You are on page 1of 14

J Pharmacokinet Pharmacodyn (2013) 40:301–314

DOI 10.1007/s10928-013-9303-7

REVIEW PAPER

In vitro, in vivo and in silico models of drug distribution


into the brain
Scott G. Summerfield • Kelly C. Dong

Received: 11 November 2012 / Accepted: 31 January 2013 / Published online: 13 February 2013
Ó Springer Science+Business Media New York 2013

Abstract Achieving sufficient brain penetration to elicit virtually eliminates paracellular diffusion in the healthy
efficacy in humans is one of the most challenging tasks for brain [1]. Expressed within the BBB are also a range of
scientists in CNS Drug Discovery. Substantial progress has transport systems whose role is to either supply CNS tissue
been made in the past decade in understanding the factors with the necessary nutrients for maintenance of brain
influencing the rate and extent of brain distribution via a homeostasis or to act as gatekeepers and expel potentially
variety of in vivo, in vitro and in silico methodologies, and undesirable xenobiotics [2]. Therefore for drugs to traverse
hence, predict their likelihood of success in man. This the BBB effectively they must pass either by means of
purpose of this review is to summarize the current transcellular diffusion or be recognised by an uptake
approaches with a special focus on parameters related to transporter.
free drug concentrations in brain which are the most Designing in the necessary properties to meet adequate
pharmacologically relevant for the majority of CNS disease potency, pharmacokinetics and also facilitate distribution
targets. Due to the dynamic and complex nature of this into the brain is an exceptionally challenging task. Attrition
targeted organ, it is inevitable that these approaches have of potential CNS agents during Drug Development is
not been able to provide a fully comprehensive assessment among the highest of all common therapeutic areas [3]. As
of brain distribution and are expected to evolve further in noted recently by Morgan et al. [4], the chances of making
the years to come. the right decision to progress or terminate a clinical drug
candidate are enhanced when one can demonstrate the
Keywords Brain  Distribution  In vivo  In vitro  presence of drug in the biophase, have the verified target
In situ  In silico  PET engagement and that this leads to the expected pharma-
cology. Drug metabolism and pharmacokinetics (DMPK)
has a role to play in all three of these ‘pillars’ although the
Introduction aim of this article is to focus on the first, namely having
techniques to show that the drug is distributed into the
Passage across the blood-brain barrier (BBB) and distri- brain. Indeed this is the necessary precursor to the other
bution into brain parenchyma are key steps for drugs acting pillars being met.
on receptors within the central nervous system (CNS). The Figure 1 shows a schematic representation of the inter-
BBB comprises a tight junctioned cellular structure that relationship between the blood and brain compartments
following administration of a potential CNS agent. Drug in
blood is in equilibration between the unbound fraction and
S. G. Summerfield (&)
GlaxoSmithKine R&D, Sir David Jack Research Centre, the fraction bound to plasma proteins and blood cell con-
Ware SG12 0DP, Hertfordshire, United Kingdom stituents. The unbound portion is available to undergo
e-mail: scott.g.summerfield@gsk.com distribution into tissues or undergo elimination from the
body, e.g. via hepatic and renal clearance. At the level of
K. C. Dong
GlaxoSmithKine R&D China, Zhangjiang Hi-tech Park, the brain, if the drug is sufficiently lipophilic then the
Shanghai 201203, China unbound portion will be able to dissolve into the cellular

123
302 J Pharmacokinet Pharmacodyn (2013) 40:301–314

environment of the BBB and pass through to the brain the more recent in vitro and in silico developments have
extracellular fluid (ECF) by means of passive transcellular been centred on unbound brain parameters, which have
diffusion. Unbound drug in the blood may also interact helped shift the focus in Drug Discovery away from
with transporters at the BBB surface to facilitate their measurements based on total concentrations that are less
passage [5, 6]. Once distributed into brain ECF then relevant with respect to distribution mechanisms and
unbound drug may distribute further into the brain paren- pharmacological activity. This review aims to cover the
chyma, either binding to the target, other receptors or non- key elements of total and unbound measurements and
specifically to surrounding lipophilic brain tissue. A variety where they make the greatest impact on the discovery
of measures of brain distribution can be taken either in effort for new CNS medicines.
terms of concentration (unbound or total), as a partition
ratio between brain and blood or plasma (unbound or total)
or as a clearance across the BBB (unbound). There are also In vivo methodologies
in vitro or in silico equivalents available to Drug Discovery
scientists for all of these measurements. The unbound Unbound concentration measurements
fraction in blood (or plasma) may be determined by means
of equilibrium dialysis, ultrafiltration or microdialysis [7]. Brain microdialysis allows for the temporal monitoring of
Similarly the unbound brain fraction may be estimated via brain ECF concentrations in concert with the blood or
equilibrium dialysis of brain homogenate [8, 9], brain tis- plasma pharmacokinetics [13, 14] and can also be used to
sue slices [10, 11], cerebrospinal fluid (CSF) [12] or brain track changes in neuro biomarkers [20]. Taking measure-
microdialysis [13, 14]. The rate of drug passage across the ments from multiple compartments enables all of the
BBB may be measured directly using in situ brain perfu- ensuing information to be woven together through physi-
sion [5] or in vitro using cell-based methods [15] or arti- ologically based pharmacokinetic (PBPK) modelling at the
ficial membranes [16, 17]. Concentrations within the brain level of the brain [21, 22]. However, this technique requires
can be also measured either as a total value (e.g. the sum of the most surgical skill of all the platforms of in vivo
the unbound and bound fractions) by homogenising excised methods available. A dialysis probe is inserted stereotaxi-
brain tissue, or alternatively, unbound by means of brain cally into the brain after exposing it through a hole in the
microdialysis [13, 14]. Few techniques have the capability cranium. Dialysis fluid is pumped slowly through the probe
to capture most or all of these parameters within a single whereupon it meets the brain ECF across a semi-permeable
experiment. The closest are in vivo microdialysis and membrane at the probe tip. The composition of this dialysis
temporal imaging techniques like positron emission fluid must match that of ECF in order to maintain the
tomography (PET) which provide significant benefits in osmotic balance and homeostasis within the brain.
this regard [18, 19]. Unbound drug diffuses across this membrane and flows
Figure 1 also demonstrates the importance of the back along the probe tubing where it is collected continu-
unbound concentration in determining the passage and ously into sampling vials. Tracking the brain kinetics of
distribution of drugs to and within the brain, which aligns drugs and their metabolites has been employed to under-
well to the fact that is also the fraction available to bathe stand efficacy discrepancies for agents such as clozapine
receptors in the appropriate cellular compartment. Many of and morphine [23, 24]. Following on the work with brain

Fig. 1 Schematic of drug distribution into the brain from the blood blood-brain barrier (BBB) and blood-CSF barrier (BCSFB). Drug
compartment. Ctotal and Cu denote the total and unbound concentra- distribution is depicted by the total concentration ratio Kp and
tions in blood. Concentrations in the brain are shown in extracellular unbound ratio Kp,uu and the rate of passage across the BBB as Kin
fluid (ECF) and cerebrospinal fluid (CSF). Dotted lines show the

