissues as wellas plasma were collected immediately after the 4-h ISF sampling inMdr1a(�/�) rats (Supplemental Table 5), at which time theKp,uu,CSF, Kp,brain,Cx, and Kp,brain,HIP values were determined (Table 3).The Kp,uu,CSF values ranged from 0.611 (verapamil) to 1.02 (antipy-rine). The Kp,brain,Cx and Kp,brain,HIP values showed large variationsacross the compounds (Kp,brain,Cx, 0.868 to 23.6; Kp,brain,HIP, 0.816 to22.2), but the corresponding Kp,uu,brain,Cx and Kp,uu,brain,HIP valuesshowed relatively smaller variations (0.646e1.75 and 0.488 to 1.69,respectively) in Mdr1a(�/�) rats.

3.3. Comparison of CNS distribution of P-glycoprotein substratesand nonsubstrates between wild-type and Mdr1a(�/�) rats

The Kp,uu,ISF,Cx, Kp,uu,ISF,HIP, Kp,uu,CSF, Kp,brain,Cx, and Kp,brain,HIPvalues of antipyrine, lamotrigine, E2074, risperidone, verapamil,and quinidine were compared betweenwild-type and Mdr1a(�/�)rats (Fig. 3). The Kp,uu,ISF, Kp,uu,CSF, and Kp,brain values of antipyrine(0.68- to 1.2-fold increase) and lamotrigine (1.2- to 1.4-fold in-crease) were minimally affected by Mdr1a knockout, consistentwith previous findings demonstrating that these compounds arenon-P-gp substrates [14].

The CNS distributions of P-gp substrates (E2074, risperidone,verapamil, and quinidine) in Mdr1a(�/�) rats were greater thanthose in wild-type rats. The effects of the absence of Mdr1a on the

Page 6: Drug Metabolism and Pharmacokinetics · In drug discovery, the cerebrospinal fluid (CSF) drug concentration (CCSF) has been used as a surrogate for the interstitial fluid (ISF)

Table 2ISF, CSF, and brain distribution of test compounds in wild-type rats.

Test compounds Wild-type rats

Kp,uu,ISF,Cx Kp,uu,ISF,HIP Kp,uu,CSF Kp,brain,Cx Kp,brain,HIP Kp,uu,brain,Cx Kp,uu,brain,HIP

Antipyrine 1.38 ± 0.08 1.12 ± 0.05 1.08 ± 0.02 0.774 ± 0.024 0.664 ± 0.027 0.587 ± 0.018 0.487 ± 0.020Carbamazepine 0.643 ± 0.040 0.749 ± 0.073 0.915 ± 0.080 1.49 ± 0.15 1.49 ± 0.11 0.604 ± 0.060 0.502 ± 0.036Lamotrigine 0.643 ± 0.059 0.666 ± 0.124 0.628 ± 0.103 1.43 ± 0.12 1.37 ± 0.12 0.478 ± 0.041 0.377 ± 0.032Midazolam 0.652 ± 0.130 0.620 ± 0.094 1.51 ± 0.18 3.11 ± 0.18 3.17 ± 0.20 0.581 ± 0.033 0.728 ± 0.046Metoclopramide 0.669 ± 0.100 0.713 ± 0.088 0.422 ± 0.013 1.83 ± 0.10 1.66 ± 0.14 0.805 ± 0.042 0.807 ± 0.067Ondansetron 0.283 ± 0.024 0.391 ± 0.056 0.243 ± 0.018 1.43 ± 0.03 1.31 ± 0.05 0.486 ± 0.009 0.432 ± 0.017E2074 0.178 ± 0.030 0.163 ± 0.023 0.169 ± 0.022 2.05 ± 0.12 2.06 ± 0.13 0.296 ± 0.018 0.323 ± 0.021Risperidone 0.119 ± 0.021 0.163 ± 0.021 0.161 ± 0.016 0.286 ± 0.039 0.200 ± 0.016 0.112 ± 0.015 0.0820 ± 0.0064Verapamil 0.0718 ± 0.0112 0.114 ± 0.023 0.172 ± 0.072 0.484 ± 0.079 0.404 ± 0.055 0.0791 ± 0.0129 0.0635 ± 0.0087Quinidine 0.0674 ± 0.0067 0.111 ± 0.028 0.0678 ± 0.0125 0.408 ± 0.053 0.409 ± 0.005 0.0220 ± 0.0029 0.0165 ± 0.0002Paliperidone 0.0600 ± 0.0092 0.0617 ± 0.0132 0.0890 ± 0.0230 0.227 ± 0.035 0.172 ± 0.027 0.166 ± 0.025 0.108 ± 0.017Desloratadine 0.0180 ± 0.040 0.0754 ± 0.0211 0.0815 ± 0.0221 1.56 ± 0.19 1.55 ± 0.20 0.0722 ± 0.0088 0.0730 ± 0.0093