123
J Pharmacokinet Pharmacodyn (2013) 40:301–314 303

microdialysis, Gupta et al. [25] introduced the concept of via serial sampling in the same animal whereas it is not
Kp,uu or the unbound brain-to-unbound plasma concentra- possible when total tissue concentrations are measured and
tion ratio. In essence, Kp,uu measures the unbound con- then corrected to unbound values because brain excision is
centration gradient across the BBB, which would equal a terminal procedure. This means numerous additional
unity for drugs undergoing passive transfer. Similarly, a animals are required to create a time course but often this
Kp,uu value less than unity would suggest passage across compromise is paid for the simplicity of the approach.
the BBB is restricted by either inherently low permeability The accuracy of the total concentration measurement
(e.g. atenolol arising from its physicochemical properties) relies on having complete tissue extraction and no inter-
or by active efflux through a transporter, such as P-glyco- ferences during the bioanalysis but there is actually little
protein (P-gp). A Kp,uu value greater than one would information available to support how well drugs are
indicate that uptake into the brain is facilitated in some extracted from brain tissue. For bioanalytical techniques
way, perhaps via active influx. such as liquid chromatography–tandem mass spectrometry
In addition to the severe surgery associated with inser- (LC–MS/MS) calibration standards and quality control
tion of a microdialysis probe, the resulting brain ECF is samples are typically prepared by spiking analyte into brain
highly diluted within the dialysate flow. This dilution effect homogenate whereas from the in vivo experiment, the drug
and the absence of protein or lipid within the dialysate resides within the parenchyma and requires efficient
often leads to adsorptive losses of the analyte over the extraction in order to reflect the associated standards.
internal surfaces of the probe and tubing, thus necessitating Supporting information for the efficient drug extraction
calibration methods to compensate for these losses [26] could come from comparing fubrain values derived from
together with highly sensitive bioanalytical measurements. spiking drug into brain homogenate versus drug dosed
Taken together with the low throughput and high probe in vivo and then brain tissue subsequently homogenised
costs, these experimental challenges restrict the utility of and being subjected to dialysis (Table 1). The close con-
brain microdialysis to more bespoke studies rather than cordance between the in vitro and ex vivo measurements
being deployed as a general screening tool. However, it is suggests that the combination of tissue homogenisation and
clear that a great deal of key information on brain dispo- solvent extraction is effective at releasing the drug.
sition is attainable if the effort is put into running these
experiments. Total concentrations measurements
Sampling CSF has been proposed as a means of taking a
surrogate unbound concentration measurement for brain The ease of measuring total brain concentrations has led to
ECF [12]. CSF is produced continuously by the choroid brain distribution often being quoted in terms of the par-
plexus and cushions the cortex from severe head move- tition ratio (Kp), which is defined as the total concentration
ments and in rats the CSF volume is ca. 300 uL and flows in brain divided by the total concentration in blood (or
at ca. 2 uL/min [27, 28]. For compounds where BBB dif- plasma). It is now generally accepted that Kp values rep-
fusion is both rapid and passive then CSF concentrations resent a rather blunt instrument for interpreting whether a
agree well with brain ECF concentrations [12]. However compound is ‘good’ or ‘poor’ in terms of brain penetration
there are instances where CSF and ECF brain concentra- or indeed efficacy [27] but can be informative with in vivo
tions do not correlate well such as when the drug is subject models to characterise active transport at the level of the
to a slow rate of uptake across the BBB, active transport or BBB [31, 32]. In these models the drug is administered to
local metabolism [28–30]. Generally, CSF concentrations naive rats or mice and then again to animals where the
appear to be a better surrogate of unbound brain concen- transporter of interest (generally P-gp) has been inactivated
trations than using unbound plasma concentrations [12]. either chemically [33, 34] or genetically [31, 32]. Kalvass
Indirect determination of unbound brain concentrations and Maurer [8] showed that the unbound brain fractions of
may be achieved by first measuring the total brain tissue several P-gp substrates were identical in wild-type and
concentration and correcting this with the in vitro unbound P-gp knockout mice [31] and hence the subsequent changes
brain fraction [8]. Despite the need for two measurements, in Kp are the result of altering BBB transport (e.g. a rate
this approach is far more amenable to screening and tri- effect) rather than altering binding (e.g. an extent effect).
aging the greater numbers of compounds within the Drug This leads to the establishment of an unbound concentra-
Discovery setting. Excision of brain tissue and the sub- tion gradient reflected in Kp,uu. Murine gene knockouts of
sequent homogenisation can be mastered by a wider range other putative BBB transporters are also available for the
of scientists and technicians than the surgical expertise study of brain distribution in selective cases [35–41] and
required for microdialysis. In addition the unbound fraction the changes observed are far lower (generally less than
methods are amenable to moderate automated throughput. two-fold) than those noted for P-gp substrates in the mdr
However, microdialysis can provide temporal information 1a/b(-/-) model. Many P-gp substrates are lipophilic

123
304 J Pharmacokinet Pharmacodyn (2013) 40:301–314

Table 1 Unbound brain fractions measurements taken from in vitro studies (control brain homogenate spiked with analyte) and in vivo studies
(iv administration followed by brain excision and homogenisation)
Unbound brain (% free)
Cpd 1 Cpd 2 Cpd 3 Acetaminophen

Rat Aa 5.8 ± 0.6 2.1 ± 0.3 0.080 ± 0.016 68.5 ± 18.5


Rat Ba 4.8 ± 0.4 2.0 ± 0.3 0.080 ± 0.044 75.4 ± 26.8
Rat Ca 4.2 ± 0.4 2.1 ± 0.3 0.089 ± 0.054 85.1 ± 21.1
Spiked brain (fresh tissue)b 4.8 ± 1.0 2.0 ± 0.1 0.075 ± 0.008 72.0 ± 5.0
Spiked brain (frozen tissue)b 4.6 ± 0.7 2.1 ± 0.1 NT NT
NT not tested
a
Drug administered by intravenous infusion with brain excision, homogenisation and extraction performed according to Ref. [9]
b
Drug spiked into control brain tissue at a concentration of 1 ug/mL [9]

bases and so would be expected to undergo extensive tissue understanding brain distribution based on the role of
distribution when freely permeable across the BBB of the unbound drug. Species differences in Kp could be shown to
knockout strain. In contrast, the physicochemical proper- arise from changes to blood/plasma binding [9] and today
ties of substrates for several of these BBB transporters may this might also be seen with the measurements of Kp,uu if
limit their inherent brain distribution, which may help to active transport is implicated. Subsequently, a collabora-
explain the small increases observed. In higher species tive effort was initiated with a PET imaging centre and a
information is far rarer on BBB transporter differences series of pig PET biodistribution studies were performed
across breeds and hence chemical knockout is generally and complemented with in vitro plasma and brain tissue
employed as an adjunct to imaging techniques such as PET binding data. Generally in vitro estimates of brain distri-
[42, 43]. One example of P-gp deficiency has been iden- bution (e.g. fuplasma/fubrain) were comparable to in vivo
tified in Collie dogs [44] although it is unclear whether any estimates from the tracer experiments indicating passive
advantage is conferred over the chemical intervention. diffusion across the BBB for many of the compounds
Aside from these instances, terminal brain penetration tested [45]. Loperamide, a known substrate for P-gp, was
measurements in higher species are performed less fre- characterised by a low in vivo Kp value in the pig but
quently because these species are often reused in pharma- showed good agreement with the in vitro Kp measurement
cokinetic studies and are employed less commonly for when co-administered with cyclosporine due to inhibition
pharmacology models of CNS conditions. Before much of of the P-gp effect.
the work cited here was published, our own company, like Total brain concentration measurements do not neces-
many others, was focusing on Kp measurements. Cross sarily just provide information on the extent of brain pen-
species information on Kp was thought to be useful since etration; they can also be used to derive information on the
the entire clinical premise was based on the brain pene- rate of drug passage across the BBB when used in con-
tration in rodents. So a series of brain penetration experi- junction with in situ brain perfusion studies [5, 46, 47].
ments were performed in pigs because of their use in PET Generally performed in rodents, in situ brain perfusion
biodistribution studies and pharmacokinetic models for involves the administration of drug via rapid perfusion into
scaling. Following 1 h iv infusions, the brains were excised the carotid artery (\1 min at a flow of 5–10 mL/min) to
and several brain regions were dissected out in order to surgically prepared animals. The brain is excised and the
measure the total concentrations (Table 2). Overall the total brain tissue concentrations are determined by means
regional Kp values were very close and no substantial of radioactivity [5] or LC–MS/MS [47]. Under this short
differences in brain regions were noted. Essentially the data perfusion paradigm drug transfer across the BBB is con-
showed that in most instances the Kp in rats and pigs were sidered to be the dominant process while back-flux of drug
different but it was not clear why. Compound A had also to the vascular compartment is negligible under these
been identified as a P-gp substrate and Kp was measured in conditions [5]. In order to benchmark compounds across
several other species (Table 3). Although the brain pene- experiments several control compounds are generally
tration was marginally higher in rats, generally the data included and this has provided an excellent and reliable
across species added nothing more. The advent of brain data set across studies reported in the literature. A non
tissue binding screens and an explicit description for the brain penetrant agent is added to the perfusion solution in
roles of plasma and tissue binding by Kalvass and Maurer order to ascertain and correct for the vascular volume (e.g.
14
[8] paved the way for a more directed approach toward C sucrose or atenolol). Antipyrine is often used as a

123
J Pharmacokinet Pharmacodyn (2013) 40:301–314 305

Table 2 Brain distribution for 14 compounds measured in terms of Kp in rodents (whole brains) and pigs (regional values)
Cpd Rat Pig brain Kbp
Kap
Mean Dorsal Cingulate and Hippocampus Caudate Nucleus Hypothalamus Cerebellum Rest Cervical
lateral infralimbic putamen accumbens of spinal
cortex cortex brain cord