Kp,uu,ISF, Kp,brain, and Kp,uu,brain values in brain cortex (Kp,uu,ISF,Cx, Kp,brain,Cx, Kp,uu,brain,Cx) and hippocampus (Kp,uu,ISF,HIP, Kp,brain,HIP, and Kp,uu,brain,HIP) and Kp,uu,CSF values of testcompounds in wild-type rats were determined 4 h after the start of infusion. Each value represents mean ± SEM of three to five animals.

Table 3ISF, CSF, and brain distribution of test compounds in Mdr1a(�/�) rats.

Test compounds Mdr1a(�/�) rats

Kp,uu,ISF,Cx Kp,uu,ISF,HIP Kp,uu,CSF Kp,brain,Cx Kp,brain,HIP Kp,uu,brain,Cx Kp,uu,brain,HIP

Antipyrine 0.941 ± 0.021 1.26 ± 0.17 1.02 ± 0.07 0.868 ± 0.012 0.816 ± 0.026 0.659 ± 0.009 0.599 ± 0.019Lamotrigine 0.782 ± 0.011 0.766 ± 0.082 0.901 ± 0.059 1.93 ± 0.16 1.77 ± 0.12 0.646 ± 0.055 0.488 ± 0.032E2074 0.462 ± 0.057 0.639 ± 0.080 0.621 ± 0.096 9.30 ± 0.94 9.30 ± 0.97 1.34 ± 0.14 1.46 ± 0.15Risperidone 0.443 ± 0.055 0.732 ± 0.131 0.796 ± 0.053 2.83 ± 0.16 2.72 ± 0.16 1.04 ± 0.06 1.11 ± 0.06Verapamil 0.438 ± 0.025 0.559 ± 0.088 0.611 ± 0.075 10.7 ± 0.9 10.7 ± 0.9 1.75 ± 0.15 1.69 ± 0.14Quinidine 0.785 ± 0.206 1.23 ± 0.22 0.835 ± 0.058 23.6 ± 1.1 22.2 ± 1.4 1.27 ± 0.06 0.896 ± 0.057

Kp,uu,ISF, Kp,brain, and Kp,uu,brain values in brain cortex (Kp,uu,ISF,Cx, Kp,brain,Cx, Kp,uu,brain,Cx) and hippocampus (Kp,uu,ISF,HIP, Kp,brain,HIP, and Kp,uu,brain,HIP) and Kp,uu,CSF values of sixselected test compounds in Mdr1a(�/�) rats were determined 4 h after the start of infusion. Each value represents mean ± SEM of three to four animals.

Fig. 3. Mdr1a knockout (KO)/wild-type (WT) ratios for the Kp,uu,ISF, Kp,uu,CSF, and Kp,brain

values. The KO/WT ratio represents the ratio of the mean Kp,uu,ISF,Cx, Kp,uu,ISF,HIP,Kp,uu,CSF, Kp,brain,Cx, or Kp,brain,HIP value of antipyrine, lamotrigine, E2074, risperidone,verapamil, or quinidine in Mdr1a(�/�) rats to that in wild-type rats. The Kp,uu,ISF,Kp,uu,CSF, and Kp,brain values were obtained from Tables 2 and 3. Open bars, Kp,uu,ISF,Cx;closed bars, Kp,uu,ISF,HIP; dotted bars, Kp,uu,CSF; hatched bars, Kp,brain,Cx; horizontal stri-ped bars, Kp,brain,HIP.

Y. Nagaya et al. / Drug Metabolism and Pharmacokinetics 31 (2016) 57e6662

ISF, CSF, and total brain concentrations of E2074 were similar, withthe Kp,uu,ISF,Cx, Kp,uu,ISF,HIP, Kp,uu,CSF, Kp,brain,Cx, and Kp,brain,HIP valuesbeing 2.6-fold, 3.9-fold, 3.7-fold, 4.5-fold, and 4.5-fold higher inMdr1a(�/�) rats than in wild-type rats, respectively. The Mdr1adefect had a greater impact on the total drug concentration in thebrain than on the ISF or CSF concentrations, for good P-gp sub-strates (risperidone, verapamil, and quinidine). The magnitude of