A 0.28 0.05 0.04 0.04 0.05 0.05 0.05 0.05 0.06 0.05 0.04
B 1.53 4.16 4.56 4.53 4.06 4.14 4.29 4.17 3.63 3.93 4.06
C 0.50 1.07 0.94 1.12 1.24 0.86 1.22 1.08 1.09 1.01 1.18
D 0.20 0.51 0.57 0.51 0.50 0.50 0.47 0.50 0.54 0.50 0.27
E 5.00 11.01 13.52 12.23 11.45 9.70 10.77 10.01 9.95 10.46 6.54
F 3.25 4.50 4.92 4.39 4.29 4.24 4.47 4.56 4.52 4.58 2.01
G 3.16 6.80 6.72 6.66 6.96 6.32 6.70 6.94 6.77 7.30 4.43
H 0.14 2.04 2.36 1.99 1.69 2.35 2.10 1.69 2.41 1.74 2.08
I 0.66 1.96 2.04 1.81 1.94 1.94 1.91 2.03 1.94 2.07 2.17
J 0.80 1.37 1.34 1.35 1.38 1.44 1.42 1.30 1.48 1.25 1.45
K 1.53 5.57 6.27 5.65 5.80 5.56 5.62 5.68 4.51 5.51 3.74
L 3.45 14.49 16.13 15.83 14.00 14.13 14.47 14.02 12.80 14.54 8.96
M 0.80 0.91 0.68 1.15 1.08 0.70 1.02 0.64 1.00 1.04 0.87
N 0.50 0.29 0.34 0.29 0.29 0.21 0.25 0.26 0.34 0.31 0.21
a
Steady-state iv infusion. A dose of 1 mg/kg was administered in all cases
b
1 h iv infusion. A dose of 1 mg/kg was administered in all cases

Table 3 Brain distribution for three compounds measured in terms of When comparing Kin or PS values for different
Kp across a range of preclinical species (no significant regional
compounds it is important to remember that these
differences)
parameters focus on the rate of uptake and provide no
Cpd Species Kap information on the extent of distribution. This is
Rat Minipig Landrace pig Monkey Dog Marmoset exemplified in Fig. 2a, b which show Kin [47] plotted in
the order of increasing Kp (steady-state) and Kp,uu values.
A 0.28 0.05 0.06 0.05 0.11 0.05
Correlations in both instances are poor because Kp and
B 1.53 4.16 1.73 1.07 3.05 3.80
Kp,uu are partition ratios and thus provide no detail on the
C 0.5 1.07 0.68 0.40 1.02 0.64
rate of drug uptake into the brain. Extent and rate
a
1 h iv infusions for all species except rat where an iv infusion to information are orthogonal parameters and are both
steady-state was performed. A dose of 1 mg/kg was administered in required to truly appreciate brain distribution, particularly
all cases
for PB-PK approaches [48]. Figure 3a, b show the use of
PBPK modelling to elucidate the interplay between PS and
moderately BBB permeable marker while diazepam is brain tissue distribution for clozapine and acetaminophen
employed as a measure of perfusion limited uptake. following iv infusion to steady-state and the measurement
Knowing the dose concentration (Cpf mg/mL) vascular of brain ECF via microdialysis. Blood kinetics, BBB
volume correction (Vi mg/g brain) and brain concentration uptake and fubrain were measured experimentally [47] and
(Cbr mg/g brain) at time t enables the intrinsic uptake fitted to a PBPK model assuming rapid distribution
(Kin mL/g brain/min) to be calculated. between brain ECF and tissue. Predicted ECF
concentrations for both clozapine and acetaminophen lie
Cbr ðtÞ
 Vi ¼ Kin t within two-fold of the measured values highlighting the
Cpf ðtÞ utility of this predictive approach. The respective Kp, PS
Kin can be viewed as a clearance and may be and fubrain values for clozapine (23, 1250 mL/h/kg and
transformed to a permeability surface product (PS) using 0.01) and acetaminophen (0.67, 25 mL/h/kg and 0.72) were
the Crone–Renkin equation, where F represents flow (e.g. also very different but both attained steady-state ECF
the Kin of diazepam) [5]. concentrations in a similar time window of 2 and 4 h. An
  important observation is the concept of the intrinsic brain
Kin half-life, which has been defined as the time taken to reach
PS ¼ F  ln 1 
F 50 % of the steady-state Kp, and is inversely proportional

123
306 J Pharmacokinet Pharmacodyn (2013) 40:301–314

to the hybrid term PSfubrain [48, 49]. If the product of PS information is also available from post-mortem dissections
and fubrain is comparable between two compounds then where brain tissue concentrations has been collected for
their time to attain brain equilibrium will be similar even if toxicology purposes [51]. On the limited data available
they possess broadly different physicochemical properties, human brain distribution and in vitro brain tissue binding
as is the case for clozapine and acetaminophen showed in measurements have shown reasonable correlations and
Fig. 3 but also for other compounds where the observation supports the view that passive permeability across the BBB
was made previously but the conection between PS and is important, at least for the compounds tested thus far [52].
fubrain had not been established [50]. Notably, there is a contention that passive permeability is
All of this work is only really performed to better the exception rather than the norm as stated by Kell et al.
understand the likely brain distribution of drug candidates [53]. However, this argument neglects to consider the
in humans. In vivo measurements in humans are largely influence of tissue binding as and the argument proposed
achieved through imaging studies (e.g. PET) but some by Kell et al. has also been refuted in a recent repost [54].

Fig. 2 Kin derived from Ref. a


[47]. Compounds are ordered
left to right in terms of
a increasing Kp and b increasing
Kp,uu (demonstrating no
correlation of Kin to Kp or Kp,uu)

123
J Pharmacokinet Pharmacodyn (2013) 40:301–314 307

a 0.14 tolerance, low cost and flexibility in adapting the lipid


membrane composition, the parallel artificial permeability
0.12
assay (PAMPA) has demonstrated reasonable correlations
0.10 to other cell-based models and in vivo evaluations in
rodents. By adapting a porcine polar brain lipid to mimic
ECF (uM)

0.08
the BBB membrane characteristics, PAMPA has also been
0.06
reported to display good correlations to in situ brain per-
0.04 fusion measurements [16]. This technique has been utilized
0.02
to provide early classification of test compounds into those
likely to be CNS penetrant and non-CNS penetrant, based
0.00
0 1 2 3 4 5 6 on permeability. However, because of the lack of active
Time (h) transporter expression or metabolic enzymatic activity,
b 7.0
PAMPA is limited in its application to generating the
passive permeability component of BBB uptake [6]. Sim-
6.0
ilarly, immobilized artificial membrane (IAM) column,
5.0 which contains stationary phase consisting of lipid mono-
ECF (uM)

4.0 layer, allows drug partitioning during chromatographic


3.0
separation. The greater retention of the column reflects
higher passive permeability although it suffers again from
2.0
the same limitation in mimicking only passive diffusion
1.0 [17].
0.0
0 1 2 3 4 5 6
Cell based in vitro methods
Time (h)

Fig. 3 Concentration-time profile of brain ECF following iv infusion Cell-based in vitro approaches to measuring permeability
to steady-state for a clozapine and b acetaminophen. Measured brain provide a more physiologically relevant screening para-
ECF concentrations (filled square) and predicted brain ECF concen-
digm over PAMPA and IAM columns. The experimental
trations (open square) are shown. Clozapine is characterised by PS
and fubrain values of 1250 mL/h/kg and 0.01, respectively. Acetami- design comprises two compartments that make contact
nophen is characterised PS and fubrain values of 25 mL/h/kg and 0.72, through a confluent cell membrane that displays a tight-
respectively. Despite significant differences in the absolute values of junction morphology. Drug in physiological buffer is
PS and fubrain between clozapine and acetaminophen the hybrid term
placed in the donor compartment and allowed to diffuse
PSfubrain are comparable, leading to comparable brain equilibration
half-lives in vivo [48, 49] across the cell layer for a given time period (t) after which
the concentration is determined from both the donor and
In summary, there are a range of in vivo techniques receiver compartments. If the cell boundary is polarised,
available to characterise the various aspects of the rate and such as by the orientation of a transport protein then the
extent of brain distribution and the ensuing information is flux across that cell line must be measured in both direc-
amenable to providing accurate and predictive PBPK tions. The apparent permeability (Papp) is the calculated as
models. However, the flip side of performing in vivo shown below:
experiment is the time required for the surgical procedures, dCr Vr
particularly for brain microdialysis and in situ brain per- Papp ¼ 
dt A  Cr ð0Þ
fusion. This has continued to drive the development of
in vitro based approaches with a view to initially triaging where Cr, Vr and A denote the receiver drug concentration,
the extensive number of molecules being synthesised dur- volume of the receiver compartment and the area of the cell
ing target validation and lead optimisation. membrane. Correcting for losses and adsorption can be also
accommodated through the use of Pexact values [55].
Caco-2 cells are a well established in vitro tool for
In vitro methodologies assessing intestinal absorption. The cell line derived from
human epithelial colorectal adenocarcinoma cells has been
Non-cell based in vitro models adapted to evaluate BBB permeability [56]. Due to its non-
brain originated epithelial nature, several studies suggested
Early in vitro screening in Drug Discovery is mainly based the limited utility of Caco-2 for assessing CNS penetration
on the use of techniques that act as a surrogate measure for [57]. Following this, the Madin–Darby Canine Kidney
transfer across the BBB. With high throughput, pH (MDCK) cell line has been widely used as an in vitro