the increase in the Kp,uu,ISF,Cx and Kp,uu,ISF,HIP values of risperidone(3.7-fold and 4.5-fold, respectively), verapamil (6.1-fold and 4.9-fold, respectively), and quinidine (12-fold and 11-fold, respec-tively) by Mdr1a knockout was comparable to that in correspond-ing Kp,uu,CSF values (4.9-fold for risperidone, 3.6-fold for verapamil,and 12-fold for quinidine). Meanwhile, a more pronounced increasewas observed for the Kp,brain,Cx and Kp,brain,HIP values of risperidone(9.9-fold and 14-fold, respectively), verapamil (22-fold and 26-fold,respectively), and quinidine (58-fold and 54-fold, respectively).

3.4. Comparison of Kp,uu,ISF, Kp,uu,CSF, and Kp,uu,brain values in rats

In the present study, the parameters were considered to besimilar if the two values were within 3-fold. This 3-fold thresholdwas chosen to allow for differences arising from random experi-mental error and for actual differences that could be considered tohave little impact on pharmacological and pharmacokinetic con-sequences in nonclinical studies [23].

In the brain cortex, the Kp,uu,ISF,Cx values showed a good agree-ment with the Kp,uu,CSF values within a 3-fold difference with theexception of desloratadine, for which the Kp,uu,CSF was 4.5-foldhigher than the Kp,uu,ISF,Cx in wild-type rats (Fig. 4A). In the hip-pocampus, the Kp,uu,ISF,HIP values agreed with the Kp,uu,CSF values,and the differences for all the test compounds were within 3-fold(Fig. 4D).

The Kp,uu,ISF,Cx values generally agreed with the Kp,uu,brain,Cx

values, but the Kp,uu,brain,Cx values of quinidine and desloratadine inwild-type rats and of verapamil in Mdr1a(�/�) rats were 0.25-fold,3.1-fold, and 4.0-fold of the Kp,uu,ISF,Cx values, respectively (Fig. 4B).Similarly, the Kp,uu,ISF,HIP values agreed with the Kp,uu,brain,HIP valueswithin a 3-fold difference with the exception of those for quinidine,for which the Kp,uu,ISF,HIP value was 6.7-fold greater than theKp,uu,brain,HIP value in wild-type rats (Fig. 4E).

Page 7: Drug Metabolism and Pharmacokinetics · In drug discovery, the cerebrospinal fluid (CSF) drug concentration (CCSF) has been used as a surrogate for the interstitial fluid (ISF)

Fig. 4. Comparison of Kp,uu,ISF, Kp,uu,CSF, and Kp,uu,brain values in rats. Relationships between Kp,uu,ISF,Cx and Kp,uu,CSF (A), Kp,uu,ISF,Cx and Kp,uu,brain,Cx (B), Kp,uu,brain,Cx and Kp,uu,CSF (C),Kp,uu,ISF,HIP and Kp,uu,CSF (D), Kp,uu,ISF,HIP and Kp,uu,brain,HIP (E), and Kp,uu,brain,HIP and Kp,uu,CSF (F). Each value is presented as the mean ± SEM of three to five animals, and the open andclosed circles represent the values in wild-type rats and Mdr1a(�/�) rats, respectively. The solid line passing through the origin represents the line of unity ± 3-fold (dashed lines). 1,antipyrine; 2, carbamazepine; 3, lamotrigine; 4, midazolam; 5, metoclopramide; 6, ondansetron; 7, E2074; 8, risperidone; 9, verapamil; 10, quinidine; 11, paliperidone; 12,desloratadine.

Y. Nagaya et al. / Drug Metabolism and Pharmacokinetics 31 (2016) 57e66 63

The Kp,uu,brain,Cx values were within or close to 3-fold of theKp,uu,CSF values, and only quinidine had a Kp,uu,CSF value thatwas 3.1-fold higher than the Kp,uu,brain,Cx value inwild-type rats (Fig. 4C). TheKp,uu,brain,HIP values were within or close to 3-fold of the Kp,uu,CSFvalues, and only quinidine had a Kp,uu,CSF value that was 4.1-foldhigher than the Kp,uu,brain,HIP value in wild-type rats (Fig. 4F).

The Kp,uu,ISF or Kp,uu,brain values were compared between thebrain cortex and hippocampus (Supplemental Fig. 1). No regionaldifferences in the Kp,uu,ISF and Kp,uu,brain values were observedexcept for the values for desloratadine, for which the Kp,uu,ISF,HIPvaluewas 4.2-fold higher than the Kp,uu,ISF,Cx value inwild-type rats.These results suggested that the brain cortex and the hippocampuscan be considered as being nearly identical in terms of drug dis-tribution at steady states, even for P-gp substrates.