123
308 J Pharmacokinet Pharmacodyn (2013) 40:301–314

permeability model due to the tighter monolayer junctions In vitro assessment for CNS delivery (extent and brain
achievable and the relatively short cell culture time distribution)
[15, 58]. Despite its non-human cerebral and epithelial
origin, MDCK has been stably transfected by a retrovirus It is widely accepted that unbound drug concentrations are
carrying the multi drug resistance (MDR1) gene in order to more relevant to understanding pharmacodynamic efficacy
incorporate P-gp, which is the most prolific efflux trans- because this is the portion of drug available to bathe the
porter of potential CNS drugs in the BBB. In a comparative receptor [74, 75]. The extent of drug biodistribution into
study of several different P-gp assay formats the MDR1– the brain has been the subject of increasing attention and is
MDCK assays faired favourably against the comparators largely governed by binding to blood constituents and to
[59]. Furthermore this study also showed a strong corre- brain tissue [76]. For passively permeable compounds good
lation between the in vitro efflux ratio between MDCK cell in vivo–in vitro correlations have been established in
lines transfected with the rodent mdr1a gene and human rodents [30], pigs [45] and humans [52]. This is because
MDR1 gene. Although P-gp is one of the most important Kp,uu is equal to unity for passively permeable compounds
efflux transporters and its role is vital in restricting the and hence Kp is corrected by the relative tissue binding as
brain penetration of many exogenous substances, active shown below:
   
transport of drugs from the brain is not limited to this efflux Cubrain Cbrain fubrain
Kp;uu ¼ ¼ 
pump alone [60–62]. BCRP, another predominant efflux Cubloodorplasma  Cblood or plasma fublood or plasma
protein in human brain, has been the subject of emerging fubrain
attention although its impact remains controversial [63]. ¼ Kp  :
fublood or plasma
This transporter plays a dominant role in limiting sorafenib
transport to the brain in vivo, which was clearly demon- Unbound fractions should not be used by themselves to
strated using the BCRP gene stably expressed into the guide the selection of compounds within Drug Discovery
MDCK cell line [64]. There is no doubt that significant because they merely represent the proportion of available
advantages are there for in vitro cell lines that express drug but not the actual unbound concentration. The
multiple active transporters in order to provide a more unbound fraction in blood, plasma or brain should be
comprehensive and cost effective evaluation of efflux lia- integrated and interpreted with total concentrations or
bilities. Recently, Poller et al. reported a study using a in vivo exposures [77]. Generally unbound brain fractions
double-transduced, polarized MDCKII cell line expressing are generated by means of tissue homogenates but evidently
both human P-gp and BCRP in order to assess the possible the entire cellular structure of the brain parenchyma is lost
interplay between these two efflux proteins for drug during this preparation [74, 76]. An alternative approach is
transportation across BBB [65]. to employ fresh in vitro brain slices, which have been shown
The next tier of in vitro permeability models involve to retain at least some transporter activity as well as cell
the use of BBB cell cultures and originated in the 1990s morphology [11]. A thin slice of brain is bathed in
[66, 67]. Primary cultured brain endothelial cells have been simulated ECF containing the drug after which the tissue
isolated and reported from several species including slice is homogenised and analysed by means of LC–MS/MS
bovine, porcine, rat and mouse brain [68–71]. In a com- analysis. This method yields an unbound brain volume of
parative study, a bovine brain microvessel endothelial cell distribution (Vu,brain) for the drug (as opposed to fubrain) that
model demonstrated a superior correlation to in vivo brain is derived from the following equation shown below where
microdialysis results relative to Caco-2 and MDCK/MDR1 Abrain denotes the amount of drug in brain slice and Cubrain
data for a series of nine reference compounds after exclu- is the concentration of free drug in the incubation solution:
sion of P-gp substrates [56]. A tantalising refinement of this Abrain
approach involves the use of human microvascular endo- Vu;brain ¼
Cubrain
thelial cells co-cultured with various concoctions of
astrocytes, glial cells and pericytes [72, 73]. The predictive The advantage of the brain slice method lies in the fact
value of humanized in vitro models has been demon- that cellular barriers, cellular pH gradients and also efflux/
strated in combination with clinical (PET) [57]. However, influx transporters are retained in addition to tissue binding
despite the intensive effort in this field, the majority of component [76]. As Vu,brain is an apparent volume term, it
these immortalized brain endothelial culture models reflects the volume of distribution into the brain tissue
struggle to achieve a sufficient tight junction morphology relative to the ECF volume (0.2 mL/g) [11, 76]. Values
and to maintain a full complement of transporter func- close to 0.2 mL/g indicate little or partial distribution into
tion or enzymatic activity that really make them most the brain parenchyma whereas progressively higher values
appealing. (e.g. [0.8 mL/g) indicate substantial nonspecific binding

123
J Pharmacokinet Pharmacodyn (2013) 40:301–314 309

into brain tissue. The use of brain tissue slices is more present in slices but not homogenised tissue. In this latter
involved than the preparation of homogenates but it is clear work Frı́den et al. [76] proposed a correction for fubrain to
that slices offer a closer resemblance to the brain in vivo account for these differences, but inevitably, in silico
and is likely to introduce fewer artefacts as a result [11]. approaches based on brain slice data would be the best
option.
Estimating PS values in silico has been facilitated by the
In silico methodologies widespread use of control compounds when conducting
these experiments. Once again lipophilicity is an important
In silico models are available for the estimation of several component of the model but other physiochemical factors
brain-related parameters including PS [78, 79], Kp (e.g. modulate this. Early literature reports from in situ brain
expressed in logarithmic form and denoted as logBB) perfusion studies had showed a strong correlation between
[80, 81] fubrain [82], Kp,uu [83] and P-gp efflux [84, 85]. increasing BBB uptake and increasing logP [5]. However,
Evidently the value of these tools is for early Drug Dis- the range of compounds tested were characterised by logP
covery where there is often a need to triage a large numbers values \1 and were not typical of the chemical space
of compounds. Of these in silico tools there is substantial generally occupied by CNS drugs [47]. In addition BBB
literature for logBB, which continues to appear despite an uptake cannot increase indefinitely and would become
accumulating body of evidence to suggest that this limited ultimately by the perfusion flow rate [6, 87]. Fig-
parameter is of little value in selecting or triaging com- ure 2 shows that many common marketed CNS drugs are
pounds [27, 75]. In addition there is a general miscon- characterised by comparable Kin values to that of diaze-
ception that logBB is related to BBB permeability [80, 81] pam, the common marker of perfusion limited uptake.
whereas it strictly refers to partitioning. Kp and its in silico Indeed some agents such as sertraline and maprotiline are
counterpart logBB are complex hybrid parameters because characterised by Kin values much greater than would be
they incorporate the relative extents of tissue binding to expected from the traditional diazepam standard [7, 47, 87].
brain and blood (or plasma) plus the concentration gradient Active transport also influences BBB uptake and the
across the BBB caused by active transport. Pharmacolog- number of examples collated from in situ perfusion studies
ical effect is not driven by the total concentration as has increased markedly [5, 6]. This strikes at the heart of
unbound drug is the portion that is available to bind to the many in silico models for estimating brain distribution.
targeted receptors [27]. An excellent example of this was Essentially any global model has to contend with the
cited by Watson et al. [77] where the in vivo D2 receptor structure activity relationships (SAR) of all BBB trans-
occupancy in rats was best correlated to Cubrain/Ki(D2) or porter interactions with drug molecules. This is highly
more explicitly the unbound brain concentration relative to problematic because the SAR associated with the substrate
the drug’s in vitro potency at the D2 receptor. Risperidone specificity for transporting gabapentin [88] or other sys-
was the least brain penetrant in terms of Kp but was offset tems [35–41] would differ greatly from that of P-gp
by the fact it possessed the highest unbound brain con- substrates.
centration married to its high D2 potency. So logBB is P-gp occupies a particular level notoriety within CNS
useful in the sense that it can predict brain distribution but Drug Discovery because of its highly promiscuous sub-
the absolute value is meaningless as is also the case for the strate specificity. P-gp substrate models have used a range
in vivo measurement of Kp.
Due to the introduction of in vitro brain tissue binding
Table 4 Compositions of various human organs as weight fractions
screens, in silico models predicting fubrain values have been of lipid, protein and water showing the higher lipid content of brain
reported in the literature [82]. Table 4 highlights the with respect to blood [86]
composition of the brain relative to plasma and other tis- % Weight fractions
sues. Based on the high lipid content of the brain it is not
surprising that fubrain is inversely proportional to logP Tissue Lipid Protein Water
[9, 82] although this accounts for only around 50 % of the Blood 0.65 18 80
overall variance. Incorporating a wider range of molecular Brain 10.7 7.9 79
characteristics into an in silico model markedly improves Fat 80 5 15
the correlation [82] but it is important to remember that Liver 7 18 72
fubrain data is still largely derived from brain homogenates Kidney 5 17 77
rather than brain slices. Substantial discrepancies have Muscle 2 17 79
been found between these two measurements [76] and have Lung 1 17.7 78
been attributed to loss of pH differences across cellular Heart 10 16.7 72.7
barriers and also the loss of latent transporter activity that is

123
310 J Pharmacokinet Pharmacodyn (2013) 40:301–314

Table 5 Brain distribution in rodents for a series of marketed drugs et al., investigated the brain distribution of acetaminophen
measured in terms of Kp for wild-type (WT) mice and mdr1a/b implanting microdialysis probes in the striatum to measure
knockout (KO) mice and Kin
brain ECF concentrations in rats, and in lateral ventricle
Rodent Kp WT Kp KO Kp ratio and cisterna magna for sampling CSF concentrations. This
Kin miceb miceb work was able to characterise the differences between the
(mL/min/g) mdr1/b mdr1/b (KO/WT)
(?/?) (-/-)
brain ECF and CSF concentration-time profiles in rodents
but then allowed the human brain ECF concentrations to be
Quinidine 0.0299e 0.28 10.07 36.5 predicted based on human CSF data by extrapolation.
e
Verapamil 0.299 0.34 5.63 16.5 Several examples of PBPK predictions of drug pharma-
Risperidone 0.849a 0.78 8.02 10.3 cokinetics in humans and drug–drug interaction have been
Metoclopramide 0.125a 1.22 7.86 6.5 accepted by regulatory agencies, so PBPK modelling and
Sumatriptan 0.00120a 0.02 0.09 3.7 its uses in supporting the filing of therapeutic CNS treat-
Digoxin 0.00197d 0.05 1.4 28.0 ments is expected to evolve in the future.
Loperamide 0.0986c 0.08 2.61 32.6
Risperidone 0.849a 0.44 5.52 12.7
Morphine 0.0104e 0.53 1.51 2.9 Conclusions
Metaclopramide 0.125a 0.79 5.48 6.9
Mesorizadine 0.837a 0.95 16.90 17.8 Distribution of drugs from the vascular compartment into
Thiothixene 2.81a 2.26 24.63 10.9 the brain is governed by both the rate of passage of
Citalopram 0.580a 5.72 17.87 3.1 unbound drug across the BBB and the extent of distribution
a in brain tissue across the intervening cellular barriers.
Taken from Ref. [47]
b Mechanisms for passive diffusion, active transport, tissue
Steady-state measurements in house
c
binding and pharmacokinetics work in concert to produce
Taken from Ref. [95]
d
the concentration-time profile observed in vivo. The range
Taken from Ref. [46]
e
of in vivo, in vitro and in silico tools has increased
Taken from Ref. [96]
markedly over recent years and have contributed to the
improved understanding of the underlying mechanisms
of novel methods to contend with this varied SAR associated with drug distribution into the brain and also in
including more complex machine learning approaches [89]. triaging compounds from new chemical templates that
However, being a P-gp substrate does not necessarily show potency in vitro. The over interpretation of in vivo Kp
prevent in vivo distribution to the brain as shown in measurements or in silico logBB values is now less com-
Table 5 for a series of marketed CNS drugs. This is mon as it much more widely appreciated that unbound drug
because the unbound concentration of drug available to concentrations are more relevant with respect to both
cross the BBB from blood together with the corresponding efficacy and brain distribution. Measurement of in vitro
PS may be sufficient to partially offset the active efflux free fractions in brain and blood (or plasma) have enabled
process and hence still afford the attainment of efficacious Kp values to be converted to Kp,uu in an attempt to
concentrations within the brain. understand the mechanisms associated with BBB passage
PBPK modelling is another in silico approach that in vivo. Assessing BBB permeability remains one of the
merges compound specific parameters (e.g. PS, fubrain) and early interventions in the lead optimisation process to tri-
system specific parameters (e.g. organ blood flow) using a age compounds that display inherently poor physico-
mathematical description of the body in order to predict PK chemical properties or those subject to P-gp efflux, or
parameters [21]. Other in vitro data, such as drug perme- perhaps BCRP efflux.
ability, may also be incorporated into whole body PBPK Literature reports looking at marketed CNS drugs have
models in order to improve their predictive power as shown shown broadly comparable findings in that many appear to
for morphine and oxycodone [90]. Contribution of active be the subject to passive diffusion with a small portion
efflux for morphine was obtained using an in vitro Caco-2 substrates for P-gp but still attain sufficient albeit reduced
cell model whilst active influx contribution to oxycodone brain concentrations. The reality of CNS Drug Discovery is
was evaluated by immortalized rat brain capillary cells. somewhat more complex however because a far wider
The incorporation of these in vitro input parameters rea- variety of chemical templates are being tested and so the
lised a significant improvement in the prediction of the chances of mismatches between in vivo and the current
unbound drug concentration-time profile. Another advan- in vitro measurements increase. This is unsurprising from
tage of PBPK approaches is the capability to determine the perspective of drug transport because contemporary
regional differences within the brain [21, 92]. Westerhout CNS drug screening has not moved much further than P-gp