The present study determined the fu,brain values using brainhomogenates. The unbound volume of distribution in a rat brainslice (Vu,brain) can be predicted by a pH partitioningmodel using thefu,brain and pKa of a compound [24]. Based on the proposed model,the Kp,uu,brain values were estimated in wild-type rats(Supplemental Fig. 2). Because of trapping in acidic intracellularcompartments, the pH partitioningmodel produces lower Kp,uu,brainvalues for basic compounds (e.g., quinidine) than the brain ho-mogenate method, resulting in larger differences between Kp,uu,-

brain and Kp,uu,ISF or Kp,uu,CSF for good P-gp substrates.

3.5. Comparison of drug CNS distributions between rats andcynomolgus monkeys

With the exception of paliperidone, we previously examined theKp,uu,ISF, Kp,uu,CSF, Kp,brain, and Kp,uu,brain values of the test compounds

at steady state in cynomolgus monkeys [14]. To evaluate speciesdifferences in the CNS distributions, the Kp,uu,ISF, Kp,uu,CSF, Kp,uu,brain,and Kp,brain values obtained in wild-type rats were compared withthose obtained in cynomolgus monkeys (Fig. 5).

The Kp,uu,ISF values in cynomolgusmonkeys tended to be slightlyoverestimated, compared with the Kp,uu,ISF,Cx values in rats, but thedifferences were within 3-fold except for that for desloratadine, forwhich the Kp,uu,ISF value in cynomolgus monkeys was 3.4-foldhigher than that in rats (Fig. 5A). The Kp,uu,CSF values in cyn-omolgus monkeys also tended to be slightly overestimated,compared with the Kp,uu,CSF values in rats, but the differences werewithin 3-fold except for that for E2074, for which the Kp,uu,CSF valuein cynomolgus monkeys was 4.3-fold higher than that in rats(Fig. 5B). Meanwhile, the Kp,uu,brain values in cynomolgus monkeyswere overestimated by more than 3-fold of the Kp,uu,brain,Cx valuesin rats for 4 of the 11 test compounds, all of which were P-gpsubstrates (E2074, risperidone, quinidine, and desloratadine)(Fig. 5D). This discrepancy was mainly attributable to higher Kp,brainvalues in cynomolgus monkeys for E2074, risperidone, anddesloratadine (Fig. 5C), and their fu,plasma and fu,brain,Cx valuesshowed minimal species differences (Supplemental Fig. 3).

4. Discussions

To understand the effects of drug candidates on the CNS quan-titatively, the monitoring of unbound drug concentrations in thebrain is indispensable. Although the CCSF is widely used as a sur-rogate for the Cu,brain in CNS drug discovery, a CCSF-to-Cu,braingradient has been suggested for the substrates of efflux trans-porters, such as P-gp, in rodents [11e13,25]. On the other hand, we

Page 8: Drug Metabolism and Pharmacokinetics · In drug discovery, the cerebrospinal fluid (CSF) drug concentration (CCSF) has been used as a surrogate for the interstitial fluid (ISF)

Fig. 5. Comparison of CNS distributions of test compounds between rats and cynomolgus monkeys. The Kp,uu,ISF (A), Kp,uu,CSF (B), Kp,brain (C), and Kp,uu,brain (D) values were comparedbetween wild-type rats and cynomolgus monkeys. Each point represents the mean ± SEM of three to five animals. The rat data were obtained from Table 2, and the data for thecynomolgus monkeys were obtained from our previous study [14]. As the ISF and brain tissues were collected form the brain cortex in the cynomolgus monkey study, the Kp,uu,ISF,Cx

and Kp,uu,brain,Cx values in rats were used for this comparison. 1, antipyrine; 2, carbamazepine; 3, lamotrigine; 4, midazolam; 5, metoclopramide; 6, ondansetron; 7, E2074; 8,risperidone; 9, verapamil; 10, quinidine; 11, desloratadine.

Y. Nagaya et al. / Drug Metabolism and Pharmacokinetics 31 (2016) 57e6664

previously demonstrated that the CCSF-to-CISF gradient was lesspronounced in cynomolgus monkeys and that the CCSF can be usedas a surrogate for the CISF in monkeys, even for P-gp substrates [14].The purpose of the present study was to investigate the utility ofCCSF as a surrogate for CISF in rats by using 12 compounds thatcovered a wide range of P-gp substrate susceptibility.

The CCSF was compared with the CISF for the test compoundsunder steady-state conditions in wild-type rats (Fig. 4). Consistentwith the results in cynomolgus monkeys [14], a CCSF-to-CISFgradient was not overly evident, even for P-gp substrates, and theCISF and CCSF of good P-gp substrates were lower than the Cu,plasma(Table 2). The Kp,uu,CSF values tended to overestimate the Kp,uu,ISF,Cxvalues slightly for good P-gp substrates (eg, desloratadine)(Fig. 4A); however, the differences were within or close to 3-fold,and the trend disappeared when it was compared with theKp,uu,ISF,HIP values (Fig. 4D). To investigate the impact of active effluxby P-gp on the CCSF-to-CISF gradient, the Kp,uu,ISF and Kp,uu,CSF valueswere determined in Mdr1a(�/�) rats. Along with an increase in theKp,uu,ISF values caused by the P-gp defect, the Kp,uu,CSF values of P-gpsubstrates, such as E2074, risperidone, verapamil, and quinidine,also increased in the Mdr1a(�/�) rats, compared with the valuesfor wild-type rats (Table 3). The magnitude of the difference in theKp,uu,CSF values between the wild-type and Mdr1a(�/�) rats wassimilar to that for the Kp,uu,ISF values (Fig. 3). These results sup-ported the idea that the CCSF is generally predictive of the CISF

within or close to a 3-fold difference, even for P-gp substrates inrats. However, caution needs to be exercised when the P-gp-mediated transport activity is similar to or greater than that ofdesloratadine (Kp,uu,CSF < 0.1 in wild-type rats). A high P-gp trans-port activity may cause a high degree of uncertainty in estimatingthe CISF when the CCSF is used as a surrogate. However, this issue isunlikely to be a major point for practical CNS drug discovery set-tings because such compounds could be screened out duringin vitro P-gp substrate liability assays. The present study alsoshowed that the Kp,uu,CSF value of midazolam (1.51) was greaterthan unity, whereas the Kp,uu,ISF values (0.620e0.652) were lowerthan unity. Although the underlying mechanism for these oppositeconcentration gradients across the BBB (CISF < Cu,plasma) and BCSFB(CCSF > Cu,plasma) remains unknown, the results for midazolam mayalso draw attention to the use of CCSF as a surrogate for CISF.

As demonstrated in the present study, the Kp,uu,CSF values of P-gpsubstrates are affected by the defect in P-gp. P-gp is expressed notonly at the BBB, but also at the blood-CSF barrier (BCSFB) in rats.However, P-gp is localized to the cytoplasm or subapical region ofthe choroid plexus epithelial cells [26], and the P-gp proteinexpression level at the rat BCSFB has been reported to be consid-erably lower than that at the BBB [27]. Therefore, our findings andliterature information cumulatively suggest that the role of P-gp, ifany, is negligible at the BCSFB, and that diffusion across the epen-dymal layer followed by P-gp-mediated active efflux at the BBB has

Page 9: Drug Metabolism and Pharmacokinetics · In drug discovery, the cerebrospinal fluid (CSF) drug concentration (CCSF) has been used as a surrogate for the interstitial fluid (ISF)

Y. Nagaya et al. / Drug Metabolism and Pharmacokinetics 31 (2016) 57e66 65

a dominant effect on the CCSF of P-gp substrates in rats, similar to itsrole in cynomolgus monkeys [14]. PK modeling may be a usefulapproach to further support this idea [28].

In the present study, the Kp,uu,brain values were also determinedusing a brain homogenate method (Fig. 4) and were estimatedusing the pH partitioning model [24] (Supplemental Fig. 2). Inparticular, when the Cu,brain was estimated as the product of thein vivo Cbrain and the predicted fu,brain values in brain slices based onthe pH partitioning model, the difference between the Cu,brain andthe CISF increased, along with a reduction in the Kp,uu,brain values(Supplemental Figs. 2A and 2C), and the CCSF-to-Cu,brain gradientwas more evident (Supplemental Figs. 2B and 2D). The underlyingmechanism causing such inconsistency between the Kp,uu,ISF andKp,uu,brain values for compounds that undergo extensive activeefflux at the BBB remains unknown; however, except for suchcompounds, both methods (microdialysis and the product of thein vivo Cbrain and the in vitro fu,brain values) provided similar esti-mates for the Cu,brain, which were predictable from the CCSF in ratswithin a 3-fold difference. Thus, the data presented in thisstudy should be helpful for performing pharmacokinetic-pharmacodynamic analyses during CNS drug discovery.