123
J Pharmacokinet Pharmacodyn (2013) 40:301–314 311

and because the expression of native transporters on cell enzymes in freshly isolated human brain microvessels. Mol
lines such as MDCK is not well understood. If Kp,uu is Pharm 8(4):1332–1341
3. Pangalos MN, Schechter LE, Hurko O (2007) Drug development
markedly lower than unity whereas in contrast in vitro for CNS disorders: strategies for balancing risk and reducing
assessment indicates no likely P-gp efflux then there are attrition. Nat Rev Drug Discov 6(7):521–532
still few experimental options to resort to. From the clinical 4. Morgan P, Van Der Graaf PH, Arrowsmith J, Feltner DE,
perspective it still remains difficult to extrapolate P-gp data Drummond KS, Wegner CD, Street SD (2012) Can the flow of
medicines be improved? Fundamental pharmacokinetic and
across species and it is unclear whether drug–drug inter- pharmacological principles toward improving phase II survival.
actions are likely in humans [92]. Drug Discov Today 17(9–10):419–424
BBB models based on primary brain cultures would 5. Smith QR (2003) A review of blood-brain barrier transport
afford a better tool for screening out compounds across a techniques. Methods Mol Med 89:193–208
6. Dagenais C, Adveef A, Tsinman O, Dudley A, Beliveau R (2009)
wider range of transporters but few Drug Discovery groups P-glycoprotein deficient mouse in situ blood-brain barrier per-
have invested the time needed to make this a standard and meability and its prediction using an in combo PAMPA model.
less tedious approach. Indeed one of the biggest issues with Eur J Pharma Sci 38(2):121–137
in vitro permeability tools is the generally poor correlation 7. Boström E, Hammarlund-Udenaes M, Simonsson US (2008)
Blood-brain barrier transport helps to explain discrepancies in
of values between laboratories running seemingly similar in vivo potency between oxycodone and morphine. Anesthesi-
screens even for simpler models based on MDR–MDCK ology 108(3):495–505
[93]. Although these instances are not in the majority it 8. Kalvass JC, Maurer TS (2002) Influence of nonspecific brain and
does become very challenging to guide the medicinal plasma binding on CNS exposure: implications for rational drug
discovery. Biopharm Drug Dispos 23(8):327–338
chemistry effort from a CNS DMPK perspective and 9. Summerfield SG, Stevens AJ, Cutler L, del Osuna Carmen M,
arguably leads to more compounds being tested in vivo Hammond B, Tang SP, Hersey A, Spalding DJ, Jeffrey P (2006)
because of the inability to triage compounds effectively. Improving the in vitro prediction of in vivo central nervous
This also appears to be genuine problem and not an artefact system penetration: integrating permeability, P-glycoprotein
efflux, and free fractions in blood and brain. J Pharmacol Exp
because the predictive power of PBPK modelling is Ther 316(3):1282–1290
excellent when in vivo measurements are made such as PS 10. Fridén M, Gupta A, Antonsson M, Bredberg U, Hammarlund-
or brain ECF concentrations. So there remains an inherent Udenaes M (2007) In vitro methods for estimating unbound drug
tension in the Drug Discovery paradigm between in vitro concentrations in the brain interstitial and intracellular fluids.
Drug Metab Dispos 35(9):1711–1719
models that afford adequately high throughput versus the 11. Fridén M, Ducrozet F, Middleton B, Antonsson M, Bredberg U,
need to provide better translational data so that the right Hammarlund-Udenaes M (2009) Development of a high-
molecules are identified and progressed more rapidly [94]. throughput brain slice method for studying drug distribution in
Perhaps the compromise lies in some form of the brain the central nervous system. Drug Metab Dispos 37(6):1226–1233
12. Liu X, Smith BJ, Chen C, Callegari E, Becker SL, Chen X,
slice methods [11] and the in silico counterparts [83] Cianfrogna J, Doran AC, Doran SD, Gibbs JP, Hosea N, Liu J,
because these assays retain cellular characteristics yet are Nelson FR, Szewc MA, Van Deusen J (2006) Evaluation of
simpler than those based brain cell cultures. cerebrospinal fluid concentration and plasma free concentration
In summary our understanding of brain distribution has as a surrogate measurement for brain free concentration. Drug
Metab Dispos 34(9):1443–1447
been improved greatly by the range of newer in vivo, 13. de Lange EC, Ravenstijn PG, Groenendaal D, van Steeg TJ
in vitro and in silico approaches and particularly those that (2005) Toward the prediction of CNS drug-effect profiles in
provide information on unbound concentrations or physiological and pathological conditions using microdialysis
unbound partitioning. Long term this can only lead to the and mechanism-based pharmacokinetic-pharmacodynamic mod-
eling. AAPS J 7(3):E532–E543
selection of better CNS drug candidates and ideally assist 14. Tunblad K, Hammarlund-Udenaes M, Jonsson EN (2005) Influ-
in improving the high attrition rates that afflict this field of ence of probenecid on the delivery of morphine-6-glucuronide to
pharmaceutical research. the brain. Eur J Pharm Sci 24(1):49–57
15. Wang Q, Rager JD, Weinstein K, Kardos PS, Dobson GL, Li J,
Acknowledgments The authors would like to thank Dr. Andy Hidalgo IJ (2005) Evaluation of the MDR-MDCK cell line as a
Ayrton for his valuable scientific input during the preparation of this permeability screen for the blood-brain barrier. Int J Pharm
manuscript. 288(2):349–359
16. Di L, Kerns EH, Bezar IF, Petusky SL, Huang Y (2009) Com-
parison of blood-brain barrier permeability assays: in situ brain
perfusion, MDR1-MDCKII and PAMPA-BBB. J Pharm Sci
References 98(6):1980–1991
17. Grumetto L, Carpentiero C, Barbato F (2012) Lipophilic and
1. Hawkins BT, Davis TP (2005) The blood-brain barrier/neurovas- electrostatic forces encoded in IAM-HPLC indexes of basic
cular unit in health and disease. Pharmacol Rev 57(2):173–185 drugs: their role in membrane partition and their relationships
2. Shawahna R, Uchida Y, Declèves X, Ohtsuki S, Yousif S, Dau- with BBB passage data. Eur J Pharm Sci 45(5):685–692
chy S, Jacob A, Chassoux F, Daumas-Duport C, Couraud PO, 18. Virdee K, Cumming P, Caprioli D, Jupp B, Rominger A, Ai-
Terasaki T, Scherrmann JM (2011) Transcriptomic and quanti- gbirhio FI, Fryer TD, Riss PJ, Dalley JW (2012) Applications of
tative proteomic analysis of transporters and drug metabolizing positron emission tomography in animal models of neurological

123
312 J Pharmacokinet Pharmacodyn (2013) 40:301–314

and neuropsychiatric disorders. Neurosci Biobehav Rev 33. Kallem R, Kulkarni CP, Patel D, Thakur M, Sinz M, Singh SP,
36(4):1188–1216 Mahammad SS, Mandlekar S (2012) A simplified protocol
19. Matthews PM, Rabiner EA, Passchier J, Gunn RN (2012) Posi- employing elacridar in rodents: A screening model in drug dis-