Interestingly, we found that the impact of Mdr1a knockout onthe Kp,brain values was 3-folde5-fold greater than that on theKp,uu,ISF values for good P-gp substrates (risperidone, verapamil,and quinidine) (Fig. 3). It appears that the difference in themagnitude of the increase caused by the P-gp defect between theKp,uu,ISF and Kp,brain values was pronounced along with in vivo P-gptransport activities. The following possibilities can be considered.The Kp,uu,ISF values of the drugs with high P-gp transport activities,which show very limited entry into the CNS, might be over-estimated in wild-type rats because of the invasive process asso-ciated with microdialysis probe implantation. Assuming thenegligible impact of probe implantation on Kp,uu,ISF values, theinconsistency is attributable to the difference in the distributionvolume in the brain parenchymal cells against ISF. Our preliminaryexperiments confirmed that Mdr1a knockout had no or minimalimpact on binding in brain homogenates. The tissue bindingproperties in intact brain parenchymal cells, including the drugtrapping capacity of lysosomes [24], might be affected in Mdr1a(�/�) rats. Or, P-gp might limit drug entry to the parenchymal cellsfrom ISF compartment since P-gp is reportedly expressed in thebrain parenchyma, but to a much lesser extent compared with theBBB [29]. A comparison of the brain slice uptakes of P-gp substratesbetween wild-type and Mdr1a(�/�) rats may further under-standing of the underlying mechanism.

In drug discovery, rodents are commonly used to evaluate drugCNS penetration in vivo. However, species differences in drug CNSpenetration are frequently debated upon the extrapolation of ro-dent data to primates when the test compounds are P-gp sub-strates. To address this issue, we compared the Kp,uu,ISF, Kp,uu,CSF,Kp,uu,brain, and Kp,brain values obtained from wild-type rats withthose we had previously determined in cynomolgus monkeys [14].Although the Kp,uu,ISF and Kp,uu,CSF values of the P-gp substrates inthe cynomolgus monkeys were similar to or slightly higher thanthose in the rats, the differences were within or close to 3-fold forthe test compounds (Fig. 5A and B). While the Kp,brain values incynomolgus monkeys were more than 3-fold of those obtained inrats for 5 of the 11 test compounds, all the outliers were P-gpsubstrates (Fig. 5C). This finding is consistent with the results of aprevious PET study in which the brain distribution of P-gp sub-strates was greater in large animals than in rodents [30]. Thefu,plasma and fu,brain values could not account for the species differ-ences (Supplemental Fig. 3), and the resulting Kp,uu,brain values inthe cynomolgus monkeys were also more than 3-fold of those inthe rats for 4 of the 11 compounds (Fig. 5D). These results suggest

that the drug distribution from the ISF to the brain parenchymamight be greater in cynomolgus monkeys than in rats for P-gpsubstrates, whereas no remarkable species differences are likely toexist in the contribution of P-gp to drug efflux between rats andcynomolgus monkeys, at least at the BBB. Thus, rats can be prac-tically used to predict Kp,uu,ISF and Kp,uu,CSF values in cynomolgusmonkeys or potentially even in humans to within or close to a 3-fold difference, even for P-gp substrates, because the P-gp proteinexpression levels at the BBB are reportedly similar between cyn-omolgus monkeys and humans [31].

In conclusion, this study directly compared the CCSF with the CISFof 12 compounds that covered a wide range of P-gp substratesusceptibility in rats. The Kp,uu,CSF values were within or close to apractically acceptable threshold: 3-fold of the correspondingKp,uu,ISF values in the brain cortex and hippocampus. Moreover, themagnitude of the increase in the Kp,uu,ISF values of the P-gp sub-strates in Mdr1a(�/�) rats was similar to that for the Kp,uu,CSFvalues. These results suggested that P-gp-mediated active efflux atthe BBB is a major determinant not only for the CISF, but also for theCCSF, and that the CCSF can be used as a surrogate for the CISF even forP-gp substrates in rats.

Conflict of interest

The authors declared no conflict of interest.

Acknowledgments

We thank Dr. Hiroko Komatsu (Eisai Co., Ltd.) for valuabletechnical advice.

Appendix A. Supplementary data

Supplementary data related to this article can be found at http://dx.doi.org/10.1016/j.dmpk.2015.10.003.

References

[1] Smith DA, Di L, Kerns EH. The effect of plasma protein binding on in vivo ef-ficacy: misconceptions in drug discovery. Nat Rev Drug Discov 2010;9:929e39.

[2] Schinkel AH, Wagenaar E, van Deemter L, Mol CA, Borst P. Absence of themdr1a P-Glycoprotein in mice affects tissue distribution and pharmacoki-netics of dexamethasone, digoxin, and cyclosporin A. J Clin Invest 1995;96:1698e705.

[3] Enokizono J, Kusuhara H, Ose A, Schinkel AH, Sugiyama Y. Quantitativeinvestigation of the role of breast cancer resistance protein (Bcrp/Abcg2) inlimiting brain and testis penetration of xenobiotic compounds. Drug MetabDispos 2008;36:995e1002.

[4] Leggas M, Adachi M, Scheffer GL, Sun D, Wielinga P, Du G, et al. Mrp4 confersresistance to topotecan and protects the brain from chemotherapy. Mol CellBiol 2004;24:7612e21.

[5] de Lange ECM, Danhof M, de Boer AG, Breimer DD. Methodological consid-erations of intracerebral microdialysis in pharmacokinetic studies on drugtransport across the blood-brain barrier. Brain Res Rev 1997;25:27e49.

[6] Liu X, Natta KV, Yeo H, Vilenski O, Weller PE, Worboys PD, et al. Unbound drugconcentration in brain homogenate and cerebral spinal fluid at steady state asa surrogate for unbound concentration in brain interstitial fluid. Drug MetabDispos 2009;37:787e93.

[7] Becker S, Liu X. Evaluation of the utility of brain slice methods to study brainpenetration. Drug Metab Dispos 2006;34:855e61.

[8] Shen DD, Artru AA, Adkison KK. Principles and applicability of CSF samplingfor the assessment of CNS drug delivery and pharmacodynamics. Adv DrugDeliv Rev 2004;56:1825e57.

[9] Liu X, Smith BJ, Chen C, Callegari E, Becker SL, Chen X, et al. Evaluation ofcerebrospinal fluid concentration and plasma free concentration as a surro-gate measurement for brain free concentration. Drug Metab Dispos 2006;34:1443e7.

[10] Mariappan TT, Kurawattimath V, Gautam SS, Kulkarni CP, Kallem R, Taskar KS,et al. Estimation of the unbound brain concentration of P-glycoprotein sub-strates or nonsubstrates by a serial cerebrospinal fluid sampling technique inrats. Mol Pharm 2014;11:477e85.

Page 10: Drug Metabolism and Pharmacokinetics · In drug discovery, the cerebrospinal fluid (CSF) drug concentration (CCSF) has been used as a surrogate for the interstitial fluid (ISF)

Y. Nagaya et al. / Drug Metabolism and Pharmacokinetics 31 (2016) 57e6666

[11] Kalvass JC, Maurer TS. Influence of nonspecific brain and plasma binding onCNS exposure: implications for rational drug discovery. Biopharm Drug Dispos2002;23:327e38.

[12] Frid�en M, Winiwarter S, Jerndal G, Bengtsson O, Wan H, Bredberg U, et al.Structure-brain exposure relationship in rat and human using a novel data setof unbound drug concentrations in brain interstitial and cerebrospinal fluids.J Med Chem 2009;52:6233e43.

[13] Kodaira H, Kusuhara H, Fujita T, Ushiki J, Fuse E, Sugiyama Y. Quantitativeevaluation of the impact of active efflux by P-glycoprotein and breast cancerresistance protein at the blood-brain barrier on the predictability of the un-bound concentrations of drugs in the brain using cerebrospinal fluid con-centration as a surrogate. J Pharmacol Exp Ther 2011;339:935e44.

[14] Nagaya Y, Nozaki Y, Kobayashi K, Takenaka O, Nakatani Y, Kusano K, et al.Utility of cerebrospinal fluid drug concentration as a surrogate for unboundbrain concentration in nonhuman primates. Drug Metab Pharmacokinet2014;29:419e26.

[15] Chu X, Zhang Z, Yabut J, Horwitz S, Levorse J, Li X, et al. Characterization ofmultidrug resistance 1a/P-glycoprotein knockout rats generated by zinc fingernucleases. Mol Pharmacol 2012;81:220e7.

[16] Zamek-Gliszczynski MJ, Bedwell DW, Bao JQ, Higgins W. Characterization ofSAGE Mdr1a (P-gp), Bcrp, and Mrp2 knockout rats using loperamide, pacli-taxel, sulfasalazine, and carboxydichlorofluorescein pharmacokinetics. DrugMetab Dispos 2012;40:1825e33.