tron emission tomography molecular imaging for drug develop- covery to assess P-gp mediated efflux at blood brain barrier. Drug
ment. Br J Clin Pharmacol 73(2):175–186 Metab Lett 16(2):134–144
20. Wotjak CT, Landgraf R, Engelmann M (2008) Listening to 34. Cutler L, Howes C, Deeks NJ, Buck TL, Jeffrey P (2006)
neuropeptides by microdialysis: echoes and new sounds? Phar- Development of a P-glycoprotein knockout model in rodents to
macol Biochem Behav 90(2):125–134 define species differences in its functional effect at the blood-
21. Westerhout J, Ploeger B, Smeets J, Danhof M, de Lange EC brain barrier. J Pharm Sci 95(9):1944–1953
(2012) Physiologically based pharmacokinetic modeling to 35. Paproski RJ, Wuest M, Jans HS, Graham K, Gati WP, McQuarrie
investigate regional brain distribution kinetics in rats. AAPS J S, McEwan A, Mercer J, Young JD, Cass CE (2010) Biodistri-
14(3):543–553 bution and uptake of 3’-deoxy-3’-fluorothymidine in ENT1-
22. Westerhout J, Danhof M, De Lange EC (2011) Preclinical pre- knockout mice and in an ENT1-knockdown tumor model. J Nucl
diction of human brain target site concentrations: considerations in Med 51(9):1447–1455
extrapolating to the clinical setting. J Pharm Sci 100(9):3577–3593 36. Ose A, Kusuhara H, Endo C, Tohyama K, Miyajima M, Kitamura
23. Cremers TI, Flik G, Hofland C, Stratford RE Jr (2012) Microdi- S, Sugiyama Y (2010) Functional characterization of mouse
alysis evaluation of clozapine and N-desmethylclozapine phar- organic anion transporting peptide 1a4 in the uptake and efflux of
macokinetics in rat brain. Drug Metab Dispos 40(10):1909–1916 drugs across the blood-brain barrier. Drug Metab Dispos
24. Bengtsson J, Ederoth P, Ley D, Hansson S, Amer-Wåhlin I, 38(1):168–176
Hellström-Westas L, Marsál K, Nordström CH, Hammarlund- 37. Smith DE, Hu Y, Shen H, Nagaraja TN, Fenstermacher JD, Keep
Udenaes M (2009) The influence of age on the distribution of RF (2011) Distribution of glycylsarcosine and cefadroxil among
morphine and morphine-3-glucuronide across the blood-brain cerebrospinal fluid, choroid plexus, and brain parenchyma after
barrier in sheep. Br J Pharmacol 157(6):1085–1096 intracerebroventricular injection is markedly different between
25. Gupta A, Chatelain P, Massingham R, Jonsson EN, Hammarlund- wild-type and Pept2 null mice. J Cereb Blood Flow Metab
Udenaes M (2006) Brain distribution of cetirizine enantiomers: 31(1):250–261
comparison of three different tissue-to-plasma partition coeffi- 38. Miyajima M, Kusuhara H, Fujishima M, Adachi Y, Sugiyama Y
cients: K(p), K(p, u), and K(p, uu). Drug Metab Dispos 34(2): (2011) Organic anion transporter 3 mediates the efflux transport
318–323 of an amphipathic organic anion, dehydroepiandrosterone sulfate,
26. Bengtsson J, Boström E, Hammarlund-Udenaes M (2008) The across the blood-brain barrier in mice. Drug Metab Dispos 39(5):
use of a deuterated calibrator for in vivo recovery estimations in 814–819
microdialysis studies. J Pharm Sci 97(8):3433–3441 39. André P, Debray M, Scherrmann JM, Cisternino S (2009) Clo-
27. Hammarlund-Udenaes M, Fridén M, Syvänen S, Gupta A (2008) nidine transport at the mouse blood-brain barrier by a new H?
On the rate and extent of drug delivery to the brain. Pharm Res antiporter that interacts with addictive drugs. J Cereb Blood Flow
25(8):1737–1750 Metab 29(7):1293–1304
28. de Lange EC, Danhof M (2002) Considerations in the use of 40. Ose A, Ito M, Kusuhara H, Yamatsugu K, Kanai M, Shibasaki M,
cerebrospinal fluid pharmacokinetics to predict brain target con- Hosokawa M, Schuetz JD, Sugiyama Y (2009) Limited brain
centrations in the clinical setting: implications of the barriers distribution of [3R,4R,5S]-4-acetamido-5-amino-3-(1-ethylprop-
between blood and brain. Clin Pharmacokinet 41(10):691–703 oxy)-1-cyclohexene-1-carboxylate phosphate (Ro 64–0802), a
29. Kodaira H, Kusuhara H, Fujita T, Ushiki J, Fuse E, Sugiyama Y pharmacologically active form of oseltamivir, by active efflux
(2011) Quantitative evaluation of the impact of active efflux by across the blood-brain barrier mediated by organic anion trans-
p-glycoprotein and breast cancer resistance protein at the blood- porter 3 (Oat3/Slc22a8) and multidrug resistance-associated
brain barrier on the predictability of the unbound concentrations protein 4 (Mrp4/Abcc4). Drug Metab Dispos 37(2):315–321
of drugs in the brain using cerebrospinal fluid concentration as a 41. Pan W, Kastin AJ (2002) TNFalpha transport across the blood-
surrogate. J Pharmacol Exp Ther 339(3):935–944 brain barrier is abolished in receptor knockout mice. Exp Neurol
30. Fridén M, Winiwarter S, Jerndal G, Bengtsson O, Wan H, 174(2):193–200
Bredberg U, Hammarlund-Udenaes M, Antonsson M (2009) 42. Tournier N, Cisternino S, Peyronneau MA, Goutal S, Dolle F,
Structure-brain exposure relationships in rat and human using a Scherrmann JM, Bottlaender M, Saba W, Valette H (2012)
novel data set of unbound drug concentrations in brain interstitial Discrepancies in the P-glycoprotein-mediated transport of (18)
and cerebrospinal fluids. J Med Chem 52(20):6233–6243 F-MPPF: a pharmacokinetic study in mice and non-human pri-
31. Doran A, Obach RS, Smith BJ, Hosea NA, Becker S, Callegari E, mates. Pharm Res 29(9):2468–2476
Chen C, Chen X, Choo E, Cianfrogna J, Cox LM, Gibbs JP, 43. Zoghbi SS, Liow JS, Yasuno F, Hong J, Tuan E, Lazarova N,
Gibbs MA, Hatch H, Hop CE, Kasman IN, Laperle J, Liu J, Liu Gladding RL, Pike VW, Innis RB (2008) 11C-loperamide and its
X, Logman M, Maclin D, Nedza FM, Nelson F, Olson E, N-desmethyl radiometabolite are avid substrates for brain per-
Rahematpura S, Raunig D, Rogers S, Schmidt K, Spracklin DK, meability-glycoprotein efflux. J Nucl Med 49(4):649–656
Szewc M, Troutman M, Tseng E, Tu M, Van Deusen JW, 44. Roulet A, Puel O, Gesta S, Lepage JF, Drag M, Soll M, Alvinerie
Venkatakrishnan K, Walens G, Wang EQ, Wong D, Yasgar AS, M, Pineau T (2003) MDR1-deficient genotype in Collie dogs
Zhang C (2005) The impact of P-glycoprotein on the disposition hypersensitive to the P-glycoprotein substrate ivermectin. Eur J
of drugs targeted for indications of the central nervous system: Pharmacol 24 460(2-3):85–91
evaluation using the MDR1A/1B knockout mouse model. Drug 45. Gunn RN, Summerfield SG, Salinas CA, Read KD, Guo Q, Searle
Metab Dispos 33(1):165–174 GE, Parker CA, Jeffrey P, Laruelle M (2012) Combining PET
32. Oostendorp RL, Buckle T, Beijnen JH, van Tellingen O, Schellens biodistribution and equilibrium dialysis assays to assess the free
JH (2009) The effect of P-gp (Mdr1a/1b), BCRP (Bcrp1) and brain concentration and BBB transport of CNS drugs. J Cereb
P-gp/BCRP inhibitors on the in vivo absorption, distribution, Blood Flow Metab 32(5):874–883
metabolism and excretion of imatinib. Invest New Drugs 27(1): 46. Murakami H, Takanaga H, Matsuo H, Ohtani H, Sawada Y
31–40 (2000) Comparison of blood-brain barrier permeability in mice

123
J Pharmacokinet Pharmacodyn (2013) 40:301–314 313

and rats using in situ brain perfusion technique. Am J Physiol functional in vitro model of rat blood-brain barrier for molecular
Heart Circ Physiol 279(3):H1022–H1028 analysis of efflux transporters. Brain Res 1150:1–13
47. Summerfield SG, Read K, Begley DJ, Obradovic T, Hidalgo IJ, 63. Nicolazzo JA, Katneni K (2009) Drug transport across the blood-
Coggon S, Lewis AV, Porter RA, Jeffrey P (2007) Central ner- brain barrier and impact of breast cancer resistance protein
vous system drug disposition: the relationship between in situ (ABCG2). Curr Topics Med Chem 9:130–147
brain permeability and brain free fraction. J Pharmacol Exp Ther 64. Agarwal S, Sane R, Ohlfest JR, Elmquist WF (2011) The role of
322(1):205-13. Erratum in: J Pharmacol Exp Ther 330(3):971-2 the breast cancer resistance protein (ABCG2) in the distribution
48. Liu X, Smith BJ, Chen C, Callegari E, Becker SL, Chen X, of sorafenib to the brain. J Pharmacol Exp Ther 336:223–233
Cianfrogna J, Doran AC, Doran SD, Gibbs JP, Hosea N, Liu J, 65. Poller B, Wagenaar E, Tang SC, Schinkel AH (2011) Double-
Nelson FR, Szewc MA, Van Deusen J (2005) Use of a physio- transduced MDCKII cells to study human P-glycoprotein
logically based pharmacokinetic model to study the time to reach (ABCB1) and breast cancer resistance protein (ABCG2) interplay
brain equilibrium: an experimental analysis of the role of blood- in drug transport across the blood-brain barrier. Mol Pharm 8(2):
brain barrier permeability, plasma protein binding, and brain 571–582
tissue binding. J Pharmacol Exp Ther 313(3):1254–1262 66. van Bree JB, Audus KL, Borchardt RT (1988) Carrier-mediated
49. Liu X, Chen C (2005) Strategies to optimize brain penetration in transport of baclofen across monolayers of bovine endothelial
drug discovery. Curr Opin Drug Discov Devel 8(4):505–512 cells in primary culture. Pharm Res 5(6):369–371
50. Hammarlund-Udenaes M, Paalzow LK, de Lange EC (1997) 67. Joó F (1992) The cerebral microvessels in culture, an update.
Drug equilibration across the blood-brain barrier—pharmacoki- J Neurochem 58(1):1–17
netic considerations based on the microdialysis method. Pharm 68. Huwyler J, Fricker G, Török M, Schneider M, Drewe J (1997)
Res (NY) 14:128–134 Transport of clonidine across cultured brain microvessel endo-
51. Svendsen CN, Hrbek CC, Casendino M, Nichols RD, Bird ED thelial cells. J Pharmacol Exp Ther 282(1):81–85
(1988) Concentration and distribution of thioridazine and 69. Abbott NJ, Hughes CC, Revest PA, Greenwood J (1992)
metabolites in schizophrenic post-mortem brain tissue. Psychiatry Development and characterisation of a rat brain capillary endo-
Res 23(1):1–10 thelial culture: towards an in vitro blood-brain barrier. J Cell Sci
52. Summerfield SG, Lucas AJ, Porter RA, Jeffrey P, Gunn RN, Read 103:23–37
KR, Stevens AJ, Metcalf AC, Osuna MC, Kilford PJ, Passchier J, 70. Coisne C, Dehouck L, Faveeuw C, Delplace Y, Miller F, Landry
Ruffo AD (2008) Toward an improved prediction of human C, Morissette C, Fenart L, Cecchelli R, Tremblay P, Dehouck B
in vivo brain penetration. Xenobiotica 38(12):1518–1535 (2005) Mouse syngenic in vitro blood-brain barrier model: a new
53. Kell DB, Dobson PD, Oliver SG (2011) Pharmaceutical drug tool to examine inflammatory events in cerebral endothelium.
transport: the issues and the implications that it is essentially Lab Invest 85(6):734–746
carrier-mediated only. Drug Discov Today 16(15–16):704–714 71. Cucullo L, Hossain M, Rapp E, Manders T, Marchi N, Janigro D
54. Di L, Artursson P, Avdeef A, Ecker GF, Faller B, Fischer H, (2007) Development of a humanized in vitro blood-brain barrier
Houston JB, Kansy M, Kerns EH, Krämer SD, Lennernäs H, model of screen for brain penetration of antiepileptic drugs.
Sugano K (2012) Evidence-based approach to assess passive Epilepsia 48(3):505–516
diffusion and carrier-mediated drug transport. Drug Discov 72. Lacombe O, Videau O, Chevillon D, Guyot AC, Contreras C,
Today 17(15–16):905–912 Blondel S, Nicolas L, Ghettas A, Bénech H, Thevenot E, Pruvost A,
55. Dagenais C, Avdeef A, Tsinman O, Dudley A, Beliveau R (2009) Bolze S, Krzaczkowski L, Prévost C, Mabondzo A (2011) In vitro
P-glycoprotein deficient mouse in situ blood-brain barrier per- primary human and animal cell-based blood-brain barrier models
meability and its prediction using an in combo PAMPA model. as a screening tool in drug discovery. Mol Pharm 8:651–663
Eur J Pharm Sci 38(2):121–137 73. Nakagawa S, Deli MA, Kawaguchi H, Shimizudani T, Shimono
56. Hakkaarainen J, Jalkanen AJ, Kääriäinen TM, Keski-Rahkonen T, Kittel A, Tanaka K, Niwa M (2009) A new blood-brain barrier
P, Venäläinen T, Hokkanen J, Mönkkönen J, Suhonen M, Fors- model using primary rat brain endothelial cells, pericytes and
berg M (2010) Comparison of in vitro cell models in predicting astrocytes. Neurochem Int 54:253–263
in vivo brain entry of drugs. Int J Pharm 402:27–36 74. Smith DA, Di L, Kerns EH (2010) The effect of plasma protein
57. Mabondzo A, Bottlaender M, Guyot A-C, Tsaouin K, Deverre JR, binding on in vivo efficacy: misconceptions in drug discovery.
Balimane P (2010) Validation of in vitro cell-based human blood- Nat Rev Drug Discov 9(12):929–939
brain barrier model using clinical positron emission tomography 75. Hammarlund-Udenaes M (2010) Active-site concentrations of
radiolligands to predict in vivo human brain penetration. Mol chemicals—are they a better predictor of effect than plasma/
Pharm 7(5):1805–1815 organ/tissue concentrations? Basic Clin Pharmacol Toxicol
58. Lundquist S, Renftel M, Brillault J, Fenart L, Cecchelli R, 106(3):215–220
Dehouck MP (2002) Prediction of drug transport through the 76. Fridén M, Bergström F, Wan H, Rehngren M, Ahlin G,
blood-brain barrier in vivo: a comparison between two in vitro Hammarlund-Udenaes M, Bredberg U (2011) Measurement of
cell models. Pharm Res 19:976–981 unbound drug exposure in brain: modelling of pH partitioning
59. Feng B, Mills JB, Davidson RE, Mireles RJ, Janiszewski JS, explains diverging results between the brain slice and brain
Troutman MD, de Morais SM (2008) In vitro P-glycoprotein homogenate methods. Drug Metab Dispos 39(3):353–362
assays to predict the in vivo interactions of P-glycoprotein with 77. Watson J, Wright S, Lucas A, Clarke KL, Viggers J, Cheetham S,
drugs in the central nervous system. Drug Metab Dispos Jeffrey P, Porter R, Read KD (2009) Receptor occupancy and
36(2):268–275 brain free fraction. Drug Metab Dispos 37(4):753–760
60. Nicolazzo JA, Katneni K (2009) Drug transport across the blood- 78. Liu X, Tu M, Kelly RS, Chen C, Smith BJ (2004) Development
brain barrier and the impact of breast cancer resistance protein of a computational approach to predict blood-brain barrier per-
(ABCG2). Curr Top Med Chem 9(2):130–147 meability. Drug Metab Dispos 32(1):132–139
61. Begley DJ (2004) ABC transporters and the blood-brain barrier. 79. Suenderhauf C, Hammann F, Huwyler J (2012) Computational
Curr Pharm Des 10(12):1295–1312 prediction of blood-brain barrier permeability using decision tree
62. Perrière N, Yousif S, Cazaubon S, Chaverot N, Bourasset F, induction. Molecules 17(9):10429–10445
Cisternino S, Declèves X, Hori S, Terasaki T, Deli M, 80. Muehlbacher M, Spitzer GM, Liedl KR, Kornhuber J (2011)
Scherrmann JM, Temsamani J, Roux F, Couraud PO (2007) A Qualitative prediction of blood-brain barrier permeability on a

123
314 J Pharmacokinet Pharmacodyn (2013) 40:301–314

large and refined dataset. J Comput Aided Mol Des 25(12): 90. Ball K, Bouzom F, Scherrmann J-M, Walther B, Declèves X
1095–1106 (2012) Development of a physiologically based pharmacokinetic
81. Fan Y, Unwalla R, Denny RA, Di L, Kerns EH, Diller DJ, model for the rat central nervous system and determination of an
Humblet C (2010) Insights for predicting blood-brain barrier in vitro-in vivo scaling methodology for the blood-brain barrier
penetration of CNS targeted molecules using QSPR approaches. permeability of two transporter substrates, morphine and oxyco-
J Chem Inf Model 50(6):1123–1133 done. J Pharm Sci 101:4277–4292
82. Wan H, Rehngren M, Giordanetto F, Bergström F, Tunek A 91. de Lange E, Ravenstijn P, Groenendaal D, van Steeg T (2005)
(2007) High-throughput screening of drug-brain tissue binding Toward the prediction of CNS drug-effect profiles in physiolog-
and in silico prediction for assessment of central nervous system ical and pathological conditions using microdialysis and mecha-
drug delivery. J Med Chem 50(19):4606–4615 nism-based pharmacokinetic-pharmacodynamic modelling. The
83. Chen H, Winiwarter S, Fridén M, Antonsson M, Engkvist O AAPS J 7(3):E532–E543
(2011) In silico prediction of unbound brain-to-plasma concen- 92. International Transporter Consortium, Giacomini KM, Huang
tration ratio using machine learning algorithms. J Mol Graph SM, Tweedie DJ, Benet LZ, Brouwer KL, Chu X, Dahlin A,
Model 29(8):985–995 Evers R, Fischer V, Hillgren KM, Hoffmaster KA, Ishikawa T,
84. Chen L, Li Y, Yu H, Zhang L, Hou T (2012) Computational Keppler D, Kim RB, Lee CA, Niemi M, Polli JW, Sugiyama Y,
models for predicting substrates or inhibitors of P-glycoprotein. Swaan PW, Ware JA, Wright SH, Yee SW, Zamek-Gliszczynski
Drug Discov Today 17(7–8):343–351 MJ, Zhang L (2010) Membrane transporters in drug development.
85. Bikadi Z, Hazai I, Malik D, Jemnitz K, Veres Z, Hari P, Ni Z, Nat Rev Drug Discov 9(3):215–236
Loo TW, Clarke DM, Hazai E, Mao Q (2011) Predicting 93. Jeffrey P, Summerfield SG (2007) Challenges for blood-brain
P-glycoprotein-mediated drug transport based on support vector barrier (BBB) screening. Xenobiotica 37(10–11):1135–1151
machine and three-dimensional crystal structure of P-glycopro- 94. Naik P, Cucullo L (2012) In vitro blood-brain barrier models:
tein. PLoS One 6(10):e25815 current and perspective technologies. J Pharm Sci 101(4):
86. Snyder WS (1975) Report of the task force on reference man. 1337–1354
Pergamon, Oxford 95. Dagenais C, Graff CL, Pollack GM (2004) Variable modulation
87. Avdeef A, Sun N (2011) A new in situ brain perfusion flow of opioid brain uptake by P-glycoprotein in mice. Biochem
correction method for lipophilic drugs based on the pH-dependent Pharmacol 67(2):269–276
Crone-Renkin equation. Pharm Res 28(3):517–530 96. Dagenais C, Zong J, Ducharme J, Pollack GM (2001) Effect of
88. del Amo EM, Urtti A, Yliperttula M (2008) Pharmacokinetic role mdr1a P-glycoprotein gene disruption, gender, and substrate
of L-type amino acid transporters LAT1 and LAT2. Eur J Pharm concentration on brain uptake of selected compounds. Pharm Res
Sci 35(3):161–174 18(7):957–963
89. Leong MK, Chen HB, Shih YH (2012) Prediction of promiscuous
p-glycoprotein inhibition using a novel machine learning scheme.
PLoS One 7(3):e33829

123

You might also like