[17] Liu X, Cheong J, Ding X, Deshmukh G. Use of cassette dosing approach toexamine the effects of P-glycoprotein on the brain and cerebral spinal fluidconcentration in wild-type and P-glycoprotein knockout rats. Drug MetabDispos 2014;42:482e91.

[18] Nagaya Y, Takenaka O, Kusano K, Yoshimura T. Species difference in themechanism of nonlinear pharmacokinetics of E2074, a novel sodium channelinhibitor, in rats, dogs, and monkeys. Drug Metab Dispos 2013;41:1004e11.

[19] Paxinos G, Watson C. The rat brain in stereotaxic coordinates. 6th ed. London:Academic Press; 2007.

[20] Bentz J, O'Connor MP, Bednarczyk D, Coleman J, Lee C, Palm J, et al. Variabilityin P-glycoprotein inhibitory potency (IC₅₀) using various in vitro experimentalsystems: implications for universal digoxin drug-drug interaction riskassessment decision criteria. Drug Metab Dispos 2013;41:1347e66.

[21] Sugimoto H, Hirabayashi H, Kimura Y, Furuta A, Amano N, Moriwaki T.Quantitative investigation of the impact of P-glycoprotein inhibition on drugtransport across blood-brain barrier in rats. Drug Metab Dispos 2011;39:8e14.

[22] Doran A, Obach RS, Smith BJ, Hosea NA, Becker S, Callegari E, et al. The impactof P-glycoprotein on the disposition of drugs targeted for indications of thecentral nervous system: evaluation using the MDR1A/1B knockout mousemodel. Drug Metab Dispos 2005;33:165e74.

[23] Maurer TS, DeBartolo DB, Tess DA, Scott DO. Relationship between exposureand nonspecific binding of thirty-three central nervous system drugs in mice.Drug Metab Dispos 2005;33:175e81.

[24] Frid�en M, Bergstr€om F, Wan H, Rehngren M, Ahlin G, Hammarlund-Udenaes M, et al. Measurement of unbound drug exposure in brain:modeling of pH partitioning explains diverging results between the brainslice and brain homogenate methods. Drug Metab Dispos 2011;39:353e62.

[25] Venkatakrishnan K, Tseng E, Nelson FR, Rollema H, French JL, Kaplan IV, et al.Central nervous system pharmacokinetics of the Mdr1 P-glycoprotein sub-strate CP-615,003: intersite differences and implications for human receptoroccupancy projections from cerebrospinal fluid exposures. Drug Metab Dispos2007;35:1341e9.

[26] Rao VV, Dahlheimer JL, Bardgett ME, Snyder AZ, Finch RA, Sartorelli A, et al.Choroid plexus epithelial expression of MDR1 P glycoprotein and multidrugresistance-associated protein contribute to the blood-cerebrospinal-fluiddrug-permeability barrier. Proc Natl Acad Sci U S A 1999;96:3900e5.

[27] Gazzin S, Strazielle N, Schmitt C, Fevre-Mntange M, Ostrow JD, Tiribelli C, et al.Differential expression of the multidrug resistance-related proteins ABCb1and ABCc1 between blood-brain interfaces. J Comp Neurol 2008;510:497e507.

[28] Kodaira H, Kusuhara H, Fuse E, Ushiki J, Sugiyama Y. Quantitative investigationof the brain-to-cerebrospinal fluid unbound drug concentration ratio understeady-state conditions in rats using a pharmacokinetic model and scalingfactors for active efflux transporters. Drug Metab Dispos 2014;42:983e9.

[29] Seegers U, Potschka H, L€oscher W. Expression of the multidrug transporter P-glycoprotein in brain capillary endothelial cells and brain parenchyma ofamygdala-kindled rats. Epilepsia 2002;43:675e84.

[30] Syv€anen S, Lindhe €O, Palner M, Krnum BR, Rahman O, Långstr€om B, et al.Species difference in blood-brain barrier transport of three positron emissiontomography radioligands with emphasis on P-glycoprotein transport. DrugMetab Dispos 2009;37:635e43.

[31] Hoshi Y, Uchida Y, Tachikawa M, Inoue T, Ohtsuki S, Terasaki T. Quantitativeatlas of blood-brain barrier transporters, receptors, and tight junction proteinsin rats and common marmoset. J Pharm Sci 2013;102:3343e55